Top Banner
462
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Toxicilogical problems 1
Page 2: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

459

Page 3: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

1

Military Publishing Ltd.

TOXICOLOGICAL PROBLEMS

Edited by

Prof. Christophor Dishovsky, MD, PhD, DSc, ERTAssoc. Prof. Julia Radenkova – Saeva, MD, PhD

SOFIA • 2014

Page 4: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

2

Copyright © 2014 Bulgarian Toxicological Society, Sofi a, Bulgaria

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, without written permission from the authors.Authors bear full responsibility for their contributions.Authors will not receive honoraria for their contributions.

First published in February 2014 by Bulgarian Toxicological Society .

A catalogue record of this book is available from National Library “St. St. Cyril and Methodius”, Sofi a.

Toxicological ProblemsEditor: Christophor Dishovsky and Julia Radenkova-Saeva

Bulgarian Toxicological Society .

ISBN 978-954-509-509-2

Page 5: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

3

CONTENTS

Preface / 11

Part 1ESTERASE STAUS ASSAY AS A NEW APPROACH TO OPC EXPOSURE ASSESMENT

Chapter 1Investigation of Esterase Status as a Complex Biomarker of Exposure to Organophosphorus Compounds Makhaeva G. F., Rudakova E. V. and Sigolaeva L. V. / 15

Chapter 2 Esterase Status of Various Species in Assessment of Exposure to Organophosphorus CompoundsBoltneva N. P., Rudakova E. V., Sigolaeva L. V. and Makhaeva G. F. / 27

Chapter 3Investigation of Mice Blood Neuropathy Target Esterase as Biochemical Marker of Exposure to Neuropathic Organophosphorus CompoundsRudakova E. V., Sigolaeva L. V. and Makhaeva G. F. / 39

Chapter 4 Layer-by-layer electrochemical biosensors for blood esterases assay Kurochkin I. N., Sigolaeva L. V., Eremenko A. V., Dontsova E. A., Gromova M. S., Rudakova E. V. and Makhaeva G. F. / 51

Chapter 5Tyrosinase-based Biosensors for Assay of Carboxylesterase, Neuropathy Target Esterase, and Paraoxonase Activities in BloodSigolaeva L., Eremenko A. V. , Rudakova E. V. ,Makhaeva G. F. and Kurochkin I. N. / 68

Chapter 6Thick Film Thiol Sensors for Cholinesterases AssayEremenko A. V., Dontsova E. A., Nazarov A. and Kurockin I. N. / 82

Chapter 7Genotyping of Esterase Genes (Ache, Pnpla6, Bche and Ces1) and Paraoxonase 1 Gene (Pon1)Nosikov V. V., Bochkarev V. V. and Nikitin A.G. / 88

Page 6: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

4

Chapter 8 Esterase Status as Biomarker of OPC Exposure and Treatment with Reactivatror of ChEDishovsky C., Ivanov T. and Petrova I. / 93

Part 2CONTEMPORARY ASPECTS OF CLINICAL TOXICOLOGY Chapter 9Multi-organ Dysfunction Syndrome – a Result of Prolonged Hypoxiafrom an Overdose of MethadoneGesheva M., Petkova M., Radenkova-Saeva J. and Loukova A. / 101

Chapter 10Serotonin Syndrome in Acute Amphetamine Intoxication Kirova E. / 106

Chapter 11Contemporary Profi le of Toxicological Morbidity at MMA in 2012, Sofi aKonov V., Neykova L. and Kanev K. / 111

Chapter 12 Clinical Case of a Welder Chronic Intoxication with Metal Aerosols Kuneva T., Apostolova D., Dimitrova T., Boyadzhieva V. and Petkova V. / 115

Chapter 13 Health Risk Assessment of Exposure to Chemicals Among MinersLyubomirova K. and Ratcheva Z. / 119

Chapter 14 Methadone Poisoning – Clinical and Psychosocial ConstellationsRadenkova – Saeva J. and Loukova A. / 124

Chapter 15 Mycotoxins – Adverse Human Health Effects Radenkova – Saeva J. / 129

Chapter 16Blood Level of Histamine- Indicator for the Expression of Abstinence Syndrome in the Phase of Alcohol Withdrawal Neykova L., Konov V., Kanev K. and Galabova G. / 137

Page 7: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

5

Chapter 17Traditional and New Treatments of Addiction – Ethical and Legal Aspects Radenkova-Saeva J. and Saeva E. / 143

Chapter 18 Quercetin Use and Human Health: Risk and Benefi tsTzankova V. and Dirimanov S. / 148

Chapter 19 Protective Effect of Aronia melanocarpa Fruit Juice in a Model of Paracetamol-induced Hepatotoxicity in RatsValcheva-Kuzmanova S. Kuzmanov K. / 160

Chapter 20Exogenic Intoxication with Neuroleptix and Antipsychotic Medications – Clinical CaseDakova S., Ramshev N., Ramsheva Z., Ramshev K. and Kanev K. / 167

Chapter 21Toxic Pneumofi brosis – Late Occurrence of Chronic Cadmium ExpositionApostolova D., Kuneva T., Petkova V. and Medzhidieva D. / 170

Chapter 22Sick Building Syndrome – Proper Management for Protect Human HealthRadenkova-Saeva J. and Saeva E. / 177

Chapter 23Case of Severe Intoxication and Anaphylactic Reaction from Multiple Bee StingsStefanova K. and Barzashka E. / 184

Chapter 24Special Features in the Treatment after Pepper Spray KO jet Expoture in Childhood ( clinical case)Stefanova K. / 189

Chapter 25 Suicidal Self-poisonings and ResilienceVanev P. and Radenkova-Saeva J. / 194

Chapter 26Toxicity of Amlodipine in Combination with Enalapril – a Case of SuicideRamshev N., Dakova S., Ramsheva Z., Ramshev K. and Kanev K. / 201

Page 8: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

6

Chapter 27Problems of Effective Clinical Communication in Cases of Acute IntoxicationYovcheva M. and Zlateva S. / 208

Chapter 28Diagnostic and Therapeutic Problems of Toxoallergic Reaction after Scolopendra Bite – 10-Years ExperienceYovcheva M., Marinov P. and Zlateva S. / 216

Chapter 29Characteristics of Acute Chemical Poisonings in Ukraine: Morbidity and MortalityLevchenko O. and Kurdil N. / 225

Part 3DRUG TOXICITY

Chapter 30Toxicology – Faculty of Pharmacy, Medical University, Sofi a – in the YearsMitcheva M., Danchev N., Astroug H., Tzankova V., Simeonova R., Kondeva-Burdina M. and Vitcheva V. / 233

Chapter 31 7-Nitroindazole, a Selective Inhibitor of Neuronal Nitric Oxide Synthase ( Nnos) Attenuates Cocaine and Amphetamine Withdrawal and Brain Toxicity in Rats Vitcheva V., Simeonova R. and Mitcheva M. / 241

Chapter 32Cytotoxic Effects Of Zn/Ag Complex On Cultured Non-Tumor CellsAlexandrova R., Abudalleh A., Zhivkova T., Dyakova L., Shishkov S., Alexandrov M., Marinescu G., Culit D. C. and Patron L. / 248

Chapter 33Effects of D-Amphetamine on Brain and Hepatocytes Isolated from Male and Female Spontaneously Hypertensive Rats (Shr Simeonova R., Kondeva-Burdina M., Vitcheva V. and Mitcheva M. / 254

Chapter 34An Update on the Signifi cance of Pharmacovigilance in Patient SafetyOzcagli E., Vynias D., Alpertunga B. and Tsatsakis A. M. / 263

Page 9: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

7

Chapter 35Effectsof Multiple Ethanol Administration on Some Brain and Hepatic Biochemical Parameters in Male and Female Spontaneously Hypertensive Rats (Shr)Simeonova R., Vitcheva V. and Mitcheva M. / 268

Chapter 36 Gold Nanoparticles Cytotoxicity Evaluationin HEP G2 Cells Tzankova V., Kachamakova M., Valoti M. / 280

Chapter 37Pilot Studies of Pharmacological and Toxicological Effects of Newly Synthesized Neuropeptides with Short ChainsStoeva S., Кlisurov R., TanchevaL., Dragomanova S., Pajpanova T., Kalfi n R., and Georgieva A. / 287

Chapter 38The Procsses of Bioactivation, Toxicity and Detoxication – Experimental Models for Evaluation of the ToxicityMitcheva M., and Kondeva-Burdina M. / 292

Chapter 39 OPRD1 Polymorphism is not Associated with Chronic Cocaine Use Vakonaki E., Manoli O., Kovatsi L., Mantsi M., DiamantaraE., Belivanis S., Tzatzarakis M. and Tsatsakis A. M. / 301

Chapter40Effect of Myosmine, Administered Alone and on Tert-Butyl Hydroperoxide-Induced Toxicity in Isolated Rat HepatocytesKondeva-Burdina M., Gorneva G.and Mitcheva M. / 307

Chapter 41Cytoprotective Effects of Saponarin from Gypsophila Trichotoma on Tert-Butyl Hydroperoxide and Cocaine-Induced Toxicity in Isolated Rat HepatocytesKondeva-Burdina M., Simeonova R., Vitcheva V. , Krasteva I. and Mitcheva M. / 313

Part 4CHEMICALS TOXICITY

Chapter 42Toxic Effects of Benzene Metabolites Hydroquinone, Catechol and Phenol in HL60 Cells Tzankova V., Velichkov B. and Valoti M. / 321

Page 10: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

8

Chapter 43Environmental Chemical Pollution Impact on Italian Campania RegionNoschese G. and Kostadinov R. / 328

Chapter 44An Update of Issues Regarding Hair Analysis of Organic PollutantsKavvalakis M., Appenzeller B. and Tsatsakis A. M. / 333

Chapter 45Effect of Solvents on the Toxicity of Some L-Valine Peptidomimetics in RatsKlisurov R., Encheva E. , Genadieva M., Tancheva L. and Tsekova D. / 341

Chapter 46Health Risk Associated with Genetic Polymorphism of Apoe in Bulgarian Workers Exposed to Carbon Disulfi de Georgieva T., Genova Z., Peneva-NikolovaV. , Panev T., Mikhailova A. and PopovT. / 345

Chapter 47Chemical Risk Assessment an Imperative in Medical Training and EducationKostadinov R., Kanev K. and Noschese G. / 354

Chapter 48Neonicotinoid Pesticides– Some Medical Intelligence Concerns Kostadinov R., Noschese G. and Popov G. / 360

Part 5CHEMICAL TERRORISM, DIAGNOSIS AND TREATMENT OF EXPOSURE TO CHEMICAL AGENTS

Chapter 49Chemical Weapon Information Revealed By Aum Shinrikyo Death RowInmate Dr. Tomomasa NakagawaTu A. T. / 369

Chapter 50Research of Mobility of Sodium Arsenite in Soils of Areas of Destruction of the Chemical WeaponPetrov V., Shumilova M., Nabokova O. and Lebedeva M. / 374

Chapter 51The Control of Formation of Dioxins in Incinerators for Reactionary Masses of Destruction of the Chemical WeaponPetrov V., Stompel S. and Bukov V. / 379

Page 11: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

9

Chapter 52The Effi ciency of Carbamates as Components of ProphylacticAntidotes Against OP Poisoning Tonkopii V. and Zagrebin A. / 384

Chapter 53A New Possibility for Separate Detection of Organophosphates and Carbamates Tonkopii V. and Zagrebin A. / 389

Chapter 54Biomarkers of Organophosphorus Compounds Poisoning and Exposure: a Review Soltaninejad K. / 393

Chapter 55Development of an in-vitro assay of apparent permeability across artifi cial membranes for antidotal oximesVoicu V., Medvedovici A. V., Miron D. S. , and Rădulescu F S. / 404

Chapter 56Assessment of the Reactivating Potency of Different Oximes in Tabun Poisoned RatsSamnaliev I. and Petrova I. / 411

Chapter 57Investigation of Prophylactic Effi cacy of Drug Mixture Consisting of Physostigmine and Procyclidine Applied Alone or Followed by Antidote Therapy in Soman Poisoning Samnaliev I. and Ivanov T. / 416

Chapter 58Opportunities for Optimization of the Antidotal Treatment of Acute Poisoning Caused by Soman by Means of Antidote Combinations Containing HI-6 and Centrally Acting CholynoliticsDishovsky C., Samnaliev I. and Draganov D. / 421

Chapter 59Toxidin - Bulgarian Ampule Form of Reactivator of Cholinesterse HI-6Dishovsky C., Stoikova S. and Petrova I. / 428

Page 12: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

10

Chapter 60 Mycotoxins: Novel Approaches for Biological Threat MitigationAgarwal P., Arora R., Chawla R., Gupta D., Zheleva A., Gadjeva V. and Stoev S. / 433

Chapter 61Signifi cance of Butyrylcholinesterase Variants for Military PersonnelDimov D. and Kanev K. / 445

Chapter 62Antidotes –the Role of the Pharmacy in Medical Response in CrisisKanev K., Paskalev K.,Chobanov N. and Kolev S. / 449

Subject Index / 454

Page 13: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

11

Preface

This book includes reports which were presented at the Fourth National Congress of Clinical Toxicology with International Participation and Annual Meeting of Bulga-rian Toxicological Society, which were held in Sofi a, Military Medical Academy, Bulgaria, from 7 to 8 November, 2013. The Congress is dedicated to the 50th years anniversary of the Toxicology Clinic,MHATEM “N. I. Pirogov” – Sofi a and the initia-tion of the Bulgarian school of clinical toxicology.

The book also includes a papers connected with the NATO project number SfP 984082 “ Esterase status for diagnostics and prognosis of OPC intoxications” (OPC Detection). They were submitted to Congress by the Project Directors and the key Project persons (http://opcdiagnostic.org/).

Some of the papers in the book were selected from the materials sent from scientists from different countries, which were close to the topics of the Congress.

The Congress and the Annual Meeting of Bulgarian Toxicological Society were orga-nized by BULGARIAN TOXICOLOGICAL SOCIETY and BULGARIAN CLINICAL TOXICOLOGY ASSOCIATION with the kind cooperation of MILITARY MEDICAL ACADEMY, Sofi a.

This book will be interesting and useful for medical students, medical doctors, specia-lists in toxicology and in the fi eld of personal and social safety, environmental protection experts, chemists, biologists, and specialists of the army and governmental anti-terrorist departments.

Printing of the book was carried out with funds of a Bulgarian Toxicological Society and the NATO project number SfP 984082 “ Esterase status for diagnostics and prognosis of OPC intoxications”.

Editors:

Prof. Christophor Dishovsky MD, PhD, DSc, ERTAssoc. Prof. Julia Radenkova – Saeva, MD, PhD

Page 14: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

12

Page 15: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

13

Part 1

ESTERASE STAUS ASSAY AS A NEW

APPROACH TO OPC EXPOSURE ASSESMENT

Page 16: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

14

Page 17: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

15

Chapter 1

Investigation of Esterase Status as a Complex Biomarker of Exposure to Organophosphorus

Compounds

Galina F. MAKHAEVAa1, Elena V. RUDAKOVAa, Larisa V. SIGOLAEVAb

aInstitute of Physiologically Active Compounds Russian Academy of SciencesChernogolovka, Moscow Region 142432 Russia

bDepartment of Chemistry, Moscow State University, Moscow 119992 Russia

Abstract. Development of biomarkers of human exposures to OPCs and their quantifi cation is a vital component of a system of prediction and early diagnosis of induced diseases. Our study was focused on investigation of esterase status as a complex biomarker of exposure to OPCs and an aid in accurate diagnosis. We suggest that this complex biomarker should be more effective and informative than standard assays of plasma BChE, RBC AChE, and lymphocyte neuropathy target esterase ( NTE). It will allow us: 1) to assess an exposure as such and to confi rm the nonexposure of individuals suspected to have been exposed; 2) to determine if the exposure was to agents expected to produce acute and/or delayed neurotoxicity; 3) to perform dosimetry of the exposure, which provides valuable information for medical treatment. To confi rm this hypothesis, we examined the changes in activity of blood AChE, NTE, BChE and carboxylesterase ( CaE) 1 h after i.p. administration of increasing doses of three OPCs with different esterase profi les: (C2H5O)2P(O)OCH(CF3)2, (C4H9O)2P(O)OCH(CF3)2 and (C3H7O)2P(O)OCH=CCl2. The esterases assay was performed in hemolysed blood by biosensor and spectrophotometric methods. Analysis of the obtained dose-dependences for blood esterases inhibition showed that blood BChE and CaE are the most sensitive biomarkers, allowing detection of low doses. Inhibition of blood NTE and AChE can be used to assess the likelihood that an exposure to OPC would produce cholinergic and/or delayed neuropathic effects. Thus, determination of esterase status allows one to improve the possibilities of diagnostics of exposure to OPCs.

Key words. Acetylcholinesterase, biomarker, blood, butyrylcholinesterase, carboxy-lesterase, neuropathy target esterase, organophosphorus compounds ( OPCs)

IntroductionThe term biomarker is used to mean biological, biochemical, and molecular markers

that can be measured by chemical, biochemical, or molecular techniques [1]. In humans, biomarkers must be present in easily and ethically obtainable tissues, one of which is blood. Biomarkers are usually divided into three categories: biomarkers of exposure, effect,

1 Laboratory of Molecular Toxicology, Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, Moscow Region 142432, Russia, E-mail: [email protected]

Page 18: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

16

and susceptibility [2]. OPC-modifi ed enzymes are more stable in the organism than intact OPCs, due to the rapid elimination of free OPCs. For this reason, new methods to identify and quantify the degree of modifi cation of biomarker proteins need to be developed.

The phosphylating properties of organophosphorus compounds ( OPCs) containing pentavalent phosphorus lead to their interactions in an organism with various serine esterases. These enzymes include primary targets, e.g., acetylcholinesterase (EC 3.1.1.7, AChE, target of acute toxicity) [3] and neuropathy target esterase (EC 3.1.1.5, NTE, target of OPC-induced delayed neuropathy, OPIDN) [4], as well as secondary ones, e.g., butyrylcholinesterase (EC 3.1.1.8, BChE) and carboxylesterase (EC 3.1.1.1, CaE), which act as stoichiometric scavengers of OPCs, i.e. alternative phosphylation sites, thereby decreasing the concentration of the active OPC available for interaction with AChE or other target sites [5-7]. Recently, some other proteins possessing esterase activity have been identifi ed as secondary targets for OPCs: acylpeptide hydrolase, fatty acid amide hydrolase, arylformamidase, albumin [8-10].

Figure 1. OPCs interaction with various serine esterases (E-OH), their possible toxic effects, role in mechanisms of toxicity and function as biomarkers

The knowledge of structural and pharmacodynamic similarities between brain and RBC AChE within a given species has provided a rational basis for using RBC AChE inhibition by anti-AChE OPCs as a surrogate measure of brain AChE inhibition by these compounds [3]. AChE activity in blood often corresponds to that in the target organs, and it can be considered as an appropriate parameter for biological monitoring of exposure to anticholinesterase agents [11].

A part of an effective chemical defense strategy is to develop methods for detecting delayed neuropathic agents via sensitive and selective biomarkers [12]. The discovery of NTE in circulating lymphocytes and platelets [13-16] enabled it to be used as a biomarker of animal and human exposure to neuropathic OPCs [16-21]. The development by our team an electrochemical method for NTE assay using tyrosinase-based biosensors enabled measuring NTE activity in whole blood [22-25] that cannot be done using the standard

PRO

ORO

X

(AChE)-O

H

POR

OOR

(AChE)-O

POR

OOR

(NTE)-O

POR

OOR

(BChE)-O

POR

OOR

(CaE)-O

PO

OOR

(NTE)-O

Acute cholinergictoxicity

Stoichiometric scavengingof E-OH inhibitors

Stoichiometric scavengingof E-OH inhibitors

(NTE)-OH

(BChE)-OH(CaE)-OH

aging Delayedneurotoxicity(OPIDN)

BIOMARKER

BIOMARKER

BIOMARKER

BIOMARKER

Page 19: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

17

colorimetric assay. The developed biosensor was used to establish correlations of NTE inhibitions in blood with that in lymphocytes and brain 24 h after dosing hens with a neuropathic OPC, O,O-dipropyl-O-dichlorvinylphosphate, PrDChVP [23]. These studies indicate that NTE in whole blood can be assayed and used as a biomarker of exposure to delayed neuropathic OPCs.

Because many OPCs react with these non-target esterases in vitro more effi ciently than with AChE, and given that plasma esterases would be the fi rst binding sites encountered by OPCs following their absorption into the blood, they tend to be more sensitive biomarkers than RBC AChE is, and can therefore detect exposure to lower doses of OPs. BChE activity measurements in either plasma (or serum) or whole blood are generally used as a sensitive biomonitor of the exposure to OPCs [3, 26, 27]. In general, AChE and BChE, which have half-lives of 5-16 days, provide excellent biomarkers of exposure to OPs [11].

The set of activities of four blood serine esterases: AChE, NTE, BChE, and CaE, as well as serum paraoxonase ( PON1), which can hydrolyze and detoxify OPCs [28], was denoted by the term, “ esterase status” of an organism [29, 30]. The esterase status incorporates aspects of susceptibility and exposure; i.e., it largely determines an individual’s sensitivity to OPCs, and it may be used as a complex biomarker of exposure to these compounds [30].

We suggested that this complex biomarker should be more effective and informative for OPC exposure assessment than standard assays of plasma BChE, RBC AChE, and lymphocyte NTE [30, 31]. It will allow us: 1) to assess an exposure as such and to confi rm the nonexposure of individuals suspected to have been exposed; 2) to determine if the exposure was to agents expected to produce acute and/or delayed neurotoxicity; 3) to perform dosimetry of the exposure, which provides valuable information for medical treatment.

To confi rm this hypothesis, we carried out a detailed examination of the changes in activity of blood AChE, NTE, BChE and CaE 1h after i.p. administration to mice increasing doses of three OPCs with different esterase profi les and different acute and delayed toxicity: two O-phosphorylated hexafl uoroisopropanols: (C2H5O)2P(O)OCH(CF3)2 (diEt-PFP) and (C4H9O)2P(O)OCH(CF3)2 (diBu-PFP), and O,O-dipropyl-O-dichlorovinyl phosphate (C3H7O)2P(O)OCH=CCl2 (PrDChVP).

DiEt-PFP has medium acute toxicity (LD50 = 200 mg/kg) and low neuropathic potential RIP = 0.07) [32]; diBu-PFP has low acute cholinergic toxicity (LD50 > 2000 mg/kg) [33] and high neuropathic potential [32]; PDChVP has high acute toxicity (10mg/kg, hens [34], 15 mg/kg, mice [35] ) and high neuropathic potential (RIP = 2.6, hens [36]).

Before performing this research, the spectrophotometric methods of AChE, BChE and CaE assay in whole blood were developed [37] that were also used for validation of biosensor measurements. New screen-printed tyrosinase-based LBL biosensors were developed [38, 39]. The basal activities of AChE, BChE, CaE and NTE in mice blood have been determined [37, 40, 41, 42]. In a detailed study it was shown that mouse blood NTE can be used as a biochemical marker of exposure to neuropathic OPCs and relationship between AChE and NTE inhibition in mouse blood after OPC exposure allows one to assess the neuropathic hazard of the compound [35, 43].

Page 20: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

18

Materials and methodsChemicalsPhenyl valerate (PV), N,N′-di-2-propylphosphorodiamidofl uoridate (mipafox, MIP),

O,O-diethyl-O-(1-trifl uoromethyl-2,2,2-trifl uoroethyl) phosphate (diet-PFP),O,O-dibutyl-O-(1-trifl uoromethyl-2,2,2-trifl uoroethyl) phosphate (diBu-PFP) and O,O-di-1-propyl O-2,2-dichlorovinyl phosphate (PrDChVP), were synthesized and characterized in the Institute of Physiologically Active Compounds, Russian Academy of Sciences (Russia) and kindly presented by Dr. Alexey Aksinenko. Synthesis of diEt-PFP and diBu-PFP is described in [32, 44]. The purity of all substances was > 99% (by spectral and chromatographic analysis data). Paraoxon (O,O-diethyl-4-nitrophenyl phosphate) was from Sigma Chemical Co (St. Louis, MO). All other chemicals were analytical grade and used without further purifi cation. Aqueous solutions were prepared using deionized water.

In vitro OPC inhibitor potency determination Commercial preparations of human RBC AChE, horse serum BChE, porcine liver

CaE (Sigma, USA) and lyophilized preparation of hen brain NTE [45] were used as the standard enzyme sources. AChE/BChE activity was determined with the colorimetric method of Ellman [46] using acetyl-/butyrylthiocholine as substrate. 4-Nitrophenyl acetate was used for spectrophotometric CaE activity assay. NTE was determined by the differential inhibition method of Johnson [47], substrate – phenyl valerate. Measurements were performed on Benchmark Plus microplate reader.

For kinetic studies of AChE, NTE, BChE and CaE inhibition by OPC in vitro, the enzyme preparation was incubated with the inhibitor in buffer with a fi nal DMSO concentration of 1% (v/v) for different times. The residual enzyme activity was then assayed in triplicate for each experiment. The slopes (k′) of each plot of log (% activity remaining) versus time were calculated by linear regression. These values of k′ were then plotted against inhibitor concentration [I], and the slope (k″) of the resultant line was derived by linear regression. The bimolecular rate constant of inhibition (ki) was calculated as a measure of inhibitory potency using the relationship, ki=2.303 k′/[I] = 2.303 k″. Each value of k′ was obtained from a line through four to six points. Plotting and regression analysis was done using Origin 6.1 software, OriginLab Corp. (Northampton, MA). Enzymes activity assay.

Animal experiments and tissues preparationIn vivo inhibition of AChE, NTE, BChE and CaE in mouse bloodIn vivo experiments were carried out on outbred male white miceCD1 (18-25 g). All

experiments and procedures with animals were carried out according to the protocols approved by the Ethics Committee of the Institute of Physiologically Active Compounds RAS (Chernogolovka, Russia).

diEt-PFP, diBu-PFP and PrDChVP were dissolved in DMSO and injected once, i.p. in a volume about 0.1 ml in 8-10 increasing doses.For each dose at least 6 animalswere used. 20 min before PrDChVP injection, mice were pretreated with atropine sulfate, 20 mg/kg in water, i.p.; control animals received atropine sulfate and DMSO. In the experiment with diBu-PFP control animals were administered only with DMSO. After 1 h, the mice were decapitated under CO2 anesthesia. Trunk blood from each animal was collected immediately

Page 21: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

19

in glass vials containing 3.8% w/v sodium citrate (0.2 ml citrate/ml blood). All blood samples were aliquoted, frozen in liquid nitrogen, and stored at -70oC before use.

AChE, NTE, BChE and CaE activities in blood of mice treated with the OPCs were determined and compared to activity in tissue samples from control animals treated with DMSO or DMSO + atropine. Analysis of dose - response was performed using Origin 6.1 software, OriginLab Corp. (Northampton, MA) to yield ED50 values.

Preparation of blood for analysis of esterases activityThe blood samples were thawed at4°C and diluted 1:100 (v:v) in cold 0.1 M sodium

phosphate pH 7.5 for preparing blood hemolysates. After thorough mixing for 2 minutes the hemolysed blood samples were aliquoted into plastic tubes Falcon, immediately frozen in liquid nitrogen to ensure complete hemolysis and stored at -20 єC until use. Before analysis, the samples were thawed slowly in ice water bath.

AChE/BChE assay Activity of AChE in hemolysed whole blood was evaluated by the rate of hydrolysis

of 1 mM acetylthiocholine by Ellman’s colorimetric method [46] in 0.1 M K,Na-phosphate buffer (pH 7.5) at 25oC in the presence of a specifi c inhibitor of BChE 0.02 mM ethopropazine [48-50]. To minimize the impact of hemoglobin absorption, the standard for Ellman assay wavelength (412 nm) was changed to 436 nm (ε436 = 10600 M-1cm-1) [50]. Activity of BChE in the hemolysed blood was measured under the same conditions with 1 mM butyrylthiocholine as a substrate.

CaE assay Spectrophotometric determination of CaE in hemolysed whole blood was carried out

with 1 mM 1-naphthyl acetate at λ 322 nm (ε322 = 2200 M-1cm-1) [51, 52] in 0.1 M K,Na-phosphate buffer (pH 8.0) at 25oC. Specifi c inhibitors of PON1/arylesterases (2 mM EDTA) and cholinesterases (40 μM eserine) were used for CaE activity discrimination [53].

Blood NTE assay NTE activity was assayed according to the differential inhibition method of Johnson

[47] with an electrochemical endpoint as described previously [22, 23]. A new sensitive, stable and reproducible planar tyrosinase biosensor of construction SPE/PDDA/Tyr was used for phenol detection. This biosensor was developed through a combination of screen-printing technology for conducting graphite support preparation and LBL technology of polyelectrolytes/oxidoreductase deposition [38,39].

Briefl y, 1:100 (v:v) blood hemolysate samples were incubated at 37 °C with 50 μM paraoxon (sample B) or 50 μM paraoxon plus 250 μM mipafox for 20 min (sample C). Substrate phenyl valerate (fi nal apparent concentration 0.54 mM) was added and the incubation was continued for the next 40 min at 37 °C. The reaction was stopped by addition of 100 μl of 1% (w/v) aqueous SDS. Total volume of reaction mixture was 600 μl.The released phenol was assayed amperometrically after 20- to 50-fold dilution of samples in 50 mM sodium phosphate with 100 mM NaCl, pH 7.0. The analytical signal was determined as the value of steady-state baseline current change (the difference between an average value of steady-state baseline current before and after analyte addition). Activity values were calculated using phenol calibration curves,

Page 22: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

20

obtained under the same conditions, and each was corrected for spontaneous hydrolysis of substrates, determined separately.

Acute toxicity determination The acute toxicity of the tested compounds was determined in outbred male white

mice weighing 18-25 g which received i.p. injections of OPCs. The observation period was 24h. The LD50 values were calculated by probit analysis using the BioStat 2006 program.

StatisticsStatistical analysis wascarried out using GraphPad Prism version 3.02 for Windows,

GraphPad Software (San Diego, CA). The results are given as means ±SEM. The level of signifi cance was set at p<0.05.

3. Results and Discussion

OPC inhibitor activity in vitro

First of all, in vitro inhibitor activity of the studied OPCs against all of the esterases in standard conditions was tested. Commercial preparations of human RBC AChE, horse serum BChE, porcine liver CaE and lyophilized preparation of hen brain NTE were used as the standard enzyme sources.

In vitro inhibitor activity of the OPCs to all of the esterases along with their acute toxicity is presented in Table 1. Table 2 shows inhibitor selectivity of the studied OPCs. The results agree with earlier published data [32, 38]

As one can see from Tables 1, 2, all of dialkylphosphates more effective inhibit the secondary targets BChE and CaE in comparison to the primary targets AChE and NTE. The medium-toxic diEt-PFP has low inhibitor activity to both primary targets and a very low neuropathic potential (RIP=0.07). The low toxic diBu-PFP has rather high activities against both the target enzymes with preferable NTE inhibition and correspondingly has a high neuropathic potential (RIP=6.6). A high toxic PrDChVP is a strong inhibitor of both AChE and NTE and has a high neuropathic potential (RIP=3.5).

Table 1. In vitro inhibitor activity (ki, M-1min-1) of dialkylphosphates diEt-PFP, diBu-PFP and PrDChVP

against individual enzymes AChE, NTE, BChE, CaE and acute toxicity of OPC.

ki, M-1min-1 LD50, mg/kg,

Mice, i.p., 24h

OPC AChE NTE BChE CaE

diEt-PFP (1.80±0.11)×103

(1.31±0.08)×102

(4.50±0.19)×103

(1.02±0.04)×105

200(161.1÷238.9)

diBu-PFP (8.44±0.34)×104

(5.6±0.18)×105

(2.47±0.43)×106 (1.09±0.08)×107 >2000

PrDChVP (5.97±0.12)×105

(2.10±0.16)×106

(1.03±0.11)×107

(4.7±0.21)×107

15(13.4÷17.3)

Page 23: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

21

Table 2. Inhibitor selectivity ofdiEt-PFP, diBu-PFP and PrDChVP to individual enzymes AChE, NTE, BChE and CaE

Selectivity = ratio of corresponding ki values: ki(EOHX) / ki(EOHY)OPC NTE/AChE =RIP BChE/AChE CaE/AChE BChE/ NTE CaE/ NTE

diEt-PFP 0.07 2.5 56.6 34.6 778.6

diBu-PFP 6.6 29.3 129 4.4 19.5

PrDChVP 3.5 17.3 78.7 4.9 23.5

OPC effects in vivo Inhibition of AChE, NTE, BChE and CaE in blood of mice was studied 1 h after

i.p. administration of increasing doses of diEt-PFP, diBu-PFP and PrDChVP. The data obtained are presented in Figs. 2-4.

diEt-PFP (Fig. 2) inhibited BChE and CaE in mice blood in dose-dependent manner. By analyzing the dose-response curves in Figure 2, the ED50 values (median effective doses) for inhibition of BChE and CaE in mouse blood by this OPC were obtained: ED50(BChE) = 46.8±1.5, ED50(CaE ) = 25.0±1.0 mg/kg. As for primary target enzymes, in the maximal administred dose AChE was inhibited by about 30% and no signifi cant inhibitory effect was observed for NTE. Based on the available data (Fig. 2) we presumed that the value of ED50(AChE) should be more than 300 mg/kg (Table 3).

This OPC has medium acute cholinergic toxicity (LD50 = 200 mg/kg, Table 1) and negligible neuropathic potential (Table 2). Thus, blood BChE and CaE are more sensitive biomarkers of exposure to this OPC than blood AChE, and they can be considered as the biomarkers of acute toxicity.

Figure 2. Dose-related BChE and CaE inhibition in mice blood given diEt-PFP in 1h after i.p. administration. The results are % control value for each esterase expressed as mean± SEM, n = 6.

Esterase activities in blood of the control animals: μmol/min/ml, Mean ± SEM: AChE – 0.829±0.035 (N=18),BChE – 0.625±0.034 (N=20), CaE – 6.02±0.27 (N=20), NTE – 0.017 ± 0.003 (N=6).

1 10 100

0

20

40

60

80

100 diEt-PFP

AChE NTE BChE CaE

bloo

d es

tera

ses

inhi

bitio

n, %

of c

ontro

l

Dose diEt-PFP, mg/kg

Page 24: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

22

For diBu-PFP (Fig.3), inhibition of all four blood esterases was clearly dose dependent. By analyzing the dose-response curves in Figure 3, the median effective doses for inhibition of AChE, NTE, BChE and CaE in mouse blood by this compound were obtained: ED50(AChE) = 154±5, ED50(NTE) = 36.3±3.6, ED50(BChE) = 25.1±3.6, ED50(CaE ) = 3.1±0.3 mg/kg (Table 3).

DiBu-PFP has low acute toxicity (LD50>2000 mg/kg, Table 1) and the high neuropathic potential (RIP = 6.6). After dosing mice with this OPC, blood NTE was inhibited in much less doses than AChE (ED50 36.3 and 154 mg/kg, correspondingly). The ratio ED50(AChE)/ED50(NTE) = 4.24 agrees with the diBu-PFP RIP value equal 6.6 (Table 2) and indicates to high neuropathic hazard for this low toxic compound. BChE and especially CaE were inhibited in this case in lower doses than NTE and AChE. Thus, blood BChE and CaE are also more sensitive biomarkers of exposure to diBu-PFP than primary target enzymes, and in the case of this low toxic OPC they can be considered as biomarkers of delayed neurotoxicity.

PrDChVP also inhibited all four blood esterases in mice in dose-dependent manner (Fig. 4). By analyzing the dose-response curves in Figure 4, the median effective doses for inhibition of AChE, NTE and BChE in mouse blood by PrDChVP were obtained: ED50(AChE) = 4.0±0.2, ED50(NTE) = 2.0±0.1, ED50(BChE) = 1.58±0.11 mg/kg. The high degree of inhibition observed for CaE (70%) in minimal administred dose of PrDChVP (0.3 mg/kg) prevented the ED50 calculation. Based on the available data we presumed that the value of ED50(CaE ) should be less than of 0.2 mg/kg (Table 3).

PrDChVP is the compound possessing the high acute toxicity (LD50=15 mg/kg, Table 1), high inhibitor activity against AChE and NTE (Table 1) and high neuropathic potential (RIP = 3.5, Table 2). It inhibits blood AChE in low doses: ED50(AChE) = 4.0±0.2 mg/kg, whereas blood NTE is inhibited by even lower doses: ED50(NTE) = 2.0±0.1 mg/kg.

The ratio ED50(AChE)/ED50(NTE) = 2 agrees with the high PrDChVP RIP value equal 3.5 (Table 2) and indicates to high neuropathic hazard for this compound. If poisoning

Figure 3. Dose-related AChE, NTE, BChE and CaE inhibition in mice blood given diBu-PFP in 1h after i.p. administration. The results are % control value for each esterase expressed as mean± SEM,

n = 6 Esterase activities in the control animals are shown in the legend to Figure 2

0.1 1 10 100 1000

0

20

40

60

80

100 diBu-PFP

AChE NTE BChE CaE

bloo

d es

tera

ses

inhi

bitio

n, %

of c

ontro

l

Dose diBu-PFP, mg/kg

Page 25: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

23

occurs with this compound, OPIDN can develop after successful treatment of acute cholinergic toxicity.

Figure 4. Dose-related AChE, NTE, BChE and CaE inhibition in mice blood given PrDChVP in 1h after i.p. administration. The results are % control value for each esterase expressed as mean± SEM, n = 6.

Esterase activities in the control animals are shown in the legend to Figure 2

BChE and especially CaE are inhibited after PrDChVP administration in lower doses than NTE and AChE (Fig.4). Thus, blood BChE and CaE are also more sensitive biomarkers of exposure to PrDChVP than primary target enzymes, and in this case they are biomarkers both of acute and delayed neurotoxicity.

The calculated ED50 values (median effective doses) for inhibition of AChE, NTE, BChE and CaE in mice blood by 3 OPCs possessing different esterase profi le, different acute toxicity and different neuropathic potential are summarized in Table 3. The ED50 values characterize sensitivity of the blood esterases as biomarkers of exposure to each OPC.

Table 3. Median effective doses (ED50, mg/kg) for diEt-PFP, diBu-PFP and PrDChVP as esterases inhibitors in mouse blood 1h after i.p. compounds administration.

OPCED50, mg/kg ED50( NTE)/

ED50(AChE)AChE NTE BChE CaE

diEt-PFP>300

30% inhibition at 200 mg/kg

No inhibition at 200 mg/kg

46.8 ± 1.5 25.0 ± 1.0 0

diBu-PFP 154 ± 5 36.3 ± 3.6 25.1± 3.6 3.1±0.3 4.24

PrDChVP 4.0 ± 0.2 2.0 ± 0.1 1.58±0.11< 0.2

70% inhibition at 0.3 mg/kg

2.0

Analysis of the results presented in Table 3 shows that scavenging esterases BChE and CaE are more sensitive biomarkers than blood AChE and NTE are, and they can therefore detect exposure to lower doses of OP toxicants. This conclusion agrees with the literature data [1,2,31]. Determining decreases in BChE and CaE activities allows us to reveal exposure to

0.1 1 10

0

20

40

60

80

100 PrDChVPbl

ood

este

rase

s in

hibi

tion,

% o

f con

trol

Dose PrDClVP, mg/kg

AChE NTE BChE CaE

Page 26: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

24

lower doses and to get information about the exposure to OP toxicants, as such [1]. BChE and CaE can be sensitive to both conventional and delayed neuropathic agents; therefore, their inhibition could serve as a general biomarker for OP agents. The simultaneous determination of AChE and NTE in blood has the potential to discriminate between exposures to acute or delayed neurotoxic agents. The ratio between NTE and AChE inhibition, measured in blood, characterizes the probability of OPIDN development versus acute cholinergic toxicity. Inhibition of all of blood esterases was shown to be dose-dependent, so, the level of blood esterases inhibition allows us to assess the level of exposure, i.e. perform dosimetry of the exposure, which provides valuable information for medical treatment.

Thus, esterase status can be considered as the effective and informative complex biomarker of exposure to OPCs. The electrochemical biosensor system to be developed in the NATO SfP project 984082 for routine on site “point-of-care” monitoring of blood esterases and esterase status determination, enables real time, sensitive and specifi c OPC exposure assessment, assisting in accurate diagnostics of poisoning, prognosis of intoxication and optimization of therapy.

This work is supported by NATO Science for Peace and Security Program (grant no SfP 984082). We are grateful to Dr. Alexey Aksinenko for the synthesis of compounds for research, as well Tatyana Galenko and Olga Serebryakova for their assistance with the experiments on mice.

References

[1] L.G. Costa, Biomarker research in neurotoxicology: The role of mechanistic studies to bridge the gap between laboratory and epidemiological investigations, Environ Health Perspect 104 (1996), (Suppl.1), 55-67.

[2] NRC (National Research Council) Biological markers in environmental health research, Environ Health Perspect 74 (1987), 3-9.

[3] C.M. Thompson., R.J. Richardson, Anticholinesterase insecticides. In: Pesticide Toxicology and International Regulation, T.C. Marrs, B. Ballantyne Editors. New York, Wiley, 2004, 89-127.

[4] R.J. Richardson, Organophosphate Poisoning, Delayed Neurotoxicity. In: Encyclopedia of Toxicology, P. Wexler Editor. Second ed. Oxford, Elsevier, 3 (4 vols.), 2005, 302-306.

[5] G.F. Makhaeva, V.L. Yankovskaya, N.V. Kovaleva, V.I. Fetisov, V.V. Malygin, N.A.Torgasheva., B.A. Khaskin, Antiesterase activity and toxicity of O,O-Dialkyl-S-ethoxycarbonyl bromomethylthiolphosphates, Russian J Bioorganic Chem 25 (1999), 6-10.

[6] M. Jokanovic, Biotransformation of organophosphorus compounds, Toxicology 166 (2001), 139-160.[7] P. Masson, O Lockridge, Butyrylcholinesterase for protection from organophosphorus poisons: catalytic

complexities and hysteretic behavior. Arch Biochem Biophys 494 (2010), 107-120[8] J.E. Casida, G.B. Quistad, Organophosphate toxicology: safety aspects of nonacetylcholinesterase

secondary targets, Chem Res Toxicol 17 (2004), 983-98.[9] E.S. Peeples, L.M. Schopfer, E.G. Duysen, R. Spaulding, T. Voelker, C.M. Thompson, O. Lockridge,

Albumin, a new biomarker of organophosphorus toxicant exposure, identifi ed by mass spectrometry, Toxicol Sci 83 (2005), 303-12.

[10] M.H. Tarhoni, T. Lister D.E. Ray, W.G. Carter, Albumin binding as a potential biomarker of exposure to moderately low levels of organophosphorus pesticides, Biomarkers 13 (2008), 343-63.

[11] J. Bajgar, Biological monitoring of exposure to nerve agents, Br J Ind Med 49 (1992), 648-653.[12] R.J. Richardson, R.M. Worden, G.F. Makhaeva, Biomarkers and biosensors of delayed neuropathic agents,

In: Handbook of Toxicology of Chemical Warfare Agents, R.C. Gupta, Ed., Academic Press/Elsevier, Amsterdam, 2009, 859-876.

[13] D. Bertoncin, A. Russolo, S, Caroldi, M. Lotti, Neuropathy target esterase in human lymphocytes, Arch Environ Health 40 (1985), 221-230.

[14] B.R. Dudek, R.J. Richardson, Evidence for the existence of neurotoxic esterase in neuronal and lymphatic tissue of the adult hen, Biochem Pharmacol 31 (1982), 1117-1121.

[15] M. Maroni, M.L. Bleecker, Neuropathy target esterase in human lymphocytes and platelets, J Appl Toxicol 6 (1986), 1-7.

Page 27: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

25

[16] R. J. Richardson, B.R. Dudek, Neurotoxic esterase: Characterization and potential for a predictive screen for exposure to neuropathic organophosphates, In: Pesticide Chemistry: Human Welfare and the Environment, J. Miyamoto, P.C. Kearney Editors. Oxford, Pergamon, 3, 1983, 491-495.

[17] M. Lotti, Biological monitoring for organophosphate-induced delayed polyneuropathy, Toxicol Lett 33 (1986), 167-172.

[18] M. Lotti, C.E. Becker, M.J. Aminoff, J.E. Woodrow, J.N. Seiber, R.E. Talcott, R.J. Richardson, Occupational exposure to the cotton defoliants DEF and merphos. A rational approach to monitoring organophosphorus-induced neurotoxicity, J Occup Med 25 (1983), 517-522.

[19] M. Lotti, A. Moretto, R. Zoppellari, R. Dainese, N. Rizzuto, G. Barusco, Inhibition of lymphocytic neuropathy target esterase predicts the development of organophosphate-induced delayed polyneuropathy, Arch Toxicol 59 (1986), 176-179.

[20] B.W. Schwab, R.J. Richardson, Lymphocyte and brain neurotoxic esterase: Dose and time dependence of inhibition in the hen examined with three organophosphorus esters, Toxicol Appl Pharmacol 83 (1986), 1-9.

[21] M. Lotti, Organophosphate-induced delayed polyneuropathy in humans: Perspectives for biomonitoring, Tr Pharmacol Sci 81 (1987), 176-177.

[22 ] L.V. Sigolaeva, A. Makower, A.V. Eremenko, G.F. Makhaeva, V.V. Malygin, I.N. Kurochkin, and F. Scheller, Bioelectrochemical analysis of neuropathy target esterase activity in blood, Anal Biochem 290 (2001), 1-9.

[23] G. F. Makhaeva, L.V. Sigolaeva, L.V. Zhuravleva, A.V. Eremenko, I.N. Kurochkin, V.V. Malygin, and R.J. Richardson, Biosensor detection of Neuropathy Target Esterase in whole blood as a biomarker of exposure to neuropathic organophosphorus compounds, J Toxicol Environ Health Part A, 66 (2003), 599-610.

[24] L. G. Sokolovskaya, L.V. Sigolaeva, A.V. Eremenko, I.V. Gachok, G.F. Makhaeva, N.N. Strakhova, V.V. Malygin, R.J. Richardson, I.N. Kurochkin, Improved electrochemical analysis of neuropathy target esterase activity by a tyrosinase carbon paste electrode modifi ed by 1-methoxyphenazine methosulfate, Biotechnol Lett 27 (2005), 1211–1218.

[25] G. F. Makhaeva, V.V. Malygin, N.N. Strakhova, L.V. Sigolaeva, L.G. Sokolovskaya, A.V. Eremenko, I.N. Kurochkin and R.J. Richardson, Biosensor assay of neuropathy target esterase in whole blood as a new approach to OPIDN risk assessment: review of progress, Hum Exp Toxicol 26 (2007), 273-282.

[26] B.W. Wilson, J. D. Henderson, Blood esterase determinations as markers of exposure, Rev Environ Contam Toxicol 128 (1992), 55-69.

[27] R. J. Richardson, Assessment of the neurotoxic potential of chlorpyrifos relative to other organophosphorus compounds: a critical review of the literature, J Toxicol Environ Health 44 (1995), 135-165.

[28] L. G. Costa, T.B. Cole, G.P. Jarvik, C.E. Furlong, Functional genomic of the paraoxonase ( PON1) polymorphisms: effects on pesticide sensitivity, cardiovascular disease, and drug metabolism, Annu Rev Med 54 (2003), 371-392.

[29] L. G. Sokolovskaya, L.V. Sigolaeva, A.V. Eremenko, I.N. Kurochkin, G.F. Makhaeva, V.V. Malygin, I.E. Zykova, V.I. Kholstov, N.V.Zavyalova Family of biosenor analyzers for assessment of “ esterase status” of organism. Russian Chemical Journal, No1-2 (N.V. 13-14) (2004), 21-31.

[30] G. F. Makhaeva, E.V. Rudakova, N.P. Boltneva, L.V. Sigolaeva, A.V. Eremenko, I.N. Kurochkin, and R.J. Richardson. Blood esterases as a complex biomarker for exposure to organophosphorus compounds, In: Counteraction to Chemical and Biological Terrorism in the East Europe Countries, NATO Security through Science Series A, 2009, 177–194.

[31] M. Maroni, C. Colosio, A. Ferioli, A. Fait, Biological Monitoring of Pesticide Exposure: a review, Introduction Toxicology 143 (2000), 1-118.

[32] G. F. Makhaeva, O.G. Serebryakova, N.P. Boltneva, T.G. Galenko, A.Yu. Aksinenko, V.B. Sokolov, I.V. Martynov, Esterase profi le and analysis of structure – inhibitor selectivity relationships for homologous phosphorylated 1-hydroperfl uoroisopropanoles, Doklady Biochem Biophys 423 (2008), 352-357. [translated from Russian Dokl Akad Nauk 423 (6) 2008, 826-831].

[33] E. V. Rudakova, G.F. Makhaeva, T.G. Galenko, A.Yu. Aksinenko, V.B. Sokolov, I.V. Martynov (2013) New selective inhibitor of mice plasma carboxylesterase, Doklady Biochem. Biophys. V. 449, P.87-89 [translated from Russian Dokl Akad Nauk, 449 (2) 2013, 232–235]

[34] J. R. Albert, S.M. Stearns, Delayed neurotoxic potential of a series of alkyl esters of 2, 2-dichlorovinyl phosphoric acid in the chicken, Toxicol Appl Pharmacol 29 (1974), 136.

[35] E. V. Rudakova, L.V. Sigolaeva, G.F. Makhaeva, Investigation of mice blood neuropathy target esterase as biochemical marker of exposure to neuropathic organophosphorus compounds, In: Problems of Toxicology, C. Dishovsky and J. Radenkova – Saeva, Editors, 2014, Sofi a, (IN PRESS).

[36] M. Lotti, M.K. Johnson, Neurotoxicity of organophosphorus pesticides: Predictions can be based on in vitro studies with hen and human enzymes, Arch Toxicol 41 (1978), 215-221.

[37] E. V. Rudakova, N.P. Boltneva, G.F. Makhaeva, Comparative analysis of esterase status of human and rat blood, Bull Exp Biol Med 152(1) (2011), 73-75 [Translated from Byulleten’ Eksperimental’noi Biologii i Meditsiny, 152 (7) (2011), 80-82.]

Page 28: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

26

[38] I. N. Kurochkin, A.V., L.V.Sigolaeva, A.V. Eremenko, E.A.Dontsova, M.S.Gromova, E.V. Rudakova, G.F. Makhaeva, Layer-by-layer electrochemical biosensors for blood esterase assay. In: Problems of Toxicology, C. Dishovsky and J. Radenkova – Saeva, Editors, 2014, Sofi a (In Press)

[39] L.V. Sigolaeva, D.V. Pergushov, C.V. Synatschke, A. Wolf, I. Dewald, I.N. Kurochkin, A. Feryc, A.H.E. MЁuller, Co-assemblies of micelle-forming diblock copolymers and enzymes on graphite substrate for an improved design of biosensor systems, Soft Matter 9 (2013), 2858–2868.

[40] L. V. Sigolaeva, G.F. Makhaeva, E.V. Rudakova, N.P. Boltneva, M.V. Porus, G.V. Dubacheva, A.V. Eremenko, I.N. Kurochkin and R.J. Richardson, Biosensor analysis of blood esterases for organophosphorus compounds exposure assessment: Approaches to simultaneous determination of several esterases, Chem-Biol Interact 187 (2010), 312–317.

[41] L. V. Sigolaeva, G.V. Dubacheva, M.V. Porus, A.V. Eremenko, E.V. Rudakova, G.F. Makhaeva, Rudy J. Richardson and I.N. Kurochkin, A layer-by-layer tyrosinase biosensor for assay of carboxylesterase and neuropathy target esterase activities in blood, Anal Methods 5(16) (2013), 3872-3879.

[42] N.P. Boltneva, E.V. Rudakova, L.V. Sigolaeva, G.F.Makhaeva, Esterase status of various species in assessment of exposure to organophosphorus compounds. In: Problems of Toxicology, C. Dishovsly and J. Radenkova – Saeva, Editors, 2014, Sofi a (IN PRESS).

[43] E.V. Rudakova, O.G. Serebryakova, N.P. Boltneva, T.G. Galenko, G.F. Makhaeva, A biochemical model in mice for assessment of neuropathic potential of organophosphorus compounds, Toxicol Reviews (Russian) No 6 (2012), 20-24.

[44] G. F. Makhaeva, A.Y. Aksinenko, V.B.Sokolov, O.G. Serebryakova, R.J. Richardson, Synthesis of organophosphates with fl uorine-containing leaving groups as serine esterase inhibitors with potential for Alzheimer disease therapeutics, Bioorg Med Chem Lett 19 (2009), 5528-5530.

[45] G.F. Makhaeva, V.V. Malygin, A stable preparation of hen brain neuropathy target esterase for rapid biochemical assessment of neurotoxic potential of organophosphates. Chem-Biol Interact 119-120 (1999), 551–557

[46] G. L. Ellman, K.D. Courtney, V. Andres, Jr., R.M. Featherstone, A new and rapid colorimetric determination of acetylcholinesterase activity, Biochem Pharmacol 7 (1961), 88-95.

[47] M. K. Johnson, Improved assay of neurotoxic esterase for screening organophosphates for delayed neurotoxicity potential, Arch Toxicol 67, (1977), 113–115.

[48] S. Padilla, T.L. Lassiter, D. Hunter, In: Methods in Molecular Medicine, Neurodegeneration Methods and Protocols. Eds J.Harry and H.A.Tilson, Humana Press Inc., N.J. Totowa, 22 (1999), 237-245.

[49] E. Reiner, A. Bosak, V. Simeon-Rudolf, Activity of cholinesterases in human whole blood measured with acetylthiocholine as substrate and ethopropazine as selective inhibitor of plasma butyrylcholinesterase, Arh Hig Rada Toksikol 55 (2004), 1-4.

[50] F. Worek, U. Mast, D. Kiderlen, C. Diepold, P. Eyer, Improved determination of acetylcholinesterase activity in human whole blood, Clin Chim Acta 288 (1999), 73–90.

[51] C. De Vriese, F. Gregoire, R. Lema-Kisoka, M. Waelbroeck, P. Robberecht, C. Delporte, Ghrelin degradation by serum and tissue homogenates: identifi cation of the cleavage sites, Endocrinology 145(11) (2004), 4997-5005.

[52] T.L. Huang, T. Shiotsuki, T. Uematsu, B. Borhan, Q.X. Li, B.D. Hammock, Structure–activity relationships for substrates and inhibitors of mammalian liver microsomal carboxylesterases, Pharm Res 13 (1996), 1495–1500.

[53] S. M. Chanda, S.R. Mortensen, V.C. Moser, S. Padilla, Tissue-specifi c effects of chlorpyrifos on carboxylesterase and cholinesterase activity in adult rats: An in vitro and in vivo comparison, Fundam Appl Toxicol, 38 (1997), 148–157

Page 29: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

27

Chapter 2

Esterase Status of Various Species in Assessment of Exposure to Organophosphorus Compounds

Natalia P. BOLTNEVAa1, Elena V. RUDAKOVAa, Larisa V. SIGOLAEVAb, Galina F. MAKHAEVAa

aInstitute of Physiologically Active Compounds Russian Academy of Sciences Chernogolovka, Moscow Region 142432 Russia

bDepartment of Chemistry, Moscow State University, Moscow 119992 Russia

Abstract. Our project supposes to use an esterase status of an organism as a complex biomarker of exposure and sensitivity to organophosphorus compounds ( OPCs). The esterase status includes fi ve blood esterases: acetylcholinesterase (AChE), neuropathy target esterase ( NTE), butyrylcholinesterase (BChE), carboxylesterase ( CaE) and paraoxonase ( PON1). The level of activity of these enzymes and their relationship to each other are an individual feature of the organism and are determined by its species, age, sex and genetic characteristics. Therefore, a set of esterases which are the most effective biomarkers in assessment of exposure to OPC may be different for various species. In this regard, it is important to determine the esterase status, because such an approach provides the most complete information on the character and extent of the exposure. In this study activities of esterases in the blood samples from humans and rodents (mice and rats) were measured by the spectrophotometric methods (AChE, BChE, CaE, PON1) or electrochemically ( NTE). The obtained sets of the esterase activities were analyzed. The results indicate signifi cant species differences in the esterase status: in humans cholinesterase activities are signifi cant and CaE activity is negligible, while in rats and mice CaE activity is higher and cholinesterase activities are low. The rodents NTE activity is lower than that one in humans. Very high PON1 arylesterase activity has been found in all species.

Key words. Esterase status, blood esterases, organophosphorus compounds, human, rodents

1. Introduction

Much attention has been given in the fi eld of organophosphorus compounds ( OPCs) toxicology to the development of biomarkers to be used as indicators of exposure, health effects and susceptibility. OPCs with anticholinesterase properties are widely used as insecticides; to a less extent they are used as therapeutic agents. Some highly toxic OPCs were produced and used in several countries as chemical warfare agents. OPC of both

1 Laboratory of Molecular Toxicology, Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, Moscow Region 142432, Russia, E-mail: [email protected]

Page 30: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

28

known and unknown structure may also arise as a result of various chemical accidents. Defending against such agents requires rapid, sensitive and specifi c detection of them and their biological effects.

Our project supposes to use of an organism “ esterase status” as a complex biomarker of exposure and individual sensitivity to OPCs [1] andplanstostudyvalidityofthisassumptioninanimalexperiments. The esterase status of an organism includes fi ve blood esterases to which OPCs interact: acetylcholinesterase (EC 3.1.1.7, AChE), butyrylcholinesterase (EC 3.1.1.8, BChE), carboxylesterase (EC 3.1.1.1, CaE), neuropathy target esterase (EC 3.1.1.5, NTE) and paraoxonase (EC 3.1.8.1, PON1).

The nervous tissue enzymes AChE and NTE are the targets of acute toxicity [2] and organophosphate-induced delayed neuropathy (OPIDN) [3] respectively, i.e. the primary targets of OPC action on an organism. RBC AChE activity refl ects the situation at target tissues (especially in peripheral compartments, e.g. neuromuscular junction) and it can be considered as an appropriate parameter for biological monitoring of exposure to OP agents as well as a valuable tool for monitoring and optimization of therapeutic measures.

The fact that an excellent correlation exists between inhibition/aging of NTE just after exposure and OPIDN development 2-3 weeks later is suffi cient to use this information for the development of biomarkers and biosensors for delayed neuropathic agents. NTE has been found in circulating lymphocytes and platelets,and it has been used as a biomarker of animal and human exposure to neuropathic OPCs [3]. We fi rst demonstrated the possibility of determining NTE in whole blood [4] andindicated that NTE inhibition in whole blood refl ected brain NTE inhibition in the experiments on hens [5,6]. We suggest that inhibition of NTE in blood can be used in conjunction with inhibition of blood AChE to assess the likelihood that an exposure to OPCs would produce cholinergic and/or delayed neuropathic effects [1,7].

Nonspecifi c esterases BChE and CaE are the secondary targets, which act as stoichiometric scavengers of OPСs, reducing the concentration of the active compound in blood [1,8-10]. Esterase status of an organism was shown to refl ect its age- and gender-related differences in sensitivity to OPCs, and can change during development. So, for example, young rats had considerably less CaE activity, and adult females had less liver CaE activity than males. These differences in detoxifying enzyme correlate with the age-related differences in behavioral and biochemical effects of OPC, as well as the gender differences seen in adult rats, and thus may be a major infl uence on the differential sensitivity to OPCs [11].

Scavenging esterases BChE and CaE tend to be more sensitive biomarkers than RBC AChE is, and they can therefore detect exposure to lower doses of OP toxicants, e.g. pesticides [1,2,12,13]. Determining decreases in BChE and CaE activities allows us to reveal exposure to lower doses and to get information about the exposure to OP toxicants, as such [1]. It should be noted that BChE and CaE can be sensitive to both conventional and delayed neuropathic agents; therefore, their inhibition could serve as a general biomarker for OP agents [1,14]. However, recently it was demonstrated that human plasma and whole blood contain extremely low level of CaE in contrast to mouse, rat, rabbit, horse, cat, and tiger that have high amounts of plasma CaE [15,16]. This suggests a low OPC-scavenging role of human blood CaE and low role of human blood CaE as a biomarker in contrast to blood CaE in rodents and other animals.

Page 31: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

29

Paraoxonase ( PON1) can hydrolyze and detoxify OPCs and acts as a catalytic scavenger. [17]. PON1 is thought to be an important determinant of an individual’s sensitivity to some OPCs, based primarily on evidence from studies on animals models [18,19]. High PON1 activity is found in serum and liver. Animals with low paraoxonase levels (e.g., birds) were more sensitive to specifi c OPCs than animals with high enzyme levels (e.g., rats and especially rabbits) [17]. The available data on the developmental time course of the appearance of PON1 in plasma showed that the serum PON1 activity is low in newborns and infants, and increases gradually during early development [20]. These data suggest that a higher sensitivity of young animals to OP toxicants could be explained, at least partially, by a defi ciency in PON1 activity [21,22]. Decreased serum PON1 activity can result in an increased sensitivity to OP toxicants upon exposure [17].

Genetic polymorphisms of esterases involved in OPCs interactions may lead to enzyme variants with a higher or lower catalytic activity and/or with different levels of expression. It allows us to suggest that polymorphisms of esterases can play essential role in sensitivity to OPCs and intoxications development.

A large number of polymorphism cases have been described for BChE [23]. Most of the identifi ed genetic variants, which are usually grouped in four categories (silent, K-variant, atypical and fl uoride-resistant), are silent, i.e. they have 0 or less than 10% of normal activity. The persons with atypical BChE have abnormal reaction to short-term acting muscle relaxants succinyldicholine (apnoea and the long- term paralysis) [24]. People with low or no BChE activity have a diminished OPC-scavenging ability and therefore may be more susceptible to OPCs toxicity. Such patients have a greater risk of development both acute and delayed OPCs neurotoxic effects.

In human populations, serum paraoxonase exhibits a substrate-dependent polymorphism as well as a large variability in plasma levels among individuals. The Q192R polymorphism, Gln(Q)/Arg(R) at position 192, imparts different catalytic activities toward some OP substrates [25]. The polymorphism at position -108 (T/C), in the promoter region of PON1, is the major contributor to differences in the level of PON1 expression and appears to have the major effect on the levels of PON1 found in plasma of individuals [26]. These two factors determine to a great extent an individual’s sensitivity to OPCs exposure [27, 28].

As we have seen, the level of activity of these blood esterases and their relationship to each other are an individual feature of the organism and are determined by its species, age, sex and genetic characteristics. Therefore, a set of esterases activities which are the most effective biomarkers in assessment of exposure to OPCs may be different for various species and may vary within a species. In this regard, it is important to determine the esterase status, because such an approach provides the more complete information on the character and extent of the exposure. Furthermore, it is important to have data on the basal activities of the esterase status enzymes for experimental animals and humans. Currently, the analysis of such information is complicated because there are only scattered data in the literature on the esterase activity for both various species and specifi c enzymes.

In this study, we performed a detailed investigation of the fi ve esterase status enzymes of human and rodents as standard laboratory animals. Individual basal activities of AChE, BChE, CaE, NTE and PON1 were measured in the blood of humans, rats, and mice and the differences in the esterase status of humans and rodents were evaluated.

Page 32: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

30

2. Materials and methods

Blood samplingWhite outbred male albino mice (18-25 g) and Wistar male rats (160-180 g) were

used in the experiments. The animals were sacrifi ced by decapitation under CO2-induced anesthesia. Trunk blood from each animal was collected immediately in glass vials containing citrate (3.8% (w/v) sodium citrate, 0.2 ml citrate/ml blood). Heparin (20 μl of 500 U/ml solution) served as the anticoagulant. The blood from 5 anonymous human (female) donors was collected in an inpatient setting using a Vacuet vacuum system into tubes with 3.8% sodium citrate as the anticoagulant. Blood samples were aliquoted, frozen in liquid nitrogen, and stored at -70oC before use.

Preparation of blood and plasma for analysisThe plasma was separated by centrifugation at 2500Чg for 15 min, frozen in liquid

nitrogen, and stored at -70oС until measurements. Frozen blood was thawed in water bath with ice, after which 1:100 hemolysate specimens were obtained by rapid dilution of one blood volume in 100 volumes of cold buffer. After thorough mixing, the aliquots of hymolysates were directly frozen in liquid nitrogen for more complete hemolysis and stored at -20oC until analysis. Before analysis the samples were thawed and kept on ice. Selection of substrates concentration was carried out based on the obtained Km values for rodent and human blood.

The volume of blood/plasma for analysis of each esterase was selected from the linear part of the dependence of the rate of hydrolysis of the corresponding substrate on the blood/plasma concentration (evaluated in a special experiment). The data allowed us to select the concentration of the substrate and the volume of blood/plasma for the standard determination of each enzyme activity.

AChE/BChE assayActivity of AChE in the whole blood and plasma was evaluated by the rate of hydrolysis

of 1 mM acetylthiocholine by Ellman’s colorimetric method [29] in 0.1 M K,Na-phosphate buffer (pH 7.5) at 25oC in the presence of a specifi c inhibitor of BChE 0.02 mM ethopropazine [30-33]. The measurements were carried out in plasma at λ 412 nm (ε412 = 14150 M-1cm-1). In order to minimize the impact of hemoglobin absorption, the measurements in the hemolysed and diluted whole blood were carried out at λ 436 nm (ε436 = 10600 M-1cm-1) [33]. Activity of BChE in the whole blood and plasma was measured under the same conditions with 1 mM butyrylthiocholine as a substrate. The selective inhibitor of AChE is not required, since AChE activity for butyrylthiocholine is very low [30].

CaE assaySpectrophotometric determination of CaE in whole blood and plasma was carried out

with 1 mM 1-naphthyl acetate at λ 322 nm (ε322 = 2200 M-1cm-1) [34,35] and 1 mM phenyl acetate at λ 270 nm (ε270 =1310 M-1cm-1) in 0.1 M K,Na-phosphate buffer (pH 8.0) at 25oC. Specifi c inhibitors of PON1/arylesterases (2 mM EDTA) and cholinesterases (40 μM eserine) were used for CaE activity discrimination [36].

PON1 assayArylesterase activity of PON1 in the whole blood and plasma was evaluated by the

4 mM phenyl acetate hydrolysis at λ 270 nm (ε270 = 1310 M-1cm-1) [37]. Paraoxonase

Page 33: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

31

activity activity of PON1 in plasma was evaluated by the rate of 1.2 mM paraoxon hydrolysis at λ 405 nm (ε405 = 18,050 M-1cm-1) [38]. Hydrolytic activity was evaluated by spectrophotometry at 25oC in 0.1 M Tris-HCl buffer (pH 8.0) containing CaCl2 and 40 μM eserine (specifi c cholinesterase inhibitor) [36].

All measurements were carried out on a Gilford-250 spectrophotometer. NTE assay NTE activity was assayed according to the differential inhibition method of Johnson

[39] with an electrochemical endpoint as described previously [4,6]. Measurements were carried out using screen-printed tyrosinase-based biosensor for phenol analysis. Briefl y, blood hemolysate samples were incubated at 37oC with 50 mM paraoxon (sample B) or 50 μM paraoxon plus 250 μM mipafox for 20 min (sample C), followed by incubation with phenyl valerate (fi nal apparent concentration 0.54 μM) for the next 40 min at 37oC. The reaction was stopped by addition of 50 μl of 1% (w/v) aqueous SDS. Phenol product was assayed amperometrically after 20-fold sample dilution in 50mM sodium phosphate with 100mM NaCl (pH 7.0). NTE activity was calculated as the difference in phenol production between samples B and C.

StatisticsStatistical analysis wascarried out using GraphPad Prism version 3.02 for Windows,

GraphPad Software (San Diego, CA). The results are given as means ±SEM. The level of signifi cance was set at p<0.05.

3. Results and Discussion

Selection of substratesTo determine CaE activity, 1-naphthyl acetate (1-NA) and 4-nitrophenyl acetate (4-

NPA) are widely used as the standard substrates [34,36]. Furthermore, we used phenyl acetate (PhA) as a substrate for spectrophotometric validation of biosensor measurements of CaE activity in blood. We performeda series of experiments to determine CaE activity in human, mice and rats blood using 1-NA, PhA, 4-NPA as substrates. The substrate hydrolysis was carried out in the presence of specifi c inhibitors of PON1/arylesterases (2 mM EDTA) and cholinesterase (40 μM eserine) for elimination of PON1 and cholinesterase [36]. The results of measurements showed a good correspondence of CaE activities obtained using 1-NA and PhA in all three blood samples while CaE activity was several times above when 4-NPA was used. The data are presented in Table 1.

Table 1. CaE activities (μmol substrate/min/ml blood) in whole blood of human, rat and mice using 1-NA PhA and 4-NPAas substrates. Results are presented as Mean ± SEM, N=5.

Object

Substrates

1-NA (1мМ) PhA (1мМ) 4-NPA (1мМ)

Human 0.23±0.01 0.25±0.02 5.5±0.40

Rat 3.27±0.21 3.59±0.43 8.5±0.50

Mouse 6.8±0.40 6.30±0.27 12.6±0.70

Page 34: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

32

We performed a series of experiments to study the effect of specifi c inhibitors of CaE, 0.1 mM BNPP [40] and 1 mM iso-OMPA [36], on the hydrolysis of 1-NA, PhA and 4-NPA to fi nd out the cause of this phenomenon. Purifi ed commercial preparation of porcine liver CaE(Sigma) and rat plasma were used as the enzyme sources. The measurements of CaE activity were performed after 0, 10, 30 and 60 min incubation with inhibitors. Residual activity of enzyme was determined for each substrate (control values of CaE activity are shown in Table 1). The results are presented in % of residual CaE activity from the control -rate of substrate hydrolysis in the absence of inhibitor in each of the experiments.

The results of the infl uence of BNPP and iso-OMPA on the hydrolysis of 1-NA, PhA and 4-NPA catalyzed by purifi ed porcine liver CaE are presented in Figure 1. As can be seen from Figure 1, BNPP, and iso-OMPA completely inhibited the hydrolysis of the studied substrates in the presence of purifi ed CaE - porcine liver CaE preparation. In these conditions, the hydrolysis of 1-NA, PhA and 4-NPA is catalyzed only by CaE. Thus, using the purifi ed preparation of CaE allows us to work with all of the mentioned substrates that corresponds to the literature data [41].

The results of the study of inhibition of hydrolysis of 1-NA, PhA and 4-NPA by specifi c inhibitors of CaE in rat plasma are presented in Figure 2.

In contrast to full inhibition of hydrolysis of 1-NA and PhA, 4-NPA hydrolysis in rat plasma (Figure 2) was inhibited only about 40-50% after 60 min incubation with 0.1 mM BNPP and 1mM iso-OMPA. That is, the hydrolysis of 4-NPA in rat plasma is catalyzed not only by CaE. These results agree with the known data on the hydrolysis of 4-NPA by serum albumin [42, 43]. When we used other substrates, 1-NA and PhA, their hydrolysis in rat plasma was fully inhibited by selective CaE inhibitors and the obtained values of CaE activity were in reasonably close agreement with each other [16,44].

0 10 20 30 40 50 600

20

40

60

80

100 porcine liver CaE 1-NA, 1mM

CaE

act

ivity

, % c

ontr

ol

time, min

BNPP 0.1 mM i-OMPA 1 mM

A)

0 10 20 30 40 50 600

20

40

60

80

100 porcine liver CaEPhA, 1mM

CaE

act

ivity

, % c

ontr

ol

time, min

BNPP 0,1mM i-OMPA 1mM

B)

0 10 20 30 40 50 600

20

40

60

80

100 porcine liver CaE4-NPA, 1mM

CaE

act

ivity

, % c

ontr

ol

time, min

BNPP 0.1mM i-OMPA 1mM

C)

Figure1. Theinfluenceof CaEspecificinhibitors0.1 mM BNPP and1 mM iso-

OMPAonthehydrolysisof

A) - 1-NA, B) - PhA, C) - 4-NPAcatalyzedbypurifiedporcineliverCaE.

Page 35: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

33

Based on these fi ndings, we excluded 4-NPA of the number of substrates used for further studies of CaE activity in blood and plasma, and spectrophotometric measurements were performed here using 1-naphthyl acetate [34,36] as substrate.

Determination of AChE, BChE, CaE and PON1 activities in human and rat plasma.The data on the AChE, BChE, CaE and PON1 activities in human and rat plasma are

presented in Table 2 and Figure 3.

Table.2. Activities of “ esterase status” enzymesin human and rat plasma.

Enzymes

Esterase activities,μmol substrate/min/ml plasma

HumanMean±SEM

n=5

RatMean±SEM

n=6

AChE 0.035±0.003 0.216±0.004

BChE 2.21±0.10 0.073±0.003

CaE 0.046±0.002 3.69±0.15

PON1/PhA 63.5±9.70 83.0±12.90

PON1/ paraoxon 0.035±0.003 0.085±0.015

In the plasma of the examined species, arylesterase activity of PON1 (with phenylacetate as the substrate) was the highest, while paraoxonase activity of PON1 (substrate paraoxon) was signifi cantly lower (Table 2, Figure 3). Both PON1 activities (arylesterase and paraoxonase) in rat plasma were higher than in human plasma. Virtually complete absence of AChE activity in human plasma and its appreciable level in rat plasma

0 10 20 30 40 50 600

20

40

60

80

100 rat plasma CaE1-NA, 1mM

CaE

act

ivity

, % c

ontr

ol

time, min

BNPP 0.1mM i-OMPA 1mM

A)

0 10 20 30 40 50 600

20

40

60

80

100 rat plasma CaEPhA, 1mM

CaE

act

ivity

, % c

ontr

ol

time, min

BNPP 0.1mM i-OMPA 1mM

B)

0 10 20 30 40 50 600

20

40

60

80

100 rat plasma CaE4-NPA, 1mM

CaE

act

ivity

, % c

ontr

ol

time, min

BNPP 0.1mM i-OMPA 1mM

C)

Figure 2.Inhibitionofhydrolysisofthreesubstrates:A)- 1-NA, B) - PhA,

C) - 4-NP AinratplasmawithspecificinhibitorsofCaE 0.1 mM BNPPand 1 mM iso-OMPA.

Page 36: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

34

was worthy of note. This fact was explained by the location of AChE protein in human blood in erythrocytes only, while in rats the AChE protein was present in both erythrocytes and plasma [33]. Activity of BChE in human plasma was 30 times higher than in rats. On the contrary, activity of CaE was extremely low in human plasma and high in rats. This was in good agreement with the data on negligible level of CaE protein and its activity in human plasma [15, 45].

Determination of AChE, BChE, CaE, NTE and PON1 activities in whole blood of human, mice and rats.

Activities of esterase status enzymes in the whole blood of humans, mice, and rats are presented in Table 3 and Figure 4. The proportions of these enzymes activities were similar to the picture in the plasma. Arylesterase activity of PON1 predominated in the blood of all studied species. Cholinesterase activities in human blood were higher than in rodents (Table 3, Figure 4). AChE in human blood was about 5-fold more active than in rodents, and BChE activity was 2- and 11-fold higher, respectively. Experiments showed that CaE activity in human blood was somewhat higher than in the plasma, presumably because of CaE presence in monocytes [46] and leukocytes [47]. However, CaE activity in the blood of humans was 15-fold lower than in rats and 29-fold lower than in mice.

Table 3.Activities of “ esterase status” enzymes in human mice and rat whole blood.U* - nmol substrate/min/ml blood for NTE activity.

Enzymes

Esterase activities,μmol substrate/min/ml blood

HumanMean±SEM

n=5

MouseMean±SEM

n=24

RatMean±SEM

n=6AChE 4.16±0.14 0.829±0.035 0.723±0.060BChE 1.4±0.08 0.710±0.061 0.128±0.010

CaE 0.25±0.01 7.15±0.18 3.59±0.43

PON1/PhA 34.6±4.1 28.2±6.40n=26 37.3±3.20

NTE, U* 56.8 ± 5.31n=3

18.5 ± 2.13n=9

15.62 ± 2.34n=4

Figure 3. AChE, BChE, CaE and PON1 activities (μmol substrate/min/ml plasma) in human and rat plasma (N = 6). A) PON1/ PhA activity is included, B) PON1/PhA activity is not included.

Page 37: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

35

Figure 4. AChE, BChE, CaE, NTE and PON1 activities (μmol substrate/min/ml blood) in human (N =5), mice (N =24)and rat (N = 6) whole blood. A) PON1/PhA is not included, B) PON1/PhA is included.

Our data on activities of AChE, BChE, CaE, and PON1 correspond well to the literature data [46,48,49,32,33] and are in good agreement with recent data on these esterase proteins distribution in human, mouse, and rat plasma and erythrocytes, obtained by PAAG native electrophoresis [50].

Spectrophotometric technique does not allow determining NTE activity in whole blood. NTE assay in whole blood became possible only using a high sensitive tyrosinase-based biosensor for phenol analysis [4]. In this study blood NTE activity in human and mice was measured with new screen-printed tyrosinase biosensor SPE/PDDA/ tyrosinase, and for the rat NTE we used the data obtained with the LBL tyrosinase biosensor of construction GR/PDDA/ tyrosinase/GA (GR – graphite rod, PDDA – poly-(dimethyldiallylammonium chloride), GA - glutaraldehyde) [51]. Current results on human and mice NTE are in good agreement with our previous data obtained using similar or other biosensors and various groups of humans (42.5±4.28 [51], 36.9±5.62 [52], 32.3±3.6 nmol/min/ml blood [4]) and mice (17.3±2.95 nmol/min/ml blood [51], 15.3±2.32 [52]). The results demonstrate that the rodents blood NTE activity is about 3-fold lower than that one in humans. Although the literature values for rodent blood NTE are apparently lacking, it is of interest to note that rat and mouse brain NTE activities are known to be lower than that of human brain [53–56].

Thus, analysis of the esterase status of humans and rodents showed high levels of arylesterase activity of PON1 in all the studied species. In human blood, esterase activity was presented (in addition to PON1) mainly by the erythrocyte AChE and plasma BChE, while CaE activity was negligible. Rodents exhibited high activity of plasma CaE, its activity being the maximum in mice, while cholinesterase activities were low. Signifi cant differences in species esterase status indicate that different esterases can be effective biomarkers of OPC exposure in various species. So, in humans, blood BChE can be the most sensitive biomarkers which have the potential to provide information about the level of exposure of an individual [57], whereas in mice, blood CaE along with BChE can play such a role [1]. It should be taken into account in the design of toxicological experiments on studying the esterase status as a complex biomarker of OPC exposure. Importantly,

Page 38: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

36

esterase activities in mice are higher than in rats, which should be taken into consideration when selecting the experimental model for studies of compounds containing ester or amide groups. Comparative data on the basal esterase activities in humans and laboratory animals (mice and rats) are essential information to use of an organism “ esterase status” as a complex biomarker of exposure and individual sensitivity to OPC.

This work is supported by NATO Science for Peace and Security Program (grant no SfP 984082). We are grateful to Olga Serebryakova for her assistance with the experiments on mice and rats.

References

[1] G.F. Makhaeva, E.V. Rudakova, N.P. Boltneva, L.V. Sigolaeva, A.V. Eremenko, I.N. Kurochkin, R.J. Richardson, Blood Esterases as a Complex Biomarker for Exposure to Organophosphorus Compounds. In: “Counteraction to Chemical and Biological Terrorism in the East Europe Countries”, Eds C. Dishovsky, A. Pivovarov, NATO Security through Science Series A, Springer, (2009,) 177-194.

[2] C.M. Thompson, R.J. Richardson, Anticholinesterase insecticides. Pesticide Toxicology and International Regulation, T.C. Marrs, B. Ballantyne Editors. New York, Wiley, (2004), 89-127.

[3] R.J. Richardson, Organophosphate Poisoning, Delayed Neurotoxicity, Encyclopedia of Toxicology, P. Wexler Editor. Second ed. Oxford, Elsevier, 3 (4 vols.) (2005), 302-306.

[4] L.V. Sigolaeva, A. Makower, A.V. Eremenko, G.F. Makhaeva, V.V. Malygin, I.N. Kurochkin, F.W. Scheller, Bioelectrochemical analysis of neuropathy target esterase activity in blood, Anal Biochem 290 (2001), 1–9.

[5] G.F. Makhaeva, V.V. Malygin, N.N. Strakhova, L.V. Sigolaeva, L.G. Sokolovskaya, A.V. Eremenko, I.N. Kurochkin, and R.J. Richardson, Biosensor assay of neuropathy target esterase in whole blood as a new approach to OPIDN risk assessment: review of progress, Hum Exp Toxicol 26 (2007), 273-282.

[6] G.F. Makhaeva, L.V. Sigolaeva, L.V. Zhuravleva, A.V. Eremenko, I.N. Kurochkin, V.V. Malygin, R.J. Richardson, Biosensor detection of neuropathy target esterase in whole blood as a biomarker of exposure to neuropathic organophosphorus compounds, J Toxicol Environ Health Part A 66 (2003), 599–610.

[7] E.V. Rudakova, G.F. Makhaeva, L.V. Sigolaeva, Assessment of Possibility of Using Mice Blood Neuropathy Target Esterase as Biochemical Marker for Exposure to Neuropathic organophosphorus Compounds, In: Problems of Toxicology, Ch. Dishovsky Editor, (IN PRESS).

[8] G.F. Makhaeva, V.L. Yankovskaya, N.V. Kovaleva, V.I. Fetisov, V.V. Malygin, N.A. Torgasheva, B.A. Khaskin, Antiesterase activity and toxicity of O,O-Dialkyl-S-ethoxycarbonyl bromomethylthiolphosphates, Russian J Bioorganic Chem 25 (1999), 6-10.

[9] M. Jokanovic, Biotransformation of organophosphorus compounds, Toxicology 166 (2001), 139-160.[10] J.E. Casida, G.B. Quistad, Organophosphate toxicology: safety aspects of nonacetylcholinesterase

secondary targets, Chem Res Toxicol 17 (2004), 983-98.[11] V.C. Moser, S.M. Chanda, S.R. Mortensen, S. Padilla, Age- and gender-related differences in sensitivity to

chlorpyrifos in the rat refl ect developmental profi les of esterase activities, Toxicol Sci 46(2) (1998), 211-22.[12] B.W. Wilson, J. D. Henderson, Blood esterase determinations as markers of exposure, Rev Environ

Contam Toxicol 128 (1992), 55-69.[13] R.J. Richardson, Assessment of the neurotoxic potential of chlorpyrifos relative to other organophosphorus

compounds: a critical review of the literature, J Toxicol Environ Health 44 (1995), 135-165.[14] R.J. Richardson, R.M. Worden, G.F. Makhaeva, Biomarkers and Biosensors of Delayed Neuropathic

Agents, Handbook of the Toxicology of Chemical Warfare Agents, R.C. Gupta Editor. Elsevier, (2009), 859-876.

[15] B. Li, M. Sedlacek, I. Manoharan, R. Boopathy, E.G. Duysen, P. Masson, O. Lockridge, Butyrylcholinesterase, paraoxonase, and albumin esterase, but not carboxylesterase, are present in human plasma, Biochem Pharmacol 70 (2005), 1673-1684.

[16] E.V. Rudakova, N.P. Boltneva, G.F. Makhaeva, Comparative analysis of esterase status of human and rat blood, Bull Exp Biol Med 152(1) (2011), 73-75.

[17] L.G. Costa, W.F. Li, R. Richter, D.M. Shid, A.J. Lusis, C.E. Furlong, PON1 and organophosphate toxicity, PON1 in health and disease, L.G. Costa and C.E. Furlong Editors. Kluver, Norwell, (2002), 165-183.

Page 39: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

37

[18] T.B. Cole, B.J. Walter, D.M. Shih, A.D. Tward, A.J. Lusis, C. Timchalk, R.J. Richter, L.G. Costa, C.E. Furlong, Toxicity of chlorpyrifos and chlorpyrifos oxon in a transgenic mouse model of the human paraoxonase ( PON1) Q192R polymorphism, Pharmacogenet Genomics 15 (2005) 589–598.

[19] W.F. Li, L.G. Costa, R.J. Richter, T. Hagen, D.M. Shih, A. Tward, A.J. Lusis,C.E. Furlong, Catalytic effi ciency determines the in-vivo effi cacy of PON1 for detoxifying organophosphorus compounds, Pharmacogenetics 10 (2000), 767–779.

[20] T.B. Cole, R.L. Jampsa, B.J. Walter, T.L. Arndt, R.J. Richter, D.M. Shih, A. Tward, A.J. Lusis, R.M. Jack, L.G. Costa, C.E. Furlong, Expression of human paraoxonase ( PON1) during development, Pharmacogenetics 13 (2003), 357-364.

[21] B.N. La Du, Structural and functional diversity of paraoxonases, Nat Med 2 (1996), 1186-1187.[22] L.G. Costa, R.J. Richter, W.F. Li, T. Cole, M. Guizzetti, C.E. Furlong, Paraoxonase (PON 1) as a biomarker

of susceptibility for organophosphate toxicity, Biomarkers 8 (2003), 1-12. [23] O. Lockridge, P. Masson, Pesticides and susceptible populations: people with butyrylcholinesterase genetic

variants may be at risk, Neurotoxicology 21 (2000), 113-26.[24] S. Deakin, I. Leviev, V. Nicaud, M-C.B. Meynet, L. Tiret, R.W. James, Paraoxonase-1 L55M polymorphism

is associated with an abnormal oral glucose tolerance test and differentiates high risk coronary disease families, J Clin Endocr Metab87(2002), 1268-1273.

[25] C.E. Furlong, T.B. Cole, G.P. Jarvik, L.G. Costa, Pharmacogenomic considerations of the paraoxonase polymorphisms, Pharmacogenomics 3 (2002), 341-348.

[26] V.H. Brophy, M.D. Hastings, J.B. Clendenning, R.J. Richter, G.P. Jarvik, C.E. Furlong, Polymorphisms in the human paraoxonase ( PON1) promoter, Pharmacogenetics 11 (2001), 77-84.

[27] L.G. Costa, T.B. Cole, C.E. Furlong, Polymorphisms of paraoxonase ( PON1) and their signifi cance in clinical toxicology of organophosphates, J Toxicol Clin Toxicol 41 (2003), 37-45.

[28] L.G. Costa, T.B. Cole, A. Vitalone, C.E. Furlong, Measurement of paraoxonase ( PON1) status as a potential biomarker of susceptibility to organophosphate toxicity, Clin Chim Acta 352 (2005), 37-47.

[29] G.L. Ellman, K.D. Courtney, V. Andres, Jr., R.M. Featherstone, A new and rapid colorimetric determination of acetylcholinesterase activity, Biochem Pharmacol 7 (1961), 88-95.

[30] R.S. Naik, W. Liu, A. Saxena,Development and validation of a simple assay for the determination of cholinesterase activity in whole blood of laboratory animals,J Appl Toxicol 33(4) (2013), 290-300.

[31] S. Padilla, T.L. Lassiter, D. Hunter, In Methods in Molecular Medicine, Neurodegeneration Methods and Protocols. Eds J.Harry and H.A.Tilson, Humana Press Inc., N.J. Totowa, 22 (1999), 237-245.

[32] E. Reiner, A. Bosak, V. Simeon-Rudolf, Activity of cholinesterases in human whole blood measured with acetylthiocholine as substrate and ethopropazine as selective inhibitor of plasma butyrylcholinesterase, Arh Hig Rada Toksikol 55 (2004), 1-4.

[33] F. Worek, U. Mast, D. Kiderlen, C. Diepold, P. Eyer, Improved determination of acetylcholinesterase activity in human whole blood, Clin Chim Acta 288 (1999), 73–90.

[34] C. De Vriese, F. Gregoire, R. Lema-Kisoka, M. Waelbroeck, P. Robberecht, C. Delporte, Ghrelin degradation by serum and tissue homogenates: identifi cation of the cleavage sites, Endocrinology 145(11) (2004), 4997-5005.

[35] T.L. Huang, T. Shiotsuki, T. Uematsu, B. Borhan, Q.X. Li, B.D. Hammock, Structure–activity relationships for substrates and inhibitors of mammalian liver microsomal carboxylesterases, Pharm Res 13 (1996), 1495–1500.

[36] S.M. Chanda, S.R. Mortensen, V.C. Moser, S. Padilla, Tissue-specifi c effects of chlorpyrifos on carboxylesterase and cholinesterase activity in adult rats: An in vitro and in vivo comparison, Fundam ApplToxicol, 38 (1997), 148–157.

[37] B.J. Kitchen, C.J. Masters, D.J. Winzor, Effects of lipid removal on the molecular size and kinetic properties of bovine plasma arylesterase, BiochemJournal 135 (1973), 93-99.

[38] C.E. Furlong, R.J. Richter, S.L. Seidel, A.G. Motulsky, Role of genetic polymorphism of human plasma paraoxonase/arylesterase in hydrolysis of the insecticide metabolites chlorpyrifos oxon and paraoxon, AmJHumGenet43 (1988), 230-238.

[39] M.K. Johnson, Improved assay of neurotoxic esterase for screening organophosphates for delayed neurotoxicity potential, Arch Toxicol67 (1977), 113–115.

[40] E. Heymann, K. Krisch, Phosphoric acid-bis-(p-nitro-phenylester), a new inhibitor of microsomal carboxylesterases, Hoppe-Seyler’s Zeitschrift fur Physiologische Chemie, 348(6) (1967), 609-619.

[41] S.S. Anand, J.V. Bruckner, W.T. Haines, S. Muralidhara, J.W. Fisher, S. Padilla, Characterization of deltamethrin metabolism by rat plasma and liver microsomes, Toxicol and Applied Pharmacol 212 (2006) 156-166.

Page 40: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

38

[42] Y. Sakurai, S.F. Ma, H. Watanabe, N. Yamaotsu, S. Hirono, Y. Kurono, U. Kragh-Hansen, M. Otagiri, Esterase-like activity of serum albumin: characterization of its structural chemistry using p-nitrophenyl esters as substrates, Pharm Res 21 (2004), 285–292.

[43] O. Lockridge, W. Xue, A. Gaydess, H. Grigoryan, S.J. Ding, L.M. Schopfer, S.H. Hinrichs, P. Masson, Pseudo-esterase activity of human albumin: slow turnover on tyrosine 411 and stable acetylation of 82 residues including 59 lysines, J Biol Chem 283 (2008), 22582–22590.

[44] T. Imai, Human carboxylesterase isozymes: catalytic properties and rational drug design, Drug Metab Pharmacokinet 21 (2006), 173–185.

[45] E.S. Peeples, L.M. Schopfer, E.G. Duysen, R. Spaulding, T. Voelker, C.M. Thompson, O. Lockridge,Albumin, anewbiomarkeroforganophosphorustoxicantexposure, identifi edbymassspectrometry, ToxicolSci83 (2) (2005), 303-312.

[46] J.A. Crow, K.L. Herring, S. Xie, A. Borazjani, P.M. Potter, M.K. Ross, Inhibitionofcarboxylesteraseac-tivityofTHP1 monocytes/macrophagesandrecombinanthumancarboxylesterase 1 byoxysterolsandfattya-cids, BiochimBiophysActa1781 (2008), 643–654.

[47] T. Minagawa, Y. Kohno, T. Suwa, and A. Tsuji, Species differences in hydrolysis of isocarbacyclin methyl ester (TEI-9090) by blood esterases, Biochem Pharmacol 49 (10) (1995), 1361-1365.

[48] A.R. Alha, A. Ruohonen, M. Telaranta, Blood and brain cholinesterase activity in human death cases, in normal human subjects and in some laboratory and domestic animals, in: Medical Protection Against Chemical Warfare Agents, SIPRI, Almqvist &Wiksell International, Stockholm, Sweden, (1976), 151–156.

[49] K. Tuovinen, E. Kaliste-Korhonen, O. Hanninen, Gender differences in activities of mouse esterase and sensitivities to DFP and sarin toxicity, Gen Pharmacol 29 (1997), 333–335.

[50] L.M. Berry, L. Wollenberg, and Z. Zhao, Esterase activities in the blood, liver and intestine of several preclinical species and humans, Drug Metab Lett 3 (2) (2009), 70-77.

[51] L.V. Sigolaeva, G.F. Makhaeva, E.V. Rudakova, N.P. Boltneva, M.V. Porus, G.V. Dubacheva, A.V. Eremenko, I.N. Kurochkin and R.J. Richardson, Biosensor analysis of blood esterases for organophosphorus compounds exposure assessment: Approaches to simultaneous determination of several esterases, Chem-Biol Interact 187 (2010), 312–317.

[52] L.V. Sigolaeva, G.V. Dubacheva, M.V. Porus, A.V. Eremenko, E.V. Rudakova, G.F. Makhaeva, Rudy J. Richardson and I.N. Kurochkin, A layer-by-layer tyrosinase biosensor for assay of carboxylesterase and neuropathy target esterase activities in blood, Anal Methods 5(16) (2013), 3872-3879.

[53] M.K. Johnson, The target for initiation of delayed neurotoxicity by organophosphorus esters: Biochemical studies and toxicological applications, in: E. Hodgson, J.R. Bend, R.M. Philpot (Eds.), Reviews in Biochemical Toxicology, Elsevier, Amsterdam, 4 (1982) 141–212.

[54] R. Novak, S. Padilla, An in vitro comparison of rat and chicken brain neurotoxic esterase, Fundam Appl Toxicol 6 (1986), 464–471.

[55] K.R. Dyer, B.S. Jortner, L.G. Shell, M. Ehrich, Comparative dose-response studies of organophosphorus ester-induced delayed neuropathy in rats and hens administered mipafox, Neurotoxicology 13 (1992), 745–755.

[56] G.F. Makhaeva, I.V. Filonenko, V.V. Malygin, A comparative study of the interaction of phosphoric acid dichlorovinyl esters with a neurotoxic esterase from the brain of hens and rats, Zh. Evol. Biokhim.Fiziol.31 (1995), 396–403.

[57] S. Wieseler, L. Schopfer, O. Lockridge, Markers of organophosphate exposure in human serum, J Mol Neuroscien 30 (2006), 93–94.

Page 41: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

39

Chapter 3

Investigation of Mice Blood Neuropathy Target Esterase as Biochemical Marker of Exposure

to Neuropathic Organophosphorus Compounds

Elena V. RUDAKOVAa1, Larisa V. SIGOLAEVAb, Galina F. MAKHAEVAa

aInstitute of Physiologically Active Compounds Russian Academy of Sciences Chernogolovka, Moscow Region 142432 Russia

bDepartment of Chemistry, Moscow State University, Moscow 119992 Russia

Abstract. A hen is a standard animal model for studying organophosphate-induced delayed neurotoxicity (OPIDN). Recently we developed a mouse biochemical model for organophosphorus compounds (OPC) neuropathic potential assessment based on brain neuropathy target esterase ( NTE) and acetylcholinesterase (AChE) assay. The aim of the present study was to investigate the possibility of using the whole blood mice NTE as a biochemical marker for exposure to neuropathic OPCs and the possibility of OPIDN risk assessment in exposed species by comparison of blood NTE and AChE inhibition. Given that NTE and AChE inhibition in brain is a biomarker of OPIDN and acute cholinergic toxicity, respectively, we compared the NTE and AChE inhibition in whole blood and in brain of mice 1 h after a single i.p administration of increasing doses of two neuropathic OPCs possessing different acute toxicity: the toxic (C3H7O)2P(O)OCH=CCl2 (PrDChVP) and the low toxic (C4H9O)2P(O)OCH(CF3)2 (diBu-PFP). For measuring blood NTE activity a highly sensitive tyrosinase LBL biosensor was used. We showed that both OPCs inhibited AChE and NTE in mice blood and brain in dose-dependent manner. The corresponding values of ED50 were calculated. A strong correlation was found between inhibition of brain and blood NTE: r = 0.994 for PrDChVP and 0.997 for diBu-PFP, as well as between brain and blood AChE inhibition: r = 0.999 for PrDChVP and 0.969 for diBu-PFP. For both OPCs the ratio ED50(AChE)/ED50( NTE) in blood was shown to corresponds to that in brain, i.e., the ratio between NTE and AChE inhibition, measured in blood, may be used to characterize the probability of OPIDN development versus acute cholinergic toxicity. The results allow us to consider mice blood NTE as a biochemical marker of exposure to neuropathic OPCs.Inhibition of NTE in blood can be used in conjunction with inhibition of blood AChE to assess the likelihood that an exposure to OPC would produce cholinergic and/or delayed neuropathic effects.

Key words. Acetylcholinesterase, neuropathy target esterase, biomarkers, blood, organophosphorus compounds

1 Laboratory of Molecular Toxicology, Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, Moscow Region 142432, Russia, E-mail: [email protected]

Page 42: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

40

1. Introduction Organophosphorus compounds (OPC) covalently phosphylate serine hydrolases, thus

inhibiting their function. When this occurs with neuropathy target esterase (EC 3.1.1.5, NTE), followed by an aging process, a latent and distal axonal degeneration occurs, and this is described as organophosphate-induced delayed neuropathy (OPIDN). If an OPC is a substantially more potent inhibitor of acetylcholinesterase (EC 3.1.1.7, AChE) than of NTE, then cholinergic toxicity could result in lethality, thus obviating the development of OPIDN. However, if the OPC is neuropathic, cholinergic toxicity will be mild, and suffi cient NTE will be inhibited and aged to initiate OPIDN.

This syndrome involves distal sensorimotor degeneration of long large-diameter axons in spinal cord and peripheral nerves with associated sensory defi cits and paralysis occurring within 1–4 weeks after exposure [1]. Mechanistic research indicates that inhibition and aging of > 70% of NTE in neural tissue initiates OPIDN [2, 3] and that inactivation of AChE is not involved.

The adult hen is currently, the most widely accepted in vivo model for the study of OPIDN and assessment of the neuropathic hazard of OPCs. However, compared to the usual laboratory animal like rats or mice, hens are diffi cult to acquire and maintain for laboratory studies, and their substantially larger size requires considerably greater amounts of test materials for dosing in vivo.

Standard laboratory animals, mice and rats, have been thought to be resistant to OPIDN, because they do not readily display clinical signs of hind limb paralysis, despite exposure to high levels of neuropathic compounds [4, 5]. However, the more detailed investigations showed that both rats and mice are sensitive to OPC exposure although resistant to the ataxia: the structural damages in long axons, spinal cord and brain caused by OPC correlate with decreased NTE activity [6, 7, 8].

Recently we demonstrated a close sensitivity of NTE from mouse and hen brain in vitro and applicability of mouse brain NTE and AChE for biochemical assessment of OPC neuropathic potential both in vitro and in vivo. It was shown that inhibition of NTE and AChE in mouse brain can be used as a suitable predictor of neuropathic hazard of OPCs [9].

The aim of the present study was: 1) to investigate the possibility of using the whole blood mice NTE as a biochemical

marker for exposure to neuropathic OPCs and 2) to study the possibility of OPIDN risk assessment in exposed species by comparison

of blood NTE and AChE inhibition.Given that NTE and AChE inhibition in brain are biomarkers of OPIDN and acute

cholinergic toxicity [10, 11, 12], respectively, we compared the NTE and AChE inhibition in whole blood and brain of mice 1 hr after a single i.p administration of increasing doses of two neuropathic OPCs possessing different acute toxicity: a toxic O,O-di-1-propyl-O-2,2-dichlorvinyl phosphate, (C3H7O)2P(O)OCH=CCl2(PrDChVP), and low toxic O,O-dibutyl-O-(1-trifl uoromethyl-2,2,2-trifl uoroethyl) phosphate, (C4H9O)2P(O)OCH(CF3)2 (diBu-PFP).

PrDChVP is a known delayed neurotoxicant which was shown to induce ataxia in hens. It has rather high neuropathic potential: its relative inhibitor potencyRIP = IC50(AChE)/

Page 43: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

41

IC50( NTE) = 2.6 [13], and high acute toxicity: LD50 = 10 mg/kg, hens [14].PrDChVP was intensively studied in our previous experiments in vitro and in vivo in hens and rats [9, 15, 16, 17]. This OPC was used as a known neuropathic OPC to develop a mouse model for biochemical assessment of neuropathic potential [9]. Thus, in the current experiment, PrDChVP was used as a standard compound to investigate NTE and AChE inhibition in mice blood compared with brain enzymes.

The second used compound was a new model OPC diBu-PFP which possesses a high neuropathic potential RIP = 4.99 according to our in vitro experiments [18, 19] and low acute toxicity: LD50 for mice, i.p. was > 2000 mg/kg [9]. This compound was studied in our experiments on mice brain NTE and AChE inhibition in vitro and in vivo [9].

2. Materials and methods

ChemicalsPhenyl valerate (PV), N,N′-di-2-propylphosphorodiamidofl uoridate (mipafox,

MIP), and O,O-di-1-propyl O-2,2-dichlorovinyl phosphate (PrDChVP), O,O-dibutyl-O-(1-trifl uoromethyl-2,2,2-trifl uoroethyl) phosphate (diBu-PFP) were synthesized and characterized in the Institute of Physiologically Active Compounds, Russian Academy of Sciences (Russia) and kindly presented by Dr. Alexey Aksinenko. Synthesis of diBu-PFP is described in [18, 19]. The purity of all substances was > 99% (by spectral and chromatographic analysis data). Paraoxon (O,O-diethyl-4-nitrophenyl phosphate) was from Sigma Chemical Co (St. Louis, MO). Protein standard (BSA) was from Sigma Chemical Co. (St. Louis, MO, USA). The Coomassie protein kit was from Pierce Chemical Co. (Rockford, IL, USA). All other chemicals were analytical grade and used without further purifi cation. Aqueous solutions were prepared using deionized water.

IC50 Determination for inhibition of brain and blood NTE and AChE in mouse brain and blood samples.

Mouse brain and blood sampling and preparation for esterases analysis, as well as the methods of AChE and NTE assay are described below. The pooled samples of blood and brain of mice were used. The IC50 values for OP inhibitors against brain and blood NTE and AChE were determined by 20 min preincubation of mice blood and brain samples with 10 to 12 different concentrations of either PrDChVP (from 10-11 to 10-5M) or diBu-PFP (from 10-11 to 10-3 M) in working buffer. Residual enzymes activity was then determined. Each measurement was made in triplicate (spectrophotometry) or in duplicate (amperometry for blood NTE).

IC50 values were calculated using Origin 6.1 software, OriginLab Corp. (Northampton, MA). Every value represents the mean ±SEM from tree or two independent experiments.

Animal experiments and tissues preparationIn vivo inhibition of NTE and AChE in mouse brain and blood

In vivo experiments were carried out on outbred male white miceCD1 (18-25 g). All experiments and procedures with animals were carried out according to the protocols approved by the Ethics Committee of the Institute of Physiologically Active Compounds RAS (Chernogolovka, Russia).

Page 44: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

42

PrDChVP and diBu-PFP were dissolved in DMSO and injected once, i.p. in a volume about 0.1 ml in increasing doses: 0.3, 0.75, 1.5, 3, 6, 12, 24, 36 mg/kg for PrDChVP and 0.5, 1, 2.5, 15, 30, 100, 250, 1000, 2000 mg/kg for diBu-PFP. At least 6 animalswere used per each dose. 20 min before PrDChVP injection, mice were pretreated with atropine sulfate, 20 mg/kg in water, i.p.; control animals received atropine sulfate and DMSO. In the experiment with diBu-PFP control animals were administered only with DMSO. After 1 h, the mice were decapitated under CO2 anesthesia. Trunk blood from each animal was collected immediately in glass vials containing 3.8% w/v sodium citrate (0.2 ml citrate/ml blood). All blood samples were aliquoted, frozen in liquid nitrogen, and stored at -70 oC until further use. Brains were immediately removed, weighed, and frozen in liquid nitrogen and stored at -70 °C until use.

NTE and AChE activities in brain and blood of mice treated with the OPCs were determined and compared to activity in tissue samples from control animals treated with DMSO or DMSO + atropine.

Analysis of dose - response was performed using Origin 6.1 software, OriginLab Corp. (Northampton, MA) to yield ED50 values.

Preparation of blood and brain for analysis of NTE and AChE activityThe blood samples were thawed at4°C and diluted 1:100 (v:v) in cold 0.1 M sodium

phosphate pH 7.5 for preparing blood hemolysates. After careful stirring for 2 minutes the hemolysed blood samples were aliquoted into plastic tubes Falcon, immediately frozen in liquid nitrogen to ensure complete hemolysis and stored at -20 єC until use. Before analysis, the samples were thawed slowly in ice water bath.

The thawed brain samples were homogenized at 4°C in 5 volumes of buffer (50 mM Tris-HCl, 0.2 mM EDTA, pH 8.0) with a Potter homogenizer. The homogenates were centrifuged for 15 min at 9000 ×g at 4°C to prepare the 9S supernatant used for enzyme assay [7]. Aliquots of the supernatants (brain 9S fraction) were stored at −70 °C until use. The concentration of protein in the mouse brain 9S homogenates was5.9 -7.6 mg/ml.

AChE assay AChE activity in brain and whole blood was determined with 1 mM of acetylthiocholine

iodide as a substrate for the spectrophotometric method of Ellman [20] using a Gilford-250 spectrophotometer in 3 ml 1 cm pathlength cuvettes at 25 °C. All data were automatically corrected for spontaneous hydrolysis of each substrate measured at the same time in the reference cuvette. For AChE assays in blood the standard for Ellman assay wavelength (412 nm) was changed to 436 nm (ε436 = 10600 M-1cm-1) [21] to reduce interference from the high absorbance of hemoglobin at 412 nm.

Brain NTE assay Brain NTE activity was assayed colorimetrically according to the differential

inhibition method of Johnson [22] with slight modifi cations. NTE activity in 9,000 ×g (9S) supernatants of whole brain homogenate [7] was determined as the difference in PV hydrolase activity between paired samples preincubated for 20 min with either paraoxon (50 μM) or paraoxon (50 μM) plus mipafox (250 μM).

Page 45: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

43

Briefl y, samples were incubated at 37 °C in 50 mM Tris-HCl/0.2 mM EDTA (pH 8.0 at 25 °C) with 50 μM paraoxon (sample B) or 50 μM paraoxon plus 250 μM mipafox for 20 min (sample C), followed by incubation with PV (fi nal apparent concentration 0.54 mM) for the next 40 min at 37°C. NTE activity was calculated as the difference in the amount of phenol released between samples B and C.

Blood NTE assay

NTE activity was assayed according to the differential inhibition method of Johnson [22] with an electrochemical endpoint as described previously [16, 23]. A new sensitive, stable and reproducible planar tyrosinase biosensor was used for phenol detection. This biosensor was developed through a combination of screen-printing technology for conducting graphite support preparation and LBL technology of polyelectrolytes/oxidoreductase deposition[24].

Briefl y, 1:100 (v:v) blood hemolysate samples were incubated at 37 °C with 50 μM paraoxon (sample B) or 50 μM paraoxon plus 250 μM mipafox for 20 min (sample C). Phenyl valerate substrate (fi nal apparent concentration 0.54 mM) was added and the incubation was continued for the next 40 min at 37 °C. The reaction was stopped by addition of 100 μl of 1% (w/v) aqueous SDS. Total volume of reaction mixture was 600 μl. The released phenol was assayed amperometrically after 20- to 50-fold dilution of samples in 50 mM sodium phosphate with 100 mM NaCl, pH 7.0. The analytical signal was determined as the value of steady-state baseline current change (the difference between an average value of steady-state baseline current before and after analyte addition). Activity values were calculated using phenol calibration curves, obtained under the same conditions, and each was corrected for spontaneous hydrolysis of substrates, determined separately.

Acute toxicity determination The acute toxicity of the tested compounds was determined in outbred male white

mice weighing 18-25 g which received i.p. injections of OPCs. The observation period was 24h. The LD50 values were calculated by probit analysis using the BioStat 2006 program.

StatisticsStatistical analysis wascarried out using GraphPad Prism version 3.02 for Windows,

GraphPad Software (San Diego, CA). The results are given as means ±SEM. The level of signifi cance was set at p<0.05.

3. Results and Discussion

In vitro inhibitor activity of PrDChVP and diBu-PFP against AChE and NTE in brain and blood preparations.

First of all we assessed the inhibitory activities of PrDChVP and diBu-PFPagainst AChE and NTE in the experiments in vitro using brain and blood preparations (fi xed-time IC50 values for 20 min incubation of the enzymes with the inhibitors). The results are shown in Table 1.

Page 46: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

44

Table 1. The fi xed-time IC50 values for OP inhibitors against AChE and NTE in mice brain and blood preparations (for 20 min of incubation).

mice brain (9S supernatant)IC50, M

mice blood,IC50, M

Inhibitors AChE NTE AChE NTE

PrDChVP (9.43±0.64) ×10-8 (2.44±0.12) ×10-8 (1.23±0.04) Ч10-7 (2.54±0.25) ×10-8

diBu-PFP (4.93±0.20) ×10-7 (6.75±0.41) ×10-8 (1.56±0.12) Ч10-6 (2.28 ± 0.17) ×10-7

As can be seen from Table 1, PrDChVP is the more potent inhibitor of AChE and NTE from both sources thandiBu-PFP. PrDChVPexhibits similar inhibitor activity against brain and blood esterases and inhibits NTE to a greater extent than AChE.Quite close inhibitor activity against brain and blood esterases was observed for diBu-PFP, although both the blood enzymes were inhibited slightly less effective than brain ones. Both in brain and in blood diBu-PFP inhibited NTE more effective than AChE.

We calculated the values of the relative inhibitor potency RIP= IC50(AChE)/IC50( NTE) for both inhibitors in mice brain and blood. This ratio characterizes the ability for an OP compound to produce OPIDN in opposite to its acute toxicity. If the RIP is less than one, the compound will cause a lethal cholinergic toxicity, and cannot produce OPIDN at doses less than the LD50; however, if the RIP is greater than one, then the compound has the potential to cause OPIDN in doses less LD50 [11, 12, 13, 25, 26].The data are shown in Table 2.

Table 2. The values of RIP for PrDChVP and diBu-PFP calculated for mice brain and blood preparations

mice brain mice blood

Inhibitors IC50(AChE)/IC50( NTE) IC50(AChE)/IC50( NTE)

PrDChVP 3.9 4.8

diBu-PFP 7.3 6.8

Inspection of Table 2 reveals that the ratios IC50(AChE)/IC50(NTE) for both OPCs in mice blood are close for those in mice brain and are rather higher than 1, which signifi es that both compounds are potential delayed neurotoxicants, especially diBu-PFP. These results are in good agreement with the data obtained using hen brain enzymes for PrDChVP, RIP = 2.6 [3], and with our in vitro data for diBu-PFP, RIP = 4.99 [18].

Thus, the results of in vitro study showed that inhibition of blood enzymes adequately refl ects the inhibitory effects of the OPCs against brain enzymes.

In vivo inhibitor activity of PrDChVP and diBu-PFP against brain and blood AChE and NTE

To investigate the possibility of using mice whole bloodNTEas a biochemical marker of exposure to neuropathic OPC as well as the possibility of OPIDN risk assessment in exposed species by comparison of blood NTE and AChE inhibition, inhibition of NTE

Page 47: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

45

and AChE in mice blood was studied 1 h after i.p. administration of increasing doses of PrDChVP and diBu-PFP compared with brain enzymes inhibition.

We determined LD50 value forPrDChVP in mice, i.p., it was 15 (13.4ч17.3) mg/kg. That corresponds well to its acute toxicity on hens - 10 mg/kg [14].

It was shown that both compounds inhibited AChE and NTE in mice brain and blood in dose-dependent manner. Figure 1 demonstrates the dose-response curves of inhibition of NTE and AChE in brain and blood of mice by PrDChVP (A, B) and diBu-PFP (C, D).

C) D)

Figure 1. Dose-related NTE and AChE inhibition in mice brain and blood given PrDChVP (A, B) and diBu-PFP (C, D) in 1h after i.p. OPC administration.The results are as % control value

for each tissue expressed as mean ± SEM. Esterase activities of the control animals: in mice brain, nmol/(minЧmg protein): AChE = 69.2±3.5 (N=20), NTE = 13.4±0.52 (N=10);

in mice blood, nmol/(minЧml blood): AChE = 829±35(N=18), NTE =18±2 (N=8)

As shown in Figure 1, inhibition of NTE and AChE in blood and brain by both OPCs was clearly dose-dependent. Figure 1 demonstrates also that both OPCs inhibit NTE and AChE in mice blood to a somewhat greater degree as compared to the enzymes in brain.

In order to use the measurement of blood NTE and AChE activity as a mirror of brain enzymes, the correlation between the inhibition of these enzymes in blood and brain should be known.

Data replotted and analyzed from Figure 1 (A, B) show a strong correlations between

1 10

0

20

40

60

80

100 brain NTE blood NTE

inhi

bitio

n of

NTE

, %

Dose PrDChLVP, mg/kg1 10

0

20

40

60

80

100 brain AChE blood AChE

inhi

bitio

n of

AC

hE, %

Dose PrDChLVP, mg/kg

A) B)

1 10 100 1000

0

20

40

60

80

100

inhi

bitio

n of

NTE

, %

Dose diBu-PFP, mg/kg

brain NTE blood NTE

1 10 100 1000

0

20

40

60

80

100

inhi

bitio

n of

ACh

E, %

Dose diBu-PFP, mg/kg

brain AChE blood AChE

Page 48: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

46

inhibition of NTE by PrDChVP in brain and whole blood (r = 0.996, p < 0.0001, n = 6) as well as between AChE inhibition by PrDChVP in brain and blood (r = 0.999, p < 0.0001, n = 6). Results are presented in Figure 2.

А) B)

Figure 2. Correlations between NTE inhibition in brain and blood (A) and between AChE inhibition in brain and blood (B) in mice dosed with PrDChVP

Data replotted and analyzed from Figure 1(C, D) show a strong correlations between inhibition of NTE by diBu-PFP in brain and blood (r = 0.967, p = 0.00035, n = 7) as well as between AChE inhibition by diBu-PFP in brain and blood (r = 0.966, p = 0.0074, n = 5). The results are presented in Figure 3.

C) D)

Figure 3. Correlations between NTE inhibition in brain and blood (C) and between AChE inhibition in brain and blood (D) in mice dosed with diBu-PFP

Thus, by directly comparison of brain and blood enzymes inhibition by PrDChVP and diBu-PFPin vivo a strong correlation was established between both tissues. This fact allows us to propose mice blood NTE as an accessible biomarker of exposure to neuropathic OPC.

To characterize the neuropathic hazard of OP compounds, an index based on the in vivo susceptibility of the relevant targets, NTE and AChE, to OP inhibitors in susceptible species

0 20 40 60 80 100

0

20

40

60

80

100

r = 0.996p < 0.0001N=6br

ain

NTE

inhi

bitio

n, %

blood NTE inhibition, %0 20 40 60 80 100

0

20

40

60

80

100

r = 0.999p < 0.0001N=6br

ain

AChE

inhi

bitio

n, %

blood AChE inhibition, %

50 60 70 80 90 100

0

20

40

60

80

100

brai

n N

TE in

hibi

tion,

%

r = 0.967p = 0.00035N=7

blood NTE inhibition, %40 50 60 70 80 90 100

0

20

40

60

80

100

r = 0.966p = 0.0074N=5bl

ood

AChE

inhi

bitio

n, %

blood AChE inhibition, %

Page 49: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

47

has been proposed [11,12]. This relative measure is a ratio ED50(AChE)/ED50(NTE) where ED50 is median effective dose, mg/kg. As this value increases, the potential neuropathic hazard of the OP compound increases.

We demonstrated that ratio ED50(AChE)/ED50(NTE) was very close for mice and hen brain after dosing animals with the known neuropathic OPC PrDChVP [9]. Similar effect was obtained earlier when the increasing doses of PrDChVP were administrated to hens and rats [15]. So, inhibition of NTE and AChE in mouse brain after exposure to OPC can be used as a suitable predictor of neuropathic hazard of OP compounds, i.e., their ability to produce OPIDN [9].

By analyzing the dose-response curves (Fig.1) the ED50 values for inhibition of NTE and AChE in mice brain and blood by PrDChVP and diBu-PFP were obtained. The values of in vivo inhibitor selectivity were calculated as the ratios ED50(AChE)/ED50(NTE ). The results are shown in Table 3.

Table 3. Inhibitory effects (ED50, mg/kg)of PrDChVP and diBu-PFP against AChE and NTE in mice brain and blood 1 h after i.p. OPC administration and OPC selectivity with respect to NTE as compared to AChE (the ratio ED50(AChE)/ED50(NTE ))

ED50, mg/kgmice brain

ED50, mg/kgmice blood

ED50(AChE)/ED50( NTE)

Compounds AChE NTE AChE NTE mice brain mice blood

PrDChVP 4.3±0.6 2.2±0.4 4.0±0.2 2.0±0.1 2 2

diBu-PFP 516±41 127±8 154±5 36.3±3.6 4.1 4.3

As can be seen from Table 3, PrDChVP inhibits brain and blood enzymes signifi cantly stronger than diBu-PFPwhich fully corresponds to the results of experiment in vitro.High inhibitor activity of PrDChVP against AChE in vivo: ED50 = 4.3 mg/kg inmice brain and 4 mg/kg in mice blood, corresponds to its high acute cholinergic toxicity (LD50 = 15 mg/kg). This compound is also a strong NTE inhibitor in vivo: ED50 = 2.2 mg/kg in brain and 2 mg/kg in blood.

The valueof the ratio ED50(AChE)/ED50(NTE) indicates the predominance of one of toxic effects on another. For PrDChVP the ratio ED50(AChE)/ED50(NTE) = 2, so, the possibility of development of delayed neurotoxicity prevails over the acute cholinergic toxicity. Thus, the intoxication with this compound leads both to high acute cholinergic and delayed neurotoxicity. What is important,the values of the ratio ED50(AChE)/ED50(NTE) are identical for blood and brain and correspond to the results of experiment in vitro (Table 2).

Low inhibitor activity of diBu-PFP against brain AChE in vivo, ED50 = 516 mg/kg, corresponds to its low acute toxicity: LD50> 2000 mg/kg. Brain NTE is inhibited by diBu-PFP at much less doses: ED50 = 127 mg/kg. The ratio ED50(AChE)/ED50(NTE) equal 4.3 indicates to high risk of OPIDN development after exposure to diBu-PFP, that agrees with the data in vitro (Table 2). So, a low toxic diBu-PFP might initiate OPIDN in doses which do not produce warning signs of acute cholinergic poisoning.

Page 50: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

48

As can be seen from Table 3, diBu-PFP inhibits NTE and AChE in blood to a greater degree as compared to enzymes in brain, i.e. blood esterases are more sensitive to inhibition with diBu-PFP than brain esterases. This difference may be caused by features of toxicokinetics of this less reactive compound. Nevertheless, inhibition of both enzymes in the blood correlates well with the enzyme inhibition in the brain (Fig. 3) and, importantly, the ratio ED50(AChE)/ED50(NTE), which characterizes the risk of delayed neurotoxicity compared to acute one, is very close for brain and blood enzymes: 4.1 and 4.3, respectively.

Thus, for both OPCs the ratio ED50(AChE)/ED50(NTE) in blood corresponds to that in brain, i.e., the ratio between NTE and AChE inhibition, measured in blood, characterizes the probability of OPIDN development versus acute cholinergic toxicity.

A) B)

Figure 4. Inhibition of NTE and AChE activities in mice blood 1 h after i.p. administration of increasing doses of PrDChVP – (A) and diBu-PFP - (B), mg/kg. Data are presented as % inhibition

of the corresponding esterase in the control animals. Esterase activities in the control animals are shown in the legend to Figure 1

Figure 4 demonstrates the dose-response curves for inhibition of NTE and AChE in mice blood 1 h after exposure to the two neuropathic OPCs possessing different acute toxicity: high toxic PrDChVP and low toxic diBu-PFP.

This fi gure clearly shows that blood AChE is inhibited by PrDChVP (A) and diBu-PFP (B) in doses corresponding to their acute toxicity. NTE in blood is inhibited by PrDChVP and diBu-PFP to a greater degree as compared to AChE, that fully corresponds to their ED50(AChE)/ED50(NTE) in brain and indicates to potential neuropathic hazard for both OPCs. And for highly toxic PrDChVP delayed neurotoxicity may develop after successful treatment of acute poisoning. Whereas after diBu-PFP intoxication, OPIDN can be developed without warning signs of acute cholinergic poisoning.

In conclusion, the data presented and discussed above allow us to consider mice blood NTE as a biochemical marker of exposure to neuropathic OPCs.Furthermore, these results indicate that mice blood NTE and AChE inhibition refl ects NTE and AChE inhibition in mice brain. Inhibition of NTE in blood can be used in conjunction with inhibition of blood AChE to assess the likelihood that an exposure to OPC would produce cholinergic and/or delayed neuropathic effects.

This work is supported by NATO Science for Peace and Security Program (grant no SfP 984082). We are grateful to Dr. A.Yu. Aksinenko for the synthesis of compounds for

1 10

0

20

40

60

80

100

blood NTEblood AChE

inhi

bitio

n of

NTE

and

AC

hE, %

Dose PrDChLVP, mg/kg1 10 100 1000

0

20

40

60

80

100

blood NTE

blood AChE

inhi

bitio

n of

NTE

and

AC

hE, %

Dose diBu-PFP, mg/kg

Page 51: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

49

research, as well Tatyana Galenko and Olga Serebryakova for their assistance with the experiments on mice.

References

[1] R.J. Richardson, Organophosphate Poisoning, Delayed Neurotoxicity. In: Encyclopedia of Toxicology, P. Wexler Editor. Second ed. Oxford, Elsevier, 3 (4 vols.), 2005, 302-306.

[2] M. K. Johnson, The target for initiation of delayed neurotoxicity by organophosphorus esters: Biochemical studies and toxicological applications. In: Reviews in biochemical toxicology, eds. E. Hodgson, J. R. Bend, and R. M. Philpot, Amsterdam, Elsevier, vol. 4, 1982, 141–212.

[3] M. Lotti, The pathogenesis of organophosphate polyneuropathy, Crit Rev Toxicol 21 (1992), 465-487.[4] M.B. Abou-Donia, Organophosphorus ester-induced delayed neurotoxicity, Annu Rev Pharmacol Toxicol21

(1981), 511-548.[5] B. Veronesi, S. Padilla, K. Blackmon, C. Pope, A murine model of OPIDN: Neuropathic and biochemical

description, Toxicol Appl Pharmacol107 (1991), 311-324.[6] B. Veronesi, A rodent model of organophosphorus-induced delayed neuropathy: distribution of central

(spinal cord) and peripheral nerve damage, Neuropath Appl Neurobiol 10 (1984), 357-368. [7] S. Padilla, B. Veronesi, The relationships between neurological damage and neurotoxic esterase inhibition

in rats acutely exposed to tri-ortho-cresyl phosphate, Toxicol Appl Pharmacol 78 (1985), 78–87.[8] E. Mutch, S.S. Kelly, P.G. Blain, F.M. Williams, Comparative studies of two organophosphorus compounds

in the mouse, Toxicol Lett 81(1) (1995), 45-53.[9] E.V. Rudakova, O.G. Serebryakova, N.P. Boltneva, T.G. Galenko, G.F. Makhaeva, A biochemical model

in mice for assessment of neuropathic potential of organophosphorus compounds, Toxicological Reviews (Russian) 6 (2012), 20-24.

[10] L.G. Costa, Biomarker research in neurotoxicology: The role of mechanistic studies to bridge the gap between laboratory and epidemiological investigations, Environ Health Perspect 104 (1996), (Suppl.1), 55-67.

[11] R.J. Richardson, Interactions of organophosphorus compounds with neurotoxic esterase, Organophosphates: Chemistry, Fate, and Effects, J.E. Chambers and P.E. Levi Editors. Academic Press, San Diego, (1992), 299-323.

[12] V.V. Malygin, V.B. Sokolov, R.J. Richardson, and G.F. Makhaeva, Quantitative structure-activity relationships predict the delayed neurotoxicity potential of a series of O-alkyl-O-methylchloroformino phenylphosphonates, J Toxicol Environ Health Part A, 66 (2003), 611-625.

[13] M. Lotti, M.K. Johnson, Neurotoxicity of organophosphorus pesticides: Predictions can be based on in vitro studies with hen and human enzymes, Arch Toxicol 41 (1978), 215-221.

[14] J.R. Albert, S.M. Stearns, Delayed neurotoxic potential of a series of alkyl esters of 2, 2-dichlorovinyl phosphoric acid in the chicken, Toxicol Appl Pharmacol 29(1974), 136.

[15] G.F. Makhaeva, I.V. Filonenko, V.V. Malygin, A comparative study of the interaction of phosphoric acid dichlorovinyl esters with a neurotoxic esterase from the brain of hens and rats, Zh Evol Biokhim Fiziol 4 (1995), 396-403. [Article in Russian]

[16] G.F. Makhaeva, L.V. Sigolaeva, L.V. Zhuravleva, A.V. Eremenko, I.N. Kurochkin, V.V. Malygin, and R.J. Richardson, Biosensor detection of Neuropathy Target Esterase in whole blood as a biomarker of exposure to neuropathic organophosphorus compounds, J Toxicol Environ Health Part A66 (2003), 599-610.

[17] G.F. Makhaeva, V.V. Malygin, N.N. Strakhova, L.V. Sigolaeva, L.G. Sokolovskaya, A.V. Eremenko, I.N. Kurochkin and R.J. Richardson, Biosensor assay of neuropathy target esterase in whole blood as a new approach to OPIDN risk assessment: review of progress, Hum Exp Toxicol 26 (2007), 273-282.

[18] G.F. Makhaeva, O.G. Serebryakova, N.P. Boltneva, T.G. Galenko, A.Yu. Aksinenko, V.B. Sokolov, I.V. Martynov, Esterase profi le and analysis of structure – inhibitor selectivity relationships for homologous phosphorylated 1-hydroperfl uoroisopropanoles, Doklady Biochem Biophys 423 (2008), 352-357. [translated from Russian Dokl Akad Nauk 423 (6) 2008, 826-831].

[19] G.F. Makhaeva, A.Y.Aksinenko, V.B.Sokolov, O.G. Serebryakova, R.J. Richardson, Synthesis of organophosphates with fl uorine-containing leaving groups as serine esterase inhibitors with potential for Alzheimer disease therapeutics, Bioorg Med Chem Lett 19 (2009), 5528-5530.

[20] G.L. Ellman, K.D. Courtney, V. Andres, Jr., R.M. Featherstone, A new and rapid colorimetric determination of acetylcholinesterase activity, Biochem Pharmacol 7 (1961), 88-95.

Page 52: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

50

[21] F. Worek, U. Mast, D. Kiderlen, C. Diepold, P. Eyer, Improved determination of acetylcholinesterase activity in human whole blood, Clin Chim Acta 288 (1999) 73-90.

[22] M. K. Johnson, Improved assay of neurotoxic esterase for screening organophosphates for delayed neurotoxicity potential, Arch Toxicol67, (1977), 113–115.

[23] L.V. Sigolaeva, A. Makower, A.V. Eremenko, G.F. Makhaeva, V.V. Malygin, I.N. Kurochkin, F. Scheller, Bioelectrochemical analysis of neuropathy target esterase activity in blood, Anal Biochem 290 (2001), 1-9.

[24] L.V. Sigolaeva, D.V. Pergushov, C.V. Synatschke, A. Wolf, I. Dewald, I.N. Kurochkin, A. Feryc, A.H.E. MЁuller, Co-assemblies of micelle-forming diblock copolymers and enzymes on graphite substrate for an improved design of biosensor systems, Soft Matter 9 (2013), 2858–2868.

[25] R.J. Richardson, T.B. Moore, U.S. Kayyali, J.H. Fowke, J.C. Randall, Inhibition of hen brain acetylcholinesterase and neurotoxic esterase by chlorpyrifos in vivo and kinetics of inhibition by chlorpyrifos oxon in vitro: application to assessment of neuropathic risk,Fundam Appl Toxicol 20 (1993), 273-279.

[26] S.J. Wijeyesakere, R.J. Richardson, Neuropathy target esterase. In: Hayes’ Handbook of Pesticide Toxicology. 3nd ed. Elsevier, 2010, 1435-1455.

Page 53: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

51

Chapter 4

Layer-by-Layer Electrochemical Biosensors for Blood EsterasesAssay

Ilya N. KUROCHKINa, Larisa V. SIGOLAEVAa, Arkadiy V. EREMENKOa,b, Ekaterina A DONTSOVAa, Maria S. GROMOVAa, Elena V. RUDAKOVAc,

Galina F. MAKHAEVAc

aM.V. Lomonosov Moscow State University, Chemical Faculty, Leninskie Gory 1/3, Moscow, 119992 RUSSIA

bN.M. Emanuel Institute of Biochemical Physics RAS, Kosygina str. 4, Moscow, 119334 RUSSIA

cInstitute of Physiologically Active Compounds RAS, Chernogolovka, Moscow Region,142432 RUSSIA

Abstract. The effi ciency of enzyme-polyelectrolyte nanofi lms, deposited on carbon screen-printed electrodes (SPE) as a highly-sensitive transducer element of biosensing platforms fabricated with “ layer-by-layer” technology was demonstrated. The present report describes several analytical possibilities of the biosensing platform based on choline oxidase (ChO) and tyrosinase nanocomposites for blood esterase assay.

Keywords. Enzyme-polyelectrolytes nanofi lms, screen printed electrodes, esterase assay.

IntroductionActivity of the mainbloodesterases, such as theAChE,BChE, CaE and NTEcan be

determinedby measuring thedegree of hydrolysis ofcholineand phenolcontainingsubstrates, respectively. Enzyme based amperometric biosensors are traditionally considered for a reliable, rapid, compact and sensitive detection of choline and phenol in aqueous medium. The biosensor detection of choline is based on its biocatalytic oxidation by choline oxidase, immobilized on the electrode surface generating hydrogen peroxide. The following registration of hydrogen peroxide can be achieved by hydrogen peroxide sensing electrodes [1].The biosensor detection of phenol is based on oxidation of phenol via catechol into o-quinone by the tyrosinase. Then o-quinone is electrochemically reduced to catechol directly at the electrode when the required potential is applied [2].

Choline oxidase and tyrosinase with high effi ciency can be incorporated into enzyme-polyelectrolyte sensor fi lms based on self-assemblingtechnologyofpolyelectrolytesdeposition or layer-by-layer technique ( LBL)[3]. Such fi lms deposited onto a conductive support can provide sensitive detection of choline or phenol [4,5]. Screen-printing by

Page 54: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

52

graphite pastes or graphite-based composites represents the most commonly encountered modern way of sensor fabrication due to its cost-effectiveness, manufacturability and electrochemical advantages.

The present report describes biosensor design and analytical possibilities of the biosensing platform based on choline oxidase- and tyrosinase-polyelectrolytes nanofi lms fabricated with “ layer-by-layer” technology and deposited on carbon screen-printed electrodes (SPE). Possible application ofthe biosensingplatformdevelopedto determine the activityof blood esterases is also described.

1. Biosensor design and analytical possibilities

1.1. MnO2hydrosols and a hydrogen peroxide sensitive layer.As indicated above, the biosensor detection of choline is based on its biocatalytic

oxidation by choline oxidase, immobilized on the electrode surface generating hydrogen peroxide. The following registration of hydrogen peroxide can be achieved by mediated layer containing electrodes. Mediated layers based onmanganese dioxidegivesome of the bestfeatures. It has been shown that MnO2 nanoparticles have special physical and chemical properties, different from common MnO2 powders providing signifi cant improvement of mediating properties on the one hand, and simplifying for the stable water mixtures on the other hand. Thus, synthesis of a stable water suspension from MnO2 nanoparticles is the essential step in the development of technology for highly sensitive biosensor production. We have studiedseveral types of MnO2nanoparticles [1,6].

Figure 1. SEM images for different crystalline modifi cations of MnO2 nanoparticles. a) amorphous MnO2, b) beta-MnO2 with average rod diameter – 100 nm, c) beta-MnO2 with average rod diameter – 50 nm, d) beta-MnO2 with average rod diameter – 25 nm, e) beta-MnO2 with average rod diameter – 15 nm, f) gamma-MnO2.

SEM images of the manganese dioxide hydrosols dried on HOPG surface are presented in Figure 1. Amorphous MnO2 (Figure 1a) represented particles close to spherical in shape,

Page 55: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

53

200-300 nm in diameter while beta-MnO2 (Figure 1b, 1c, 1d, 1e) were different from the formed one by the presence of nano-size fi lamentous crystals of varying from 100 nm (b) to 50 nm (c) and 25 nm (d) and 15 nm (e) in diameter respectively. Figure 1f presents SEM photograph of gamma-MnO2 with the characteristic “wrinkled” structures on the surface of HOPG. The crystal phase of the MnO2 was analyzed by powder X-ray diffraction[1,6].

Possibilities of SPEs covered with different crystalline modifi cations of manganese dioxide for hydrogen peroxide detection have been studied.Screen-printed carbon electrodes (SPE) were made using semi-automated machine Winon (model WSC-160B, China) with 200 mesh screen stencil. Polyvinyl chloride substrate of 0.2 mm thickness and conductive graphite paste (Coates Screen, Germany) were used. Each SPE consisted of a round-shaped working area (3 mm diameter), a conductive track (30 mm Ч 1.5 mm), and a square extremity (3 mm Ч 7 mm) for the electrical contact. Peroxide-sensitive layer was formed by dropping 5 mkl of appropriate MnO2 sol solution on the working area of the electrode followed by drying at a room temperature for 40 min. Then the electrode was rinsed with bidistilled water and dried at the temperature of 60 ◦C for 1 h.

Voltammograms obtained in the presence of hydrogen peroxide showed higher levels of oxidation current in comparison to similar data obtained in Hepes buffer for all of the samples. Oxidation of hydrogen peroxide leads to reduction of MnO2 and formation of Mn (II) and (III) oxides and subsequent oxidation of these oxides with reiterated generation of MnO2 on the electrode surface. Amperometric responses to 10mkM H2O2 were not changed at working potential range from 250 mV to 500 mV.

Comparison of amperometric responses for electrodes coated with different MnO2 nanoparticles is given in Table 1.

Table 1. Amperometric responses to 100 nM hydrogen peroxide for electrodes based on different crystalline modifi cations of MnO2.

Type of MnO2 nanoparticles Electrode response, nAAmorphous 100 + 20

Beta-phase, rod diameter 100 nm 110 + 22Beta-phase, rod diameter 50 nm 115 + 23Beta-phase, rod diameter 25 nm 175 + 35Beta-phase, rod diameter 15 nm 210 + 42

Gamma-phase 233 + 36

These data showthat gamma-MnO2 based electrodes aremore sensitiveat the detection ofhydrogen peroxidethan theelectrodespreparedusing othercrystalline modifi cations.

The calibration curves for hydrogen peroxide were obtained at working potential of 250 mV (A) and 480 mV (B). In both cases, the electrode response increased linearly over the entire range of examined H2O2 concentrations.The detection limit at 250 mV calculated according to the equation: y=23.6⋅x-0.3 (signal to noise ratio = 0.3 nA), was 4.5⋅10-8M (3σ), the linear range was 4.5⋅10-8-1.0⋅10-4 M, and the sensitivity was estimated as 377±1 mA·M-1⋅cm-2.Similar results were obtained for potential 480 mV with hydrogen peroxide linear range 2.2⋅10-8-1.0⋅10-4 M. The detection limit calculated for hydrogen peroxide was 2.2⋅10-8 M (3σ) (the equation was y=36.1⋅x+0.1 at signal to noise ratio = 0.3 nA), and sensitivity was 515±3 mA·M-1⋅cm-2.

Page 56: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

54

Operational stability of peroxide-sensitive electrodes was investigated at potentials 250 mV and 480 mV, in Hepes buffer (pH=7.5). Ten measurements were performed for each electrode for 10-5 M H2O2 with R.S.D. 5.5±1.7% for 250 mV and 5.0±3.0% for 480 mV. Storage of the MnO2-modifi ed electrodes at room temperature for two months did not lead to any changes in analytical characteristics of the tested sensors.

1.2. Choline oxidase screen-printed amperometric sensors based on the layer-by-layer technology.

Choline oxidase biosensors based on MnO2-modifi ed SPE were manufactured according to the following procedure. Choline oxidase (ChOx) was dissolved in 50 mM Hepes buffer, containing 30 mM KCl (pH 7.5). Polyelectrolytes (poly(dimethyldiallylammonium chloride) (PDDA) and sodium polyanethol sulfonate, (PAS)) were dissolved in bidistilled water at concentration 5 mg ml−1. For preparation of PDDA/ChOx nanofi lms, a 5 mkl drop of PDDA solution was put on the surface of MnO2-modifi ed electrodes and in 10 min (before the drop dried) the electrodes were rinsed with bidistilled water for 1–2 min. The electrodes were then dried and a 5 mkl drop of ChOx solution was put on the electrodes’ surface (the optimal concentration of ChOx in the solution was estimated as 0.5 mg/ml). After 10 min of adsorption, the electrodes were rinsed with water and dried. The same procedure was used to prepare complex nanofi lms containing several interpolyelectrolyte layers (PDDA/PAS)2 and several enzyme/polyelectrolyte layers (PDDA/ChOx)n on the electrode surfaces.

The general schemes for biosensor architecture and for choline detection by amperometric choline oxidase based biosensors are shown in Figure 2. Biosensor response at different concentrations of choline oxidase in solution for adsorption was evaluated (Table 2). The analytical response increases from 0.05 mg mL-1 to 4.00 mg mL-1 of choline oxidase concentration in solution for adsorption. For validation of sensor fabrication reproducibility we have use R.S.D. % referred to data from fi ve different sensor electrodes. It should be noted, that minimal R.S.D. (13-16%) was observed, when concentration of choline oxidase vary from 0.05-0.5 mg mL-1. Thus, the selected optimum of enzyme concentration for effective biosensor functioning was 0.5 mg mL-1.

Figure 2. Architecture of enzyme/polyelectrolyte layers of MnO2-based choline oxidase biosensor a) PDDA/ChOx, b) (PDDA/PAS)2/PDDA/ChOx, c) scheme of choline detection.

Page 57: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

55

Table 2. Dependence of analytical response (0.1 mM of choline) and reproducibility of MnO2/PDDA/ChOx biosensor on choline oxidase concentration. Measurement conditions: 50 mM Hepes, pH 7.5, 30 mM KCl, 480 mV vs Ag/AgCl.

ChOx concentration, mg·ml-1 ∆I, nA R.S.D. % of the response to choline

for fi ve different biosensors

0.05 58 16

0.10 98 13

0.50 239 13

1.00 276 24

2.00 318 29

4.00 331 37

Investigation of operational stability of the developed choline oxidase biosensors shows that there is a trend for decrease in analytical response from measurement to measurement.

The decrement of sensor analytical response was 2.1±0.1% per one measurement (R.S.D. was 7.0±0.3% per ten measurements) at working potential 480 mV. The operation stability of MnO2 layer was investigated at different hydrosol concentrations. Aiming the above reason, the concentrations of MnO2 in hydrosol were chosen for linear (OD = 0.17) and saturated (OD = 1.5) response-concentration curve areas. Sensor operation stability was determined in the same ranges of concentrations, i.e. the decrement of analytical response was estimated as 0.2±0.4% (R.S.D. = 2.0±0.1%). All this proves that hydrogen peroxide-sensitive layer remains intact. It follows, that the observed decay in sensor response is connected with the enzyme layer. Consequently, the improved stabilization of choline oxidase layers was carried out with two additional PDDA/PAS layer’s variants (Figure 3b). The decrement of analytical response in this case was 0.6±0.2% (R.S.D. 1.9±0.4%) for MnO2/(PDDA/PAS)2/PDDA/ChOx electrodes, at working potential 480 mV.

The pH dependence values (in the range from 7.0 to 8.2.) of the electrochemical response for MnO2/(PDDA/PAS)2/PDDA/ChOx electrodes have shown the increase of response with pH increasing. It is known that pH-optimum of choline oxidase is about 8.0. However, the observed operation stability of choline oxidase biosensors was better at pH 7.5 (decrement of analytical response 0.6±0.2% and R.S.D. = 1.9±0.4%) than at pH 8.2 (decrement of analytical response 3.2±0.0% and R.S.D. = 11.1±0.4%). The biosensor response to 0.1 mM choline of the electrodes stored at 4oC and tested every week kept 75% of initial activity for 3 weeks of storage.

The typical steady-state response of choline oxidase biosensor to 0.1 mM of choline is shown in Figure 3a. The time needed to reach the 90% of the fi nal response was about 10 seconds. Figure 3b shows the dependence of choline oxidase biosensor sensitivity on

Page 58: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

56

number of PDDA/ChO layers. The electrode sensitivity increases at magnifi cation of PDDA/ChO layers from 1 to 3, and reaches upper limit.Figure 3c and 3d illustrate the dependence of the MnO2/(PDDA/PAS)2/PDDA/ChOx sensor responses at different concentrations of choline. Obtained choline calibration curve showed a good linearity in the range between 3.0⋅10-7-1.0⋅10-4 M. The corresponding regression equation was: y=2.9⋅x+3.5, where y represents the current in nA and x - the choline concentration in mkM. The sensitivity 59±3 mA·M-1⋅cm-2 and the detection limit 300 nM (3σ) were calculated. It should be noted that the highest sensitivity of the developed biosensors was obtained when three enzyme-containing layers were deposited on the electrode surface: MnO2/(PDDA/PAS)2/(PDDA/ChOx)3 (Figure 3c and 3d).

Figure 3. a)Analytical response to 0.1 mM of choline and b) dependence of choline oxidase biosensor sensitivity on number of PDDA/ChO layers с), d) dependence of the MnO2/(PDDA/PAS)2/PDDA/ChOx (line I) and MnO2/(PDDA/PAS)2/(PDDA/ChOx)3 (line II) sensor responses at the different concentrations of choline.

Measurement conditions: 50 mM Hepes, 30 mM KCl, pH 7.5, 480 mV vs. Ag/AgCl.

The limit of detection was estimated as 130 nM (3σ) and sensitivity was estimated as 103±3 mA·M-1⋅cm-2.These analytical parameters are the best known for amperometric choline oxidase based biosensors.

1.3. Tyrosinase based biosensor.The tyrosinase (Tr) based biosensors were prepared as described below. The SPEs were

fabricated on polyvinyl chloride substrate of 0.2 mm thickness by means of conductive graphite paste (Gwent, UK) screen-printed by a semi-automated machine Winon (model WSC-160B, China) with a 200 mesh screen stencil. Each SPE consisted of a round-

Page 59: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

57

shaped working area (3 mm diameter), a conductive track (30 mm×1.5 mm), and a square extremity (3 mm×7 mm) for electrical contact. Polyelectrolyte PDDAwas dissolved in 50 mM PB, pH 7.0 at concentration 5 mg⋅ml−1. For preparation of PDDA/Tr nanofi lms, a 5 mkl drop of PDDA solution was put on the surface of SPE and after 50 min the electrodes were rinsed with bidistilled water for 2 min. The electrodes were then dried and a 5 mkl drop of Tr solution was put on the electrodes’ surface (the optimal concentration of Tr in the solution was estimated as 5⋅10-6 M). After 10 min of adsorption, the electrodes were rinsed with water for 2 min. The application ofall componentswasperformedin a climatic chamber at 250C and relative humidity about 60%. At the fi nalstage the electrodeswere driedat ambient conditions.The general scheme for phenol detection by amperometric tyrosinase based biosensors are shown in Figure 4.

Figure 4. Tyrosinase based biosensor

Analytical characteristics of tyrosinase based biosensors for phenol detection are presented in Table 3

Table 3. Analytical characteristics of tyrosinase based biosensors for phenol detection.

Parameter Value

Limit of Detection, nM 12

Linear range, M 25•10-9 – 1•10-5

Sensitivity, A/(M•cm2) 0.44

Operational stability,Decrement for the response (%)/measurement -0.5

Residual Biosensor Activity after 1 month(storage temperature +40C), % from initial value 75

Tyrosinase

Catehol

o-Quinone

Tyrosinase

H2O

1/2 O2

Electrode�

1/2 O2

H2O

I, nA

t, s 0

Biosensorresponse

Pheno

Page 60: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

58

Thus, obtained analytical parameters are the best known for SPE amperometric tyrosinase based biosensors.

1.4. The effects of various interfering substances on choline oxidase and tyrosinase biosensors.

An important factor for evaluating the analytical performance for medical and environmental applications of an electrochemical choline biosensor is the interference of sampling “contaminating” compounds, easily oxidized at positive potentials. Therefore, these potential aberrations introduced by interfering compounds were additionally investigated using couple of substances commonly found in biological fl uids (ascorbic and uric acids) and environmental objects (heavy metals like Cd2+, Co2+, Cu2+). Two groups of samples were examined: “pure, non-contaminated” control samples, and those, spiked with an interfering compounds. The summarized results are shown in Table 4. The interference for the normal physiological level of ascorbic acid (5·10-5 M) increased the MnO2/(PDDA/PAS)2/PDDA/ChOx biosensor response to 394% and decreased to 0.5% at concentration 5·10-7 M in the sample spiked with choline. Similar effect was found for uric acid (see Table 4). Thus, at least, 200-fold dilution of real blood samples is necessary to eliminate the interference of analogous concomitant compounds on studied electrode response.

Heavy metals (Cd2+ and Co2+) are reversible inhibitors of choline oxidase. The inhibition effect of Cd2+ and Co2+ ions is negligible at the concentrations lower than 10-5 M Cd2+ and 10-4 М Co2+, respectively (Table 4). In our experiments, Cu2+ does not show any infl uences on choline oxidase electrode function in concentration < 1 mkM.

Table 4. The effect of interfering compounds on analysis of choline. Electrodes: MnO2/(PDDA/PAS)2/PDDA/ChOx. Measurement conditions: 50 mM Hepes, pH 7.5, 30 мМ KCl, 480 mV vs Ag/AgCl, choline concentration in the electrochemical cell 0.1 mM.

Components of biological liquids

Normal level in human blood serum

Measured concentration

Response change, %

Ascorbic acid 50 mkM

50 mkM 394±125 mkM 29±32 mkM 13.7±3.3

0.5·mkM 0.5±4.5

Uric acid 500 mkM

50 mkM 129±475 mkM 24±112 mkM -1.0±2.4

0.5·mkM 15±10

Ions of heavy metals Maximum permissible concentration in fresh water .

Cd2+ 10 mg⋅m-3 (90 nM)1 mM -40±0

0.1 mM -19.5±1.510 mkM -4.5±0.5

Co2+ 0.01 mg⋅ L-1 (170·nM)1 mM -26.5±0.5

0.1 mM -10±010 mkM -5±0

Cu2+ 2 mg⋅L-1 (30 mkM)1 mM -89±4

10 mkM -40.5±1.51 mkM -6±2

Page 61: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

59

The infl uence of potentially interfering agents found in blood on biosensor phenol detection are presented in Table 5. The concentrations of interfering agents were selected to be close to their normal levels in blood plasma. Additional dilutions (1/10 and 1/100, v/v) were tested for the cases where interference was observed. For adrenaline, the full range of its concentrations in plasma was investigated from the normal state (0.5 nМ) to 100 times this level in stress conditions. As can be seen from Table 5, some interference was observed at the maximum levels of interfering agents; however, their effects were readily attenuated with sample dilution.

Table 5. Infl uence of interfering components from blood plasma on biosensor detection of phenol.a

Interfering agent Concentration in plasma (M)

Concentration introduced (М)

Change in analytical signalb

(Iint-I0)/(I0×100%)

Glucose 5 ×10-35 ×10-3

5 ×10-4

5 ×10-5

-15.0-11.0-0.5

Ascorbic acid 5 ×10-55 ×10-5

5 ×10-6

5 ×10-7

-13.3-8.4-0.4

Adrenaline 5 ×10-8 (under stress)5 ×10-10 (normal)

5 ×10-8

5 ×10-100.00.0

Uric acid 5 ×10-45 ×10-4

5 ×10-5

5 ×10-6

-7.4-4.0-1.5

L-tyrosine 2 ×10-5 2 ×10-5 0.012

a Measurement conditions: 100 mM NaCl, 50 mМ Na phosphate, pH 7.0, -150 mV vs. Ag/AgCl, [ phenol] = 10 μM.

bIint = analytical response (current) to 10 μM phenol in the presence of a specifi ed concentration of an interfering agent; I0 = analytical response (current) to 10 μM phenol in the absence of interfering agent.

2. Biosensor analysis of blood esterases.This part of the paper demonstrates the possibility of the developed electrochemical

biosensorsto detect the activities of the mainbloodesterases, such as theAChE,BChE, CaE and NTE.Blood samplewas preparedas follows: fresh or thawed after storage at -700C whole blood diluted with a buffer solution 100 times and quickly frozen at -700C (or in liquid nitrogen) for the destruction of the blood corpuscles. Thereafter, samples of blood (hemolysate) could be stored for several months at -700C until use without signifi cant decrease in activity of target enzymes.

We have developed the following schemes of analysis of cholinesterase (Fig. 5), CaE (Fig. 6) and NTE (Fig. 7).

In accordancewith the scheme (Fig. 5) to determine the activity of acetylcholinesterase, the required amount of the hemolysate were incubated with selective BChE inhibitor (iso-OMPA) for 10 minutes, then added substrate acetylcholine and incubated for a time t1. To determine the activity of AChE/BChE, hemolysate was incubated with acetylcholine/butyrylcholine for a time t2 (hereinafter, such a mixture comprising a hemolysate inhibitor, the substrate is called the incubation mixture). An aliquot of the incubation mixture was transferred into an electrochemical cell, diluting it 25 times. As was shown in preliminary experiments, 25-fold dilution of the incubation mixture taking place in the electrochemical

Page 62: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

60

cell results in a complete stop of the enzymatic hydrolysis. Next, we measured the amount of choline, accumulated at the time of stopping the reaction by dilution, using choline oxidase biosensor. Biosensor response resulting in the incubation mixture is proportional to the enzyme activity corresponding hemolysate.

In accordancewith the scheme (Fig. 6), to determine the activity of CaE the hemolysate was pretreated with 2 mM EDTA and 40 μM eserine for 10 min at room temperature to inhibit PON1 and cholinesterases, respectively. Then samples were incubated for 5 min with 1 mM phenyl acetate at room temperature and assayed for phenol after 50-fold dilution.

Principle of NTE activity assay is summarized in Fig. 7. If A is the total phenyl valerate hydrolyzing activity (PVase activity), B is the activity preinhibited with paraoxon (nonneuropathic OP), and C is the residual activity after both paraoxon and mipafox (neuropathic OP) preinhibition, then the NTE activity can be calculated as the difference between B and C. To determine the NTE activity in blood, wehave usedthe hemolysate diluted 200 times.

Figure 5. The general scheme of measuring the activity of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) in the blood.I*BChE – the inhibited form of the BChE.

Figure 6. The general scheme of measuring the activity of CaE in the blood.I*AChE, I*BChE, I* PON1 – the inhibited form of the enzymes.

Page 63: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

61

2.1. Optimization of cholinesterase activity assay.Investigation of the infl uence of dilution of hemolysate in the measuring cell on

the response of choline oxidase biosensor showed that dilution of hemolysate in the electrochemical cell 50 (or more) timeshas no appreciable effect on the analytical characteristics of the biosensor. In this regard, the hemolysate diluted in the incubation mixture of 2 times followed by 25-fold dilution in the cell (50-fold dilution of the hemolysate) was optimal and was used for AChE and BChE assay.

To estimate the correctness of the measurement method of cholinesterase activity we investigated the kinetics of accumulation of choline in the enzymatic hydrolysis of large and small substrate concentrations (acetylcholine (ACh) and butyrylcholine (BCh)) in the blood of mice. Over the entire time range and for different concentrations of substrates, we observed a linear dependence of the sensor response on the time of incubation. Obviously, the longer the incubation the higher is the sensitivity. However, the total time of the procedure for determining the activity of enzymes should be as short as possible. To determine the initial rate of enzymatic hydrolysis of ACh and BCh the 30 min time of incubation was chosen, located in the linear region. Dependences of the rate of enzymatic hydrolysis of ACh and BCh on the concentration of the respective substrates are hyperbolic curves with saturation. 90% of the response is achieved at concentrations of BCh and ACh 3 and 10 mM,respectively.

We have investigated the concentration dependences of human erythrocyte AChE activity and horse serum BChE activity in the presence and absence of the hemolysate in the incubation mixture. The obtained dependences are parallel straight lines, indicating to the contribution to the response of only own AChE or BChE activity of the hemolysate and the absence of infl uence of the matrix on the amperometric signal. We calculated the limits of detection of these enzymes on the basis of the minimum detectable concentrations of choline, calculated at a ratio of S / N = 3. The limits of detection of AChE and BChE was 4.3 and 0.8 nmol / min x ml, respectively.

2.2. Optimization of CaE and NTE activity assay.Before attempting assay of CaE and NTE activities in blood samples, the electrochemical

detection of commercial porcine liver CaE and hen brain NTE activity by the tyrosinase

Figure 7. The general scheme for measuring the activity of NTE.

0

10

20

90

100

NTE activity(B-C)

C

B

A

Incu

batio

n w

ith 5

0 μM

of p

arao

xon

and

250

μMof

mip

afox

for 2

0 m

in

at 3

7 o C

Incu

batio

n w

ith 5

0 μM

of p

arao

xon

for 2

0 m

in

at 3

7 o C

Phen

yl v

aler

ate

hydr

olys

ing

activ

ity, %

Page 64: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

62

biosensor was tested in the absence and presence of blood. Serial dilutions of each esterase preparation were made and the activity of each sample was measured in the absence and presence of an aliquot of diluted blood homogenate. Preliminary experiments with a range of dilutions (20- to 200-fold) of whole human or mouse blood homogenates showed that neither the response of the biosensor to phenol nor its recovery time for subsequent measurements were affected by the presence of blood if it was diluted at least 100-fold. The experiments showed that only an upward parallel shift of the calibration curve occurred in the presence of blood for CaE (Fig. 8) or NTE (Fig. 9). These results confi rm the absence of a matrix effect of blood on the biosensor assay for CaE and NTE activity. The upward shift in each enzyme calibration curve is due to the intrinsic activities of CaE and NTE in whole homogenized blood, as discussed further below.

Figure 8. Amperometric assay of porcine liver CaE activity in the absence (lower line, open circles) or presence (upper line, closed circles) of human blood homogenate (1/500 v/v). Sensor response (Y) = relative to response at standard [ phenol] = 10 μM. Data are mean ± SD (n = 7, absence of blood; n = 5, presence of blood). Y(absence) = (0.70 ± 0.013)X – (0.0008 ± 0.0082), R2 = 0.987; Y(presence) = (0.76 ± 0.050)X +

(0.290 ± 0.034), R2 = 0.910. Slopes of the two lines were not statistically different (p = 0.12); Y-intercepts were statistically different (p < 0.0001). The change in intercept caused by addition of blood was due to intrinsic

CaE activity in blood.

2.3. Validation in the experiments on in vitro concentration-dependent inhibition of human blood choline esterases.

Selective in vitro inhibition of AChE and BChE activities in whole blood hemolysate was performed using mouse blood and the following inhibitors: iso-OMPA for BChE and (-)Huperzine A - for AChE. Measurement of the residual esterase activity in blood was carried out by two methods: electrochemical method in optimal conditions according to the developing format and spectrophotometric analysis by the standard method of Ellman. In the spectrophotometric method for determining the activity of AChE and BChE we used the thio analogues of substrates: acetylthiocholine and butyrylthiocholine.

The titration curves of enzymatic activities (% inhibition depending on the concentration of the inhibitor) obtained by the biosensor and spectrophotometric methods were quite

Page 65: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

63

close, even if the use of different substrates (Fig. 10). In both cases, there was almost 100% inhibition of the target enzyme by corresponding specifi c inhibitor.

Table 6 shows the IC50 values for each enzyme obtained by the two methods. The table shows that the compared analytical parameter has similar values when using the biosensor and spectrophotometric methods, which confi rms the validity and correctness of the defi ned activities of enzymes and is a successful outcome of the fi rst phase of validation of the developed assay format.

These data confi rm the authenticity and validity of the biosensor measurement and demonstrate the promising of the new biosensor for biomonitoring of OPC exposure on living organisms, including humans.

Figure 9. Amperometric assay of hen brain PVase (main graph) and NTE (inset) activities. Data are mean ± SD, n = 5. Open squares, dashed line: PVase activity of partially purifi ed hen brain NTE preparation

preincubated with 50 μM paraoxon at 37°C for 20 min (activity “B”, absence of blood); Y = (0.010 ± 0.00047)X – (0.026 ± 0.0146), R2 = 0.953. Closed squares, solid line: same as open squares, dashed line, but

in the presence of 1/200 (v/v) diluted human blood homogenate preincubated with 50 μM paraoxon at 37°C for 20 min (activity “B”, presence of blood); Y = (0.0097 ± 0.00048)X + (0.060 ± 0.015), R2 = 0.947. Open circles, dashed line: PVase activity of partially purifi ed hen brain NTE preparation preincubated with 50 μM paraoxon

and 250 μM mipafox at 37°C for 20 min (activity “C”, absence of blood); Y = (0.0031 ± 0.00037)X – 0.023 ± 0.012), R2 = 0.754. Closed circles, solid line: same as open circles, dashed line, but in the presence of 1/200 (v/v) diluted human blood homogenate pretincubated with 50 μM paraoxon and 250 μM mipafox at 37°C for 20 min; Y = (0.0037 ± 0.00034)X + (0.0178 ± 0.011), R2=0.837. PVase was measured after 40 min incubation of samples with 0.56 mM PV at 37°C. Phenol was assayed amperometrically after 20-fold dilution of samples. Inset, open triangles, dashed line: NTE activity (activity “B-C”) in the absence of 1/200 (v/v) diluted human

blood homogenate; Y = (0.0070 ± 0.00045)X – (0.0010 ± 0.014), R2 = 0.988. Inset, closed triangles, solid line: NTE activity (activity “B-C”) in the presence of 1/200 (v/v) diluted human blood homogenate; Y = (0.0060 ± 0.00051)X + (0.042 ± 0.016), R2 = 0.978. Sensor responses are given as electrochemical signal normalized to response to standard phenol concentration (10 μM). Slopes of “B” lines (absence and presence of blood) were not statistically different (p = 0.473), but Y-intercepts were different (p < 0.0002). Slopes of “C” lines

(absence and presence of blood) were not statistically different (p = 0.255), but Y-intercepts were different (p < 0.012). Slopes of “B-C” lines (absence and presence of blood) were not statistically different (p = 0.184), and Y-intercepts were not statistically different (p = 0.087). The change in Y-intercepts caused by addition of blood

was due to intrinsic PVase activity in blood.

Page 66: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

64

A) B)Figure 10. Titration curves of AChE by (-)huperzine A (A) and BChE by iso-OMPA (B) in blood mice

obtained using spectrophotometric (●) and electrochemical (Δ) techniques. The insert shows the correlation (R) between the results of measurements obtained by electrochemical and spectrophotometric methods. For AChE

R = 0.999, for BChE R = 0.987.

Table 6. The IC50 values for in vitro inhibition of specifi c inhibitors of AChE and BChE mouse blood based on the results of spectrophotometric and electrochemical methods.

(-)Huperzine A,IC50±SE, M

iso-OMPA,IC50±SE, M

Enzyme AChE BChE

Spectrophotometry (2.9±0.4)⋅10-9 (1.4±0.1)⋅10-6

Biosensor (0.80±0.06)⋅10-9 (0.85±0.05)⋅10-6

2.4. Validation in the experiments on in vitro concentration-dependent inhibition of human blood CaE and NTE.

Activity of CaE in blood homogenates from humans and mice were measured in paired samples using the (PDDA/ tyrosinase/GA) biosensor and the spectrophotometric method. In addition, NTE activity was measured in aliquots from the paired samples using only the biosensor method, because the activity of this enzyme cannot be measured spectrophotometrically in homogenates of whole blood. The results in Table 2 show that there is excellent agreement between the biosensor and spectrophotometric results for CaE activity for both human and mouse blood. In addition, these values are in good agreement with previously reported electrochemical measurements in hemolyzed blood samples.

Table 7. CaE and NTE activities in human or mouse blood homogenates.

Subject NumberaCaEb (μmol/min/

ml whole blood)NTEc

(nmol/min/ml whole blood)spectrophotometric biosensor biosensor

H-1H-2H-3H-4

H-Mean ± SEM (n = 4)

0.560.140.100.13

0.23 ± 0.11d

0.410.180.120.14

0.22 ± 0.07d

41.450.430.625.2

36.9 ± 5.62e

Page 67: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

65

M-1M-2M-3M-4

M-Mean ± SEM (n = 4)

6.257.245.805.43

6.18 ± 0.39f

4.267.796.645.46

6.04 ± 0.76f

9.119.817.614.6

15.3 ± 2.32g

a H = human; M = mouse.b CaE activity determined either spectrophotometrically or electrochemically (via biosensor).c NTE activity can only be determined electrochemically (via biosensor) in blood homogenates.d-g Mean values with the same letter are not signifi cantly different from each other; values with different

letters are signifi cantly different from each other (2-way repeated measures ANOVA on log-transformed data to correct for unequal variances,Tukey-Kramer post-hoc test for all pairwise comparisons,α = 0.05).

With respect to other published data on blood CaE levels, data could be found only for plasma. Apparent CaE activity in human plasma is very low; when determined with 1-naphthyl acetate, it is reported to be 0.019 ± 0.001 μmol/min/ml. Using nondenaturing gradient gel electrophoresis and staining for esterase activity, CaE was undetectable in human plasma. However, CaE is found in monocytes, which are the likely source of the activity detected in whole blood homogenates in our studies.

Our results also show that the apparent CaE activity of rodent (mouse) blood is higher than that of humans, which is in accord with qualitatively high levels detected on gels in mouse plasma and quantifi ed as 3.52 ± 0.15 μmol/min/ml in rat plasma.

Spectrophotometric technique does not allow determining NTE activity in whole blood. As we demonstrated earlier, NTE assay in whole blood is possible only using a phenol biosensor. Activity of NTE in human blood determined in this work with the new LBL biosensor (Table 6) was close to that obtained earlier (0.19 ± 0.02 nmoles/min per mg of protein; equivalent to 32.3 ± 3.6 nmol/min/ml blood) . We also found that the mean value for mouse blood NTE activity was 41.5% of that measured in human blood; similarly, the mean value for NTE activity in mouse platelets was previously determined by Husain to be 42.8% of that in human platelets.

2.4.1. Biosensor assay of mouse blood CaE following its inhibition in vivo

A biomonitoring assessment of the tyrosinase biosensor was carried out by comparing electrochemical and spectrophotometric measurements of blood CaE activity changes ex vivo after dosing mice in vivo with DEHFPP. This compound was previously found to be a relatively potent OP inhibitor of CaE in vitro (ki = 1.20×105 M-1min-1). As shown in Fig. 6, blood CaE activity was inhibited in a dose-dependent manner by DEHFPP treatment, yielding effective dose 50% (ED50) values [mean (95% CI) (n)] of 25.5 (23.2, 28.0) mg/kg (6) and 21.1 (18.9, 23.5) mg/kg (4) for spectrophotometric and electrochemical assays, respectively. Although the ED50 values were signifi cantly different from each other (p< 0.007), an excellent correlation between biosensor and spectrophotometric measurements was found (r = 0.99) (Fig. 6, inset), indicating that the differences between the two methods are systematic and providing validation of the biosensor assay.

Page 68: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

66

Figure 11. Main graph: dose-related inhibition of CaE activity in mouse blood 1h after dosing with DEHFPP (structure shown on graph). CaE activity in whole blood was determined by spectrophotometric

(fi lled circles) and biosensor (circles) methods and presented as % inhibition of control activity. Data are mean ± SE, n = 4-6. Control CaE activity (mean ± SE) = 6.02 ± 0.27 μmol/min/ml blood (n =20) and 6.04 ±1.52

(n =5) for spectrophotometric and biosensor methods, respectively. ED50 for CaE inhibition, mean (95% CI) = 25.5 (23.2, 28.0) mg/kg (n = 6) and 21.1 (18.9, 23.5) mg/kg (n = 4) for spectrophotometric and electrochemical

assays, respectively. Inset, fi lled diamonds: correlation of blood CaE % inhibition obtained with biosensor and spectrophotometric methods; mean ± SE, r = 0.99, p <0.0001, n = 7.

3. ConclusionThe effi ciency of enzyme-polyelectrolytes nanofi lms, deposited on carbon screen-

printed electrodes (SPE), as a highly-sensitive transducer element of biosensing platforms, fabricated with “ layer-by-layer” technology was demonstrated. The present report describes several analytical possibilities of the biosensing platform based on choline oxidase (ChO) and tyrosinase nanocomposites.

The fi rst one is SPE amperometric sensors based on aqueous gamma-phase-MnO2 as hydrogen peroxide oxidation mediating reagent. The revealed analytical characteristics of the choline oxidase biosensor are promising for highly-sensitive monitoring of cholinesterases and their inhibitors in medical, biological and environmental samples.

The second one is phenol registration. Highly sensitive tyrosinase SPE biosensors based on nanostructured polyelectrolyte fi lms were developed for these purposes. The methodologies of neuropathy target esterase, carboxylesterase activity assay in the blood were designed using developed tyrosinase SPE biosensors.

4. AcknowledgementsThe fi nancial support for this work from Russian Foundation for Basic Research

(project no. 10-08-00895-a, 11-08-01306-a and 13-08-01078) and NATO grant SfP #984082 are gratefully acknowledged.

Page 69: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

67

References

[1] E.A.Dontsova,Y.S.Zeifman,I.A.Budashov,A.V.Eremenko,S.L.Kalnov,I.N.Kurochkin,Screen-printed carbon electrode for choline based on MnO2 nanoparticles and choline oxidase/polyelectrolyte layers,SensorsandActuatorsB:Chemical,159 (2011) 261-270.

[2] L.V.Sigolaeva, A.Makower, A.V.Eremenko, G.F.Makhaeva, V.V.Malygin, I.N.Kurochkin, F.W.Scheller. Bioelectrochemical Analysis of Neuropathy Target Esterase Activity in Blood, Anal.Biochem.,290 (2001) 1-9.

[3] I.N. Kurochkin, A.V. Eremenko, G.F. Makhaeva, L.V. Sigolaeva, G.G. Dubacheva, and R.J. Richardson.Multi-strip assay and multimodal biosensors for environmental and medical monitoring of neurotoxicants.NATO Security through Science Series A, (2009) 219–229.

[4] L.V. Sigolaeva, G.F. Makhaeva, E.V. Rudakova, N.P. Boltneva, M.V. Porus, G.V. Dubacheva, A.V. Eremenko, I.N. Kurochkin, and R.J. Richardson, Biosensor analysis of blood esterases for organophosphates exposure assessment: Approaches to simultaneous determination of several esterases.ChemBiol Interact,187 (2010) 177-184.

[5] M.S. Gromova, L.V. Sigolaeva, M.A. Fastovets, E.G. Evtushenko, I.A. Babin, D.V. Pergushov, S.V. Amitonov, A.V. Eremenko, I.N. Kurochkin, Improved adsorption of choline oxidase on a polyelectrolyte LBL fi lm in the presence of iodide anions, Soft Matter, 7 (2011) 7404-7409.

[6 A.V. Eremenko, E.A. Dontsova, A.P. Nazarov, E.G. Evtushenko, S.V. Amitonov, S.V. Savilov, L.F. Martynova, V.V. Lunin, I.N. Kurochkin, Manganese Dioxide Nanostructures asa Novel Electrochemical MediatorforThiol Sensors, Electroanalysis, 3 (2012), 573-580.

Page 70: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

68

Chapter 5

Tyrosinase-Based Biosensors for Assay of Carboxylesterase, Neuropathy Target Esterase,

and Paraoxonase Activities in blood

Larisa V. SIGOLAEVAa, Arkadi V. EREMENKOb, Elena V. RUDAKOVAc,Galina F. MAKHAEVAc, Ilya N. KUROCHKINa1

aDepartment of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russia

bN.M. Emanuel Institute of Biochemical Physics, Russian Academy of Scienses, Moscow 119334 Russia

cInstitute of Physiologically Active Compounds, Russian Academy of Sciences, Moscow Region, Chernogolovka, 142432 Russia

Abstract. A tyrosinase-based phenol biosensors were formed via layer-by-layer ( LBL) approach to deposit poly(dimethyldiallylammonium chloride) (PDDA) and tyrosinase onto graphite-based substrates, followed by optional glutaraldehyde (GA) crosslinking. The good analytical parameters of phenol detection thogether with low detection limit enabled phenol measurements in highly diluted blood samples, which also minimized interference from extraneous (non-analyte) substances in blood (e.g., adrenaline, ascorbate, glucose, L-tyrosine, and urate). Applicability of the biosensor to analysis of carboxylesterase ( CaE), paraofonase 1 ( PON1) and neuropathy target esterase ( NTE) activities in mouse and human blood was demonstrated. Parallel biosensor and spectrophotometric CaE analyses were carried out 1 h after intraperitoneal injection of mice with the model dialkylphosphate, (C2H5O)2P(O)OCH(CF3)2 (DEHFPP). Dose-related inhibition of CaE activity was observed, yielding ED50 values [mean (95% CI) (n)] of 25.5 (23.2, 28.0) mg/kg (6) and 21.1 mg/kg (18.9, 23.5) (4) for spectrophotometric and electrochemical assays, respectively. Although the ED50 values were signifi cantly different from each other (p< 0.007), excellent correlation between biosensor and spectrophotometric measurements was found (r = 0.99), indicating that the differences between the two methods are systematic and providing validation of the biosensor assay.

Key words. biosensor; blood assay; carboxylesterase ( CaE); layer-by-layer ( LBL); neuropathy target esterase ( NTE); paraoxonase ( PON1); phenol; tyrosinase

1 Laboratory of Postgenomic Chemistry, Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russia E.mail: [email protected]

Page 71: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

69

1. Introduction

Detection and quantifi cation of phenolic compounds in medical and environmental matrices is frequently required because phenolic compounds are commonly involved in health care and environmental monitoring [1]. In biochemistry and medicine there is a particular need for monitoring enzymatic processes that proceed with the release of phenols and that could be carried out in highly diluted blood samples to minimize interference from extraneous (non-analyte) substances. Obviously, increased dilution results in decreased concentration of the target analyte, thus requiring increased sensitivity of the analytical method. The present work describes the application of a highly sensitive electrochemical technique for measuring phenol produced three toxicologically and biochemically important enzymes in blood samples from humans and experimental animals: carboxylesterase, paraoxonase, and neuropathy target esterase. At least two of them ( carboxylesterase and neuropathy target esterase) have low-level activities in blood therefore the alternative analytical methods are highly required.

Carboxylesterase (EC 3.1.1.1, CaE) is involved in biotransformation of various drugs, environmental toxicants, and carcinogens containing ester groups [2-5]. It is one of the main enzymes involved in the detoxifi cation of organophosphorus (OP) compounds and serves as a determinant of individual sensitivity to these agents [6,7]. Measurment of blood CaE activity is important for the development and use of ester-containing drugs and could be benefi cial for biomonitoring individuals who have occupational or accidental contact with OP compounds. However, because humans have a low level of blood CaE activity [7], its spectrophotometric determination is problematic. In addition to aforementioned carboxylesterase activity in some species in relation to agrochemical exposure may be useful for ecosystem-wide environmental monitoring projects [8]. Fish, amphibians and birds have blood carboxylesterases which are generally more sensitive to OP compounds exposure than brain acetylcholinesterase. A signifi cant advantage of carboxylesterase activity-based biomarkers is that assays can be nondestructive in contrast to use of brain acetylcholinesterase activity.

Human serum paraoxonase (aryldialkylphosphatase, EC 3.1.8.1, PON1), is a calcium-dependent hydrolase that is tightly associated with high-density lipoproteins. PON1 is thought to be an important determinant of an individual’s sensitivity to some OPCs, based primarily on evidence from studies on animals models [9,10]. PON1 hydrolyzes the active metabolites (oxons) of certain OP insecticides (e.g., chlorpyriphos oxon and diazoxon) as well as nerve agents such as soman, sarin, tabun, and less VX, and it plays a central role in their detoxifi cation and toxicity. Animals with low paraoxonase levels (e.g., birds) were more sensitive to specifi c OP compounds than animals with high enzyme levels (e.g., rats and especially rabbits) [11]. In human populations, serum paraoxonase exhibits a substrate-dependent polymorphism as well as a large variability in plasma levels among individuals. These determine to a great extent an individual’s sensitivity to OP compounds and a requirement of new alternative approaches to fast and reliable PON1 activity determination.

Neuropathy target esterase (EC 3.1.1.5, NTE) is a specifi c target and biomarker of OP compound-induced delayed neuropathy (OPIDN). A good correlation between the inhibition/aging of brain/lymphocyte/ blood NTE within hours of exposure and the

Page 72: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

70

subsequent induction of OPIDN [7, 12-15] enables application of blood NTE activity determination for earlier diagnosis of OPIDN. However, because of the low level of NTE activity in blood and interference from hemoglobin absorbance, the colorimetric assay cannot be used to determine NTE activity in whole blood [16].

Given that currently used spectrophotometric methods for assay of blood CaE and NTE activity suffer from limited sensitivity or interference, substitution of these analytical methods with a more sensitive and less interference-prone biosensor technique would be desirable. Because of their sensitivity and indifference to spectrophotometric interference, tyrosinase-based biosensors have proved to be promising for this purpose. These devices employ the enzymatic oxidation of phenol via catechol into o-quinone, a reaction that consumes molecular oxygen. Electroreduction of quinone to catechol directly on the graphite electrode can be used as a detection reaction for the quantifi cation of phenol. The use of such biosensors allows the detection limit for the monitoring of phenols to reach nanomolar and even subnanomolar [17-23] levels.

Our prior experience with biosensor fabrication on the basis of sequential adsorption driven by electrostatic interactions of cationic and anionic species on a charged support ( layer-by-layer, LBL technique) resulted in a highly sensitive biosensors for phenol or choline assay. These biosensor coatings comprise tyrosinase [24-26] or choline oxidase [27, 28], respectively, fabricated via LBL deposition of poly(dimethyldiallylammonium chloride) (PDDA) and respective enzyme onto a polished graphite rod (GR), and its functionality was optionally preserved by glutaraldehyde (GA) crosslinking.

The present report describes the analytical properties of GR/PDDA/Tyrosinase/GA graphite-substrate biosensor as well as the new one SPE/PDDA/Tyrosinase, wherein the similar biosensor design has been realized on the surface of graphite-based screen-printed electrode (SPE). The application of both type of biosensorsto for the electrochemical analysis of CaE, PON1, and NTE in blood samples of humans and experimental animals (mice) was also developed that enables this approach to be of practical use in biomedical chemistry and toxicology.

2. Materials and methodsChemicals and enzymes Carboxylesterase ( CaE) from porcine liver (EC 3.1.1.1), activity 177 U/mg of protein

for phenyl acetate; eserine (physostigmine) hemisulfate; glutaraldehyde (GA); graphite rods, 3 mm in diameter; mushroom tyrosinase (EC 1.14.18.1), activity 3800-4700 U/mg for L-tyrosine; paraoxon; phenol; phenyl acetate; and poly(dimethyldiallylammonium chloride) (PDDA, Mw = 400000–500000, 20% (w/w) aqueous solution) were purchased from Sigma-Aldrich (Munich, Germany). Human recombinant paraoxonase ( PON1) (EC 3.1.8.1), activity 68 U/ml was a kind gift of Prof. J.F. Tieber, The University of Texas Southwestern Medical Center, US. Phenyl valerate (PV) and mipafox were obtained from Oryza (Chelmsford, MA, USA). A lyophilized hen brain membrane fraction consisting of combined mitochondrial and microsomal pellets (P2+P3) and preinhibited with diethyl 4-nitrophenyl phosphate ( paraoxon) was used as a source of NTE. It was prepared as described [29] and had a specifi c activity of about 15 nmoles/min for PV per mg of protein. The model diethylphosphate OP inhibitor, (C2H5O)2P(O)OCH(CF3)2, diethyl

Page 73: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

71

1,1,1,3,3,3-hexafl uoropropan-2-yl phosphate (DEHFPP), was synthesyzed in the Institute of Physiologically Active Compounds, RAS (Chernogolovka, Russia) according to [30] . All other chemicals were of analytical grade and were used without further purifi cation. Aqueous solutions were prepared using Milli Q (Millipore) deionized water.

Blood sampling and preparation of blood for analysisBlood samples from human and mouse subjects were obtained according to protocols

approved by the Insitute of Physiologically Active Compounds, RAS (Chernogolovka, Russia). Blood from white outbred male mice was obtained following sacrifi ce by decapitation under CO2-induced anesthesia. Trunk blood from each animal was collected immediately in glass vials containing citrate (3.8% w/v sodium citrate; 0.2 ml citrate/ml blood). Blood from 4 anonymous human (female) donors stabilized with citrate (0.2 ml citrate/ml blood) was obtained by venipuncture. All blood samples were aliquoted, frozen in liquid nitrogen, and stored at -70 °C before use. For whole blood homogenates, thawed blood samples were diluted 1/10 (v/v) in the appropriate diluting buffer (see below for buffer for each enzyme), homogenized (Potter glass-tefl on homogenizer, 10 times), and the necessary amount of homogenate was used for further analysis. For subsequent steps, homogenates were stored refrigerated at +4 °С except for incubations as noted below.

In vivo experimentsIn vivo experiments were carried out according to protocols for the use and care

of laboratory animals approved by the Insitute of Physiologically Active Compounds, RAS (Chernogolovka, Russia). DEHFPP was dissolved in Me2SO and administrated intraperitoneally into white outbred male mice (25-30 g) (about 0.1 ml/mouse) at doses from 1 to 200 mg/kg to groups of 4-6 mice per dose. Control animals received Me2SO only. At 1 h after administration, mice were decapitated and blood was collected immediately as described above, then transferred to Eppendorf tubes, frozen in liquid nitrogen, and stored at -70 C before the assay. Activity of CaE in blood samples was measured electrochemically and spectrophotometrically as described below. CaE activity in blood from mice treated with DEHFPP was compared to activity in blood from animals treated with Me2SO only (control) to calculate percent inhibition relative to control.

Design and preparation of tyrosinase-based biosensorsTyrosinase biosensors based on GR substrate were fabricated as previously described

[24-26, 31]. Briefl y, the tops of graphite rods were polished to a smooth shiny surface by means of sandpaper with progressively decreasing grain size and then washed in deionized water. The rods were then immersed in 0.5% (w/v) PDDA solution in 0.05 M sodium phosphate, pH 7.0, for 10 min. After rinsing thoroughly for 1-2 min with deionized water and drying in the air stream, the rods were dipped into 1Ч10-4 M tyrosinase solution prepared in 0.05 M sodium phosphate, pH 7.0, for 10 min followed by rinsing thoroughly for 1-2 min with deionized water. To stablilize the sensors by cross-linking with GA, graphite sensors coated with PDDA/ tyrosinase fi lm were dipped in aqueous 1% (w/v) solution of GA for 60 min followed by rinsing thoroughly for 1-2 min with deionized water, after which the sensors were stored under air at +4є.

The tyrosinase biosensors based on SPE substrate were fabricated as follows: bare SPE were produced on poly(vinyl chloride) substrates of 0.2 mm thickness by means

Page 74: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

72

of conductive graphite paste (Gwent, UK) screen-printed by a semiautomated machine (model WSC-160B, Winon, China) with a 200 mesh screen stencil. The polymer was adsorbed onto SPEs by covering the working area (0.049 mm2) of each SPE by a 10 μl drop of the 5 g/L solution of PDDA in 0.05 M sodium phosphate, pH 7.0, followed by 50 min adsorption. After that time, the substrate was rinsed with Milli-Q water and dried using a stream of air. The enzyme was adsorbed in a similar way for 10 min from a 1Ч10-4 M tyrosinase solution prepared in 0.05 M sodium phosphate, pH 7.0, followed by rinsing with Milli-Q water and drying with a stream of air. After that the biosensors were stored at +4єC until further use.

The design and principle of measurement of the PDDA/ tyrosinase/GA biosensors are shown in Fig. 1.

Figure 1. Design of (PDDA/Tyrosinase/GA) biosensor and principle of phenol assay.

Electrochemical measurements of phenol

All the electrochemical measurements were performed in 0.05 M sodium phosphate buffer with 0.1 M NaCl (pH 7.0) in a two-electrode electrochemical cell (V = 1 ml) under water-saturated air conditions. GR/PDDA/Tyrosinase/GA or SPE/PDDA/Tyrosinase biosensors served as a working electrode, and counter and reference electrodes were Ag/AgCl. The cell was monitored by a IPC-2000 potentiostat, designed in the Institute of Physical Chemistry, RAS (Moscow, Russia). The amperometric current response was recorded at -150 mV versus the Ag/AgCl reference electrode. The activity of tyrosinase-based sensors was determined as the value of a steady-state baseline current change (the difference between average values of steady-state baseline current before and after phenol addition).

CaE assayThe activity of CaE was assayed in 0.1 M sodium phosphate, pH 8.0, by measurement

of phenol released after a 5-min incubation of enzyme with 1 mM phenyl acetate at room temperature. In order to discriminate CaE activity, a 10-min preincubation with inhibitors of PON1/arylesterase (2 mM EDTA) and cholinesterases (40 μM eserine) was used [32-35].

PON1 assayThe activity of PON1 was assayed in 0.1 M Tris, pH 8.0, by measurement of phenol

released after enzymatic hydrolysis of 2 mM phenyl acetate at room temperature. In order

-

Gra

phite

+

+

+

+

+

+

++

+

+

--

-

-- E

--

- E

---

-- E

--

-- E

--

-

-- E

+ Phenol

Quinone

2H2OO2

Gra

phite

-

E

Catechole-

--

-

-- E

+

+

+

+Poly(dimethyldiallyl-ammonium chloride) Tyrosinase

Glutaraldehydecrosslinks

-

Gra

phite

+

+

+

+

+

+

++

+

+

--

-

-- E

--

- E

---

-- E

--

-- E

--

-

-- E

+

Gra

phite

+

+

+

+

+

+

++

+

+

--

-

-- E

--

- E

---

-- E

--

-- E

--

-

-- E

+ Phenol

Quinone

2H2OO2

Gra

phite

-

E

Catechole-

Phenol

Quinone

2H2OO2

Gra

phite

-

E

Catechole-e-

--

-

-- E

--

-

-- E

+

+

+

+Poly(dimethyldiallyl-ammonium chloride) Tyrosinase

Glutaraldehydecrosslinks+

+

+

+

+

+

+

+Poly(dimethyldiallyl-ammonium chloride) Tyrosinase

Glutaraldehydecrosslinks

Page 75: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

73

to discriminate PON1 activity prior to substrate addition a pair of samples was preincubated for 10 min in 0.1 M Tris, pH 8.0 with 1мМ CaCl2 or in 0.1 M Tris, pH 8.0 with 2 mM of EDTA. PON1 activity was calculated as the difference in the amount of phenol released between samples in pair.

NTE assay NTE activity is operationally defi ned as the enzymatic hydrolysis of the optimal non-

physiological substrate, phenyl valerate (PV), that is resistant to inhibition by diethyl 4-nitrophenyl phosphate ( paraoxon, nonneuropathic OP) and sensitive to inhibition by N,N′-diisopropylphosphorodiamidic fl uoride (mipafox, neuropathic OP) under specifi ed conditions of preincubation with inhibitors and subsequent incubation with substrate [36]. If “B” is the phenyl valerate hydrolyzing activity preinhibited with paraoxon, and “C” is the residual activity after both paraoxon and mipafox preinhibition, then the NTE activity can be calculated as the difference between “B” and “C”. For blood NTE assay, the differential inhibition method of Johnson [36] was used with an electrochemical endpoint as described previously [16]. Briefl y, samples were incubated at 37 °C in 50 mM Tris-HCl/0.2 mM EDTA (pH 8.0 at 25 °C) with 50 μM paraoxon (sample B) or 50 μM paraoxon plus 250 μM mipafox for 20 min (sample C), followed by incubation with PV (fi nal apparent concentration 0.54 mM) for the next 40 min at 37°C. NTE activity was calculated as the difference in the amount of phenol released between samples B and C.

Electrochemical esterase activity determinationsElectrochemical determination of esterase activity was carried out after the enzymatic

reaction was stopped by addition of 100 μl of 1% (w/v) aqueous SDS. The released phenol was assayed amperometrically after 20- to 100-fold dilution of samples in 50 mM sodium phosphate with 100 mM NaCl, pH 7.0. The analytical signal was determined as the value of steady-state baseline current change (the difference between an average value of steady-state baseline current before and after analyte addition). Activity values were calculated using phenol calibration curves, obtained in the same conditions, and each was corrected for spontaneous hydrolysis of substrates, determined separately.

Spectrophotometric CaE and PON1 activity determinationSpectrophotometric determination of CaE activity was carried out with 1 mM phenyl

acetate by measuring appearance of phenol product at 270 nm (ε270 = 1310 M-1min-1) [37,38] using a Gilford-250 spectrophotometer in 3 ml 1-cm pathlength cuvettes at 25 °C. All data were automatically corrected for spontaneous hydrolysis of substrate measured at the same time in the reference cuvette.

Spectrophotometric determination of PON1 activity was carried out with 2 mM phenyl acetate by measuring appearance of phenol product at 270 nm (ε270 = 1310 M-1min-1) using a Shimadzu UV-1800 spectrophotometer in 3 ml 1-cm pathlength cuvettes at 25 °C. All data were automatically corrected for spontaneous hydrolysis of substrate measured at the same time in the reference cuvette.

Statistical analysesData are presented as mean ± SD, mean ± SE, or mean and 95% confi dence interval

(CI) (n ≥ 3 separate experiments), as indicated in each instance. Statistical analyses were

Page 76: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

74

carried out using GraphPad Prism 6.0a.130 for Mac OS X, GraphPad Software (San Diego, CA, USA) and OriginPro 8.6, OriginLab (Northampton, MA, USA). The level of statistical signifi cance of differences between means was set at p< 0.05.

3. Results and discussion

Analytical characteristics of the tyrosinase biosensorsHerein described tyrosinase biosensors were fabricated using polished graphite rods

(GR) ot screen-printed electrodes (SPE) as initial substrates to be modifi ed via LBL technique fi rstly by poly(dimethyldiallylammonium chloride) (PDDA) and secondly by tyrosinase. The resultunt SPE/PDDA/Tyrosinase biosensor was use as it is, wherears functionality of GR/PDDA/Tyrosinase was additionally preserved by glutaraldehyde (GA) crosslinking.

The change in steady-state baseline current as a function of phenol concentration (calibration curve for phenol) was examined for both types of tyrosinase biosensors and the analytival parameters of phenol detection were derived from respective calibration curves and were summarized in Table 1. The operational stability of PDDA/Tyrosinase/GA biosensors was examined for >10 repeated measurements (typically for 25-30 repeated measurements) of a standard analyte concentration (10 μM of phenol). It can be characterized quantitatively as a percentage of the analytical signal change per a single measurement and can be calculated according to the formula: ΔI = 100% × tgI/I1, where tgI is the slope of the dependence of the analytical signal on the number of repeated measurements normalized to the initial analytical signal I1 and given in percentage (Table 1).

Table 1. Comparative analytical characteristics of tyrosinase biosensors

Biosensor type GR/PDDA/Tyrosinase/GA [31] SPE/PDDA/Tyrosinase

Linearity, M 1×10-8– 1×10-5 25×10-9 – 1×10-5

Detection limit for phenol(S/N=3), nM 6 12

Sensitivity, mA/(M×cm2) 600 440

Repetabilty (relative SD for 1Ч10-5 M phenol, n = 10), %

6 2.5

Operational stability, ∆I, % -0.9 ± 0.4 -0.5±0.2

Storage stability, 100% after 20 days of storage at +4°C in a dry state

75% after 30 days of storage at +4°C in a dry state

Reprodusibility of biosensor preparation 15 10

These results demonstrate that fast and reliable detection of phenol in the nanomolar range is possible by using both type of tyrosinase biosensors. These analytical characteristics indicated that both type biosensors were potentially suitable for monitoring enzymatic

Page 77: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

75

processes that proceed with release of phenol. Preliminary experiments with a range of dilutions (20- to 200-fold) of whole human or mouse blood homogenates showed that neither the response of the biosensor to phenol nor its recovery time for subsequent measurements were affected by the presence of blood if it was diluted at least 100-fold.

The infl uence of potentially interfering agents found in blood (including glucose, L-ascorbic acid, adrenaline, uric acid and L-tyrosine) on biosensor phenol detection were also examined [31]. The concentrations of interfering agents were selected to be close to their normal levels in blood plasma. Additional dilutions (1/10 and 1/100, v/v) were tested as well. Inspite on some interference that was observed at the maximum levels of interfering agents; their effects were neglidible at 1/100 sample dilution.

Therefore, further studies were conducted to examine responses of the biosensors to activities of blood esterases.

Matrix effect of blood on biosensor assay of CaE, PON1, and NTE activitiesBefore attempting assay of CaE, PON1, and NTE activities in blood samples, the

electrochemical detection of commercial porcine liver CaE, human recombinant PON1, and hen brain NTE activity by the tyrosinase biosensors was tested in the absence and presence of blood. Serial dilutions of each esterase preparation were made and the activity of each sample was measured in the absence and presence of an aliquot of diluted blood homogenate. Preliminary experiments with a range of dilutions (20- to 200-fold) of whole human or mouse blood homogenates showed that neither the response of the biosensor to phenol nor its recovery time for subsequent measurements were affected by the presence of blood if it was diluted at least 100-fold. The experiments showed that only an upward parallel shift of the calibration curve occurred in the presence of blood for CaE (Fig. 3A), PON1 (Fig. 3B) or NTE (Fig. 4). These results confi rm the absence of a matrix effect of blood on the biosensor assay for CaE, PON1, and NTE activity. The upward shift in each enzyme calibration curve is due to the intrinsic activities of CaE, PON1, and NTE in whole homogenized blood, as discussed further below.

Figure 3. Amperometric assay of porcine liver CaE activity (A) and human recombinant PON1 activity (B) depending on the enzyme concentration obtained in the absence (lower lines, open circles) or presence (upper lines, closed circles) of human blood homogenate. Conditions for CaE: 1/500 v/v blood sample pretreated with 2 mM EDTA and 40 μM eserine for 10 min at room temperature to inhibit PON1 and cholinesterases, respectively. Conditions for PON1: 1/500 v/v blood sample pretreated with 100 μM paraoxon for 20 min at room temperature to inhibit CaE and cholinesterases. Samples were then incubated with phenyl acetate at room temperature at specifi ed conditions and assayed for phenol after 50ч100 fold dilution. Sensor response (Y) = relative to response at standard [ phenol] = 10 μM. Data are mean ± SD. The change in intercepts caused by addition of blood was due to intrinsic CaE/ PON1 activities in blood.

0 20 40 600.0

0.4

0.8

1.2

Sesn

or re

spon

se (r

elat

ive

units

)

[CaE], mU/ml

A

0 20 40 60

0.0

0.2

0.4

0.6

0.8

[PON1], mU/ml

Sens

or re

spon

se (r

elat

ive

units

)

B

Page 78: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

76

Figure. 4. Amperometric assay of hen brain PVase (main graph) and NTE (inset) activities depending on the enzyme concentration obtained in the absence (lower lines, open circles) or presence (upper lines, closed circles) of human blood. Open squares: PVase activity of partially purifi ed paraoxon pretreated hen brain NTE preparation (activity “B”, absence of blood). Closed squares: same as open squares, but in the presence of 1/200 (v/v) diluted human blood homogenate preincubated with 50 μM paraoxon at 37°C for 20 min (activity “B”, presence of blood). Open circles: PVase activity of partially purifi ed paraoxon pretreated hen brain NTE preparation preincubated with 250 μM mipafox at 37°C for 20 min (activity “C”, absence of blood). Closed circles: same as open circles, but in the presence of 1/200 (v/v) diluted human blood homogenate pretincubated with 50 μM paraoxon and 250 μM mipafox at 37°C for 20 min. PVase was measured after 40 min incubation of samples with 0.56 mM PV at 37°C. Phenol was assayed amperometrically after 20-fold dilution of samples. Inset, open triangles: NTE activity (activity “B-C”) in the absence of 1/200 (v/v) diluted human blood homogenate; Inset, closed triangles: NTE activity (activity “B-C”) in the presence of 1/200 (v/v) diluted human blood homogenate. Sensor responses are given as electrochemical signal normalized to response to standard phenol concentration (10 μM). Data are mean ± SD. The change in Y-intercepts caused by addition of blood was due to intrinsic PVase activity in blood.

Biosensor assay of CaE, PON1, and NTE in human and mouse blood

Activity of CaE in blood homogenates from humans and mice were measured in paired samples using the (GR/PDDA/ tyrosinase/GA) biosensor and the spectrophotometric method. Activity of PON1 in blood homogenates from humans were measured in paired samples using the (SPE/PDDA/ tyrosinase) biosensor and the spectrophotometric method. In addition, NTE activity was measured in aliquots from the paired samples using only the biosensor method, because the activity of this enzyme cannot be measured spectrophotometrically in homogenates of whole blood. The results in Table 2 show that there is good agreement between the biosensor and spectrophotometric results for CaE for both human and mouse blood and PON1 activities for human blood. In addition, these values are in good agreement with previously reported electrochemical measurements in hemolyzed blood samples [35].

0 10 20 30 40 500.0

0.2

0.4

0.6

0.8

10 20 30 40 50

0.1

0.2

0.3

0.4

[NTE], μg/mlSe

nsor

resp

onse

(rel

ativ

e un

its)

[PVase], mg/ml

B

C

(B-C)

Page 79: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

77

Table 2. CaE, PON1, and NTE activities in human or mouse blood homogenates.

Subject and Sample Number

CaE(μmol/min/ml whole blood)

PON1(μmol/min/ml whole blood)

NTE(nmol/min/ml whole blood)

spectrophot.(n=4)

biosensor(n=4)

spectrophot.(n=8)

biosensor(n=8)

biosensor(n=4)

Human blood

12

6. 345678

0.560.140.100.13

----

0.410.180.120.14

----

17.125.227.041.713.422.327.724.5

18.129.431.747.713.719.727.423.6

41.450.430.625.2

----

Mean±SEM 0.23 ± 0.11 0.22 ± 0.07 26.4 ± 10.5 24.9 ± 8.4 36.9 ± 5.62Mouse blood

1234

6.257.245.805.43

4.267.796.645.46

----

----

9.119.817.614.6

Mean±SEM 6.18 ± 0.39 6.04 ± 0.76 - - 15.3 ± 2.32

With respect to other published data on blood CaE levels, data could be found only for plasma. Apparent CaE activity in human plasma is very low; when determined with 1-naphthyl acetate, it is reported to be 0.019 ± 0.001 μmol/min/ml [37]. Using nondenaturing gradient gel electrophoresis and staining for esterase activity, CaE was undetectable in human plasma [39]. However, CaE is found in monocytes [40], which are the likely source of the activity detected in whole blood homogenates in our studies. Our results also show that the apparent CaE activity of rodent (mouse) blood is higher than that of humans, which is in accord with qualitatively high levels detected on gels in mouse plasma [39] and quantifi ed as 3.52 ± 0.15 μmol/min/ml in rat plasma [37].

Very different published data on arylesterase activities of PON1 can be found in the literature, e.g., PON1 activity in human serum reported by [41] was on the level of 113 ± 14 μmol/min/ml, while the same was in the range of 80-240 μmol/min/ml according to [42]. According to our former data arylesterase activity of PON1 in human plasma and in hemolysed human blood was found to be 63.5±9.70 (N=5) and 34.6±4.1 μmol/min/ml, respectively [34]. Taking into account a volume fraction of plasma in whole blood of about 50% as well as a large variability of PON1 arylesterse activity in plasma levels among individuals we can consider our results as quite reasonable.

Spectrophotometric technique does not allow determining NTE activity in whole blood . As we demonstrated earlier, NTE assay in whole blood is possible only using a phenol biosensor [16]. Activity of NTE in human blood determined in this work with the new LBL biosensor (Table 2) was close to that obtained earlier (0.19 ± 0.02 nmoles/min per mg of protein; equivalent to 32.3 ± 3.6 nmol/min/ml blood ) [16]. We also found that the mean value for mouse blood NTE activity was 41.5% of that measured in human blood ; similarly, the mean value for NTE activity in mouse platelets was previously determined by Husain to be 42.8% of that in human platelets [43].

Page 80: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

78

Biosensor assay of mouse blood CaE following its inhibition in vivoA biomonitoring assessment of the tyrosinase biosensor was carried out by comparing

electrochemical and spectrophotometric measurements of blood CaE activity changes ex vivo after dosing mice in vivo with DEHFPP. This compound was previously found to be a relatively potent OP inhibitor of CaE in vitro(ki = 1.20×105 M-1min-1) [30]. As shown in Fig. 5, blood CaE activity was inhibited in a dose-dependent manner by DEHFPP treatment, yielding effective dose 50% (ED50) values [mean (95% CI) (n)] of 25.5 (23.2, 28.0) mg/kg (6) and 21.1 (18.9, 23.5) mg/kg (4) for spectrophotometric and electrochemical assays, respectively. Although the ED50 values were signifi cantly different from each other (p< 0.007), an excellent correlation between biosensor and spectrophotometric measurements was found (r = 0.99) (Fig. 5, inset), indicating that the differences between the two methods are systematic and providing validation of the biosensor assay.

Figure 5. Main graph: dose-related inhibition of CaE activity in mouse blood 1h after dosing with DEHFPP (structure shown on graph). CaE activity in whole blood was determined by spectrophotometric (fi lled circles) and biosensor (circles) methods and presented as % inhibition of control activity. Data are mean ± SE, n = 4-6. Control CaE activity (mean ± SE) = 6.02 ± 0.27 μmol/min/ml blood (n =20) and 6.04 ±1.52 (n =5) for spectrophotometric and biosensor methods, respectively. ED50 for CaE inhibition, mean (95% CI) = 25.5 (23.2, 28.0) mg/kg (n = 6) and 21.1 (18.9, 23.5) mg/kg (n = 4) for spectrophotometric and electrochemical assays, respectively. Inset, fi lled diamonds: correlation of blood CaE % inhibition obtained with biosensor and spectrophotometric methods; mean ± SE, r= 0.99, p<0.0001, n = 7.

The LBL technique was successfully applied to deposit a PDDA/ tyrosinase/GA fi lm on a graphite substrates to produce a highly sensitive tyrosinase-based biosensor for nanomolar phenol detection. The biosensor exhibited good sensitivity, stability, and analytical characteristics. Cross-linking by glutaraldehyde imparted improved operational stability without decreasing tyrosinase activity. The biosensor enabled rapid and precise assays of blood esterases based on their hydrolysis of phenyl esters to the phenol analyte. Quantitative determination of CaE and PON1 activities in blood samples showed good agreement between biosensor and spectrophotometric assays for these enzymes. Biosensor assay of NTE activity in human and mouse blood gave values in close agreement with previous determinations of human blood NTE using an earlier biosensor version, and the

Page 81: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

79

ratio of mouse to human blood NTE activities was essentially identical to the previously reported ratio of activities for NTE in mouse and human platelets. Biosensor determination of changes in blood CaE ex vivo after dosing mice in vivo with an OP inhibitor of the enzyme yielded values highly correlated with those from a spectrophotometric method, thus validating the use of the biosensors for biomonitoring applications. The application of the biosensors could be expanded to include other enzymes that could use phenyl esters as substrates, so much so the new biosensor setups are already developed with further improvement in their activity and stability [44].

AcknowledgmentsThe research was supported by ISTC 3130; RFBR 07-04-12235, 11-03-00712-a; and

NATO SfP 984082.

References

[1] D. Puig, D. Barcelo, Determination of phenolic compounds in water and waste water, Trends Anal. Chem. 15 (1996), 362-375.

[2] P.M. Potter, R.M. Wadkins, Carboxylesterases - Detoxifying Enzymes and Targets for Drug Therapy, Curr. Med. Chem. 13 (2006), 1045-1054.

[3] M.R. Redinbo, S. Bencharit, P.M. Potter, Human carboxylesterase 1: from drug metabolism to drug discovery, Biochem. Soc. Trans. 31 (2003), 620–624.

[4] T. Satoh, M. Hosokawa, The mammalian carboxylesterases: from molecules to functions, Annu. Rev. Pharmacol. Toxicol. 38 (1998), 257–288.

[5] M.K. Ross, J.A. Crow, Human carboxylesterases and their role in xenobiotic and endobiotic metabolism, J. Biochem. Mol. Toxicol. 21 (2007), 187-96.

[6] M. Jokanovic, Current understanding of the mechanisms involved in metabolic detoxifi cation of warfare nerve agents, Toxicol. Lett. 188 (2009), 1-10.

[7] G.F. Makhaeva, E.V. Rudakova, N.P. Boltneva, L.V. Sigolaeva, A.V. Eremenko, I.N. Kurochkin, R.J. Richardson, Blood esterases as a complex biomarker for exposure to organophosphorus compounds. In: Counteraction to Chemical and Biological Terrorism in the East Europe Countries, NATO for Peace and Security Series, Dishovsky C. and Pivovarov A., (Eds.), Springer, 2009, 177-194.

[8] C.E. Wheelock, B.M. Phillips, B.S. Anderson, J.L. Miller, M.J. Miller, B.D. Hammock, Applications of carboxylesterase activity in environmental monitoring and toxicity identifi cation evaluations (TIEs), Rev. Environ. Contam. Toxicol. 195 (2008), 117-78.

[9] L.G. Costa, T.B. Cole, A. Vitalone, C.E. Furlong, Measurement of paraoxonase ( PON1) status as a potential biomarker of susceptibility to organophosphate toxicity, Clin. Chim. Acta 352 (2005), 37–47.

[10] B. Li, M. Sedlacek, I. Manoharan, R. Boopathy, E.G. Duysen, P. Masson, O. Lockridge, Butyrylcholinesterase, paraoxonase, and albumin esterase, but not carboxylesterase, are present in human plasma, Biochem. Pharmacol. 70(11) (2005), 1673-1684.

[11] L.G. Costa, W.F. Li, R. Richter, D.M. Shid, A.J. Lusis, C.E. Furlong In: Paraoxonase ( PON1) in health and disease, L.G. Costa and C.E. Furlong (Eds.) Kluver, Norwell, 2002, 165-183.

[12] M.K. Johnson, The target for initiation of delayed neurotoxicity by organophosphorus esters: biochemical studies and toxicological applications. In: Reviews in Biochemical Toxicology, E. Hodgson, J.R. Bend, and R.M. Philpot (Eds.), Amsterdam, Elsevier, 1982, 4, 141-212.

[13] G.F. Makhaeva, L.V. Sigolaeva, L.V. Zhuravleva, A.V. Eremenko, I.N. Kurochkin, V.V. Malygin, R.J. Richardson, Biosensor detection of Neuropathy Target Esterase in whole blood as a biomarker of exposure to neuropathic organophosphorus compounds, J. Toxicol. Environ. Health, Part A 66 (2003), 599-610.

[14] L.G. Costa, Biomarker research in neurotoxicology: the role of mechanistic studies to bridge the gap between the laboratory and epidemiological investigations, Environ. Health Persp. (Suppl. 1) 104 (1996). 55–67.

[15] G.F. Makhaeva, V.V. Malygin, N.N. Strakhova, L.V. Sigolaeva, L.G. Sokolovskaya, A.V. Eremenko, I.N. Kurochkin, R.J. Richardson, Biosensor assay of neuropathy target esterase in whole blood as a new approach to OPIDN risk assessment: review of progress, Hum. Exp. Toxicol. 26 (2007), 273-282.

Page 82: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

80

[16] L.V. Sigolaeva, A. Makower, A.V. Eremenko, G.F. Makhaeva, V.V. Malygin, I.N. Kurochkin, F. Scheller, Bioelectrochemical analysis of neuropathy target esterase activity in blood , Anal. Biochem. 290 (2001), 1-9.

[17] S. Liu, J. Yu, H. Ju, Renewable phenol biosensor based on a tyrosinase-colloidal gold modifi ed carbon paste electrode, J. Electroanal. Chem. 540 (2003), 61-67.

[18] D. Shan, Ch. Mousty, S. Cosnier, Sh. Mu, A composite poly azure B-clay-enzyme sensor for the mediated electrochemical determination of phenols, J. Electroanal. Chem. 537, (2002), 103-109.

[19] G. Wang, J.-J. Xu, L.-H. Ye, J.-J. Zhu, H.-Y. Chen, Highly sensitive sensos based on the immobilization of tyrosinase in chitosan, Bioelectrochemistry 57 (2002), 33-38.

[20] Y. Qu, M. Ma, Zh. Wang, G. Zhan, B. Li, X. Wang, H. Fang, H. Zhang, Ch. Li, Sensitive amperometric biosensor for phenolic compounds based on graphene-silk peptide/ tyrosinase composite nanointerface, Biosens. Bioelectron.44 (2013), 85-88.

[21] Ch.-J. Yuan, Ch.-L. Wang, T.Y. Wu, K.-C. Hwang, W.-C. Chao, Fabrication of a carbon fi ber paper as the electrode and its application toward developing a sensitive unmediated amperometric biosensor, Biosens. Bioelectron. 26 (2011), 2858-2863.

[22] L. Yang, H. Xiong, X. Zhang, Sh. Wang, A novel tyrosinase biosensor based on chitosan-carbon-coated nickel nanocomposite fi lm, Bioelectrochemistry 84 (2012), 44-48.

[23] B.C. Janegitz, R.A. Medeiros, R.C. Rocha-Filho, O. Fatibello-Filho, Direct electrochemistry of tyrosinase and biosensing for phenol based on gold nanoparticles electrodeposited on a boron-doped diamond electrode, Diamond and Related Materials 25 (2012), 128-133.

[24] G.V. Dubacheva, L.G. Sokolovskaya, L.V. Sigolaeva, A.A. Yaroslavov, A.V. Eremenko, I.N. Kurochkin, Tyrosinase biosensors based on nanostructured polyelectrolyte fi lms, Sensornie systemi (Russian)20 (2006), 326-333.

[25] G.V. Dubacheva, M.V. Porus, L.G. Sokolovskaya, L.V. Sigolaeva, D.V. Pergushov, A.A. Yaroslavov, I.N. Kurochkin, S.D. Vartfolomeev, Nanostructured polyelectrolyte fi lms as a base for the creation of high sensitive tyrosinase biosensors, Nanotechnologies in Russia (Russian) 2 (2007), 154-159.

[26] G.V. Dubacheva, M.V. Porus, L.V. Sigolaeva, D.V. Pergushov, D.R. Tur, V.S. Papkov, A.B. Zezin, A.A. Yaroslavov, A.V. Eremenko, I.N. Kurochkin, S.D. Varfolomeev, Nanostructured polyelectrolyte fi lms for engineering highly sensitive tyrosinase biosensors: specifi cs of enzyme-polyelectrolyte structures, Nanotechnologies in Russia (Russian) 3 (2008), 219–225.

[27] M.V. Porus, G.V. Dubacheva, L.V. Sigolaeva, A.V. Eremenko, I.N. Kurochkin, Determination of cholinesterase activities by using bielectrode sensor system, Sensornie Sistemi (Russian) 22 (2008), 88-95.

[28] M.S. Gromova, L.V. Sigolaeva, M.A. Fastovets, E.G. Evtushenko, I.A. Babin, D.V. Pergushov, S.V. Amitonov, A.V. Eremenko, I.N. Kurochkin, Improved adsorption of choline oxidase on a polyelectrolyte LBL fi lm in the presence of iodide anion, Soft Matter 7 (2011), 7404-7409.

[29] G.F. Makhaeva, V.V. Malygin, A stable preparation of hen brain neuropathy target esterase for rapid biochemicqal assessment of neurotoxic potential of organophosphates, Chem. Biol. Interact. 119-120 (1999), 551-557.

[30] G.F. Makhaeva, A.Y.Aksinenko, V.B.Sokolov, O.G. Serebryakova, R.J. Richardson, Synthesis of organophosphates with fl uorine-containing leaving groups as serine esterase inhibitors with potential for Alzheimer disease therapeutics, Bioorg. Med. Chem. Lett. 19 (2009), 5528-5530.

[31] L.V. Sigolaeva, G.V. Dubacheva, M.V. Porus, E.A. Eremenko, E.A. Rudakova, G.F. Makhaeva, R.J. Richardson, I.N. Kurochkin, A layer-by-layer tyrosinase biosensor for assay of carboxylesterase and neuropathy target esterase activities in blood , Anal. Meth. 5(16) (2013), 3872-3879.

[32] S.H. Sterri, B.A. Johnsen, F. Fonnum, A radiochemical assay method for carboxylesterase, and comparison of enzyme activity towards the substrates methyl [1-14C] butyrate and 4-nitrophenyl butyrate, Biochem. Pharmacol. 34 (1985), 2779-2785.

[33] S.M. Chanda, S.R. Mortensen, V.C. Moser, S. Padilla, Tissue-specifi c effects of chlorpyrifos on carboxylesterase and cholinesterase activity in adult rats: an in vitro and in vivo comparison, Fundam. Appl. Toxicol. 38 (1997), 148-57.

[34] E.V. Rudakova, N.P. Boltneva, G.F. Makhaeva, Comparative analysis of esterase status of human and rat blood , Bull. Exp. Biol. Med. 152 (2011), 73-75.

[35] L.V. Sigolaeva, G.F. Makhaeva, E.V. Rudakova, N.P. Boltneva, M.V. Porus, G.V. Dubacheva, A.V. Eremenko, I.N. Kurochkin, R.J. Richardson, Biosensor analysis of blood esterases for organophosphorus compounds exposure assessment: Approaches to simultaneous determination of several esterases, Chem. Biol. Interact. 187 (2010), 312-317.

Page 83: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

81

[36] M.K. Johnson, Improved assay of neurotoxic esterase for screening organophosphates for delayed neurotoxicity potential, Arch. Toxicol. 67 (1977), 113–115.

[37] C. De Vriese, F. Gregoire, R. Lema-Kisoka, M. Waelbroeck, P. Robberecht, C. Delporte, Ghrelin degradation by serum and tissue homogenates: identifi cation of the cleavage sites, Endocrinology 145 (2004), 4997-5005.

[38] T.L. Huang, T. Shiotsuki, T. Uematsu, B. Borhan, Q.X. Li and B.D. Hammock, Structure-activity relationships for substrates and inhibitors of mammalian liver microsomal carboxylesterases, Pharm. Res. 13 (1996), 1495–1500.

[39] B. Li, M. Sedlacek, I. Manoharan, R. Boopathy, E.G. Duysen, P. Masson and O. Lockridge, Butyrylcholinesterase, paraoxonase, and albumin esterase, but not carboxylesterase, are present in human plasma, Biochem. Pharmacol. 70 (2005), 1673-1684.

[40] J.A. Crow, K.L. Herring, S. Xie, A. Borazjani, P.M. Potter and M.K. Ross, Inhibition of carboxylesterase 1 is associated with cholesteryl ester retention in human THP-1 monocyte/macrophages, Biochim. Biophys. Acta. 1781 (2008), 643-654.

[41] M. Rosenblat, R. Karry, M. Aviram, Paraoxonase 1 ( PON1) is a more potent antioxidant and stimulant of macrophage cholesterol effl ux, when present in HDL than in lipoprotein-defi cient serum: Relevance to diabetes, Atherosclerosis 187 (2006) 74-81.

[42] C.E. Furlong, R.J. Richter, S.L. Seidel, A.G. Motulsky, Role of genetic polymorphism of human plasma paraoxonase/arylesterase in hydrolysis of the insecticide metabolites chlorpyrifos oxon and paraoxon. Am. J. Hum. Genet. 43(3) (1988),230-238.

[43] K. Husain, Phenyl valerate and choline ester hydrolases in the platelets of human, hen, rat and mouse, Hum. Exp. Toxicol. 13 (1994), 157-159.

[44] L.V. Sigolaeva, D.V. Pergushov, C.V. Synatschke, A. Wolf, I. Dewald, I.N. Kurochkin, A. Feryc, A.H.E. Mьller, Co-assemblies of micelle-forming diblock copolymers and enzymes on graphite substrate for an improved design of biosensor systems, Soft Matter 9 (2013), 2858–2868.

Page 84: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

82

Chapter 6

Thick Film Thiol Sensors for Cholinesterases Assay

Arkadiy EREMENKOa,b1, Ekaterina DONTSOVAa, Artem NAZAROVa, Ilya KUROCKINa

aM.V. Lomonosov Moscow State University, Chemical Faculty, Leninskie Gory 1/3, Moscow, RUSSIA

bN.M. Emanuel Institute of Biochemical Physics of the RAS, Kosygina str. 4, Moscow, RUSSIA

Abstract. A special design of thick fi lm sensors and electrochemical detector for cholinesterases assay and their inhibitors in aqueous samples has been developed. For this assay, thiol sensitive sensors based on screen printed graphite electrode modifi ed with nanoparticles of manganese dioxide were used. High sensitivity of manganese dioxide modifi ed thick fi lm sensors towards thiocholine and therefore low detection limit of butyrylcholinesterase (1 pM) enabled their use for subnanomolar detection of an organophosphate pesticide diazinon, and other irreversible inhibitors of BChE. These developed electrochemical thick fi lm sensors may be well suited for the analysis of choline esterases in biological fl uids.

Keywords. Thick fi lm thiol sensitive sensor, manganese dioxide, Gypsophila trichotoma, isolated rat hepatocytes, cytoprotection, antioxidant

IntroductionCholinesterases have important physiological functions: acetylcholinesterase (AChE)

is involved in neural signal transduction catalyzing hydrolysis of neurotransmitter acetylcholine [1,2], butyrylcholinesterase (BChE) act as stoichiometric scavenger of organophosphates [3,4]. These enzymes can be inhibited by pesticides. Currently, activities of blood esterases and concentrations of their inhibitors are determined by using techniques for registration of ‘exogenous’ choline generated by biocatalytic hydrolysis of choline esters. The application of modern electrochemical sensors is a good alternative to routinely used methods due to high sensitivity achieved in recent years.

This paper describes the thick fi lm graphite sensors based on gamma-phase-MnO2 as thiols oxidation mediating reagent. The proposed approach for the sensors manufacturing is technological and highly productive. At the same time, it achieves an excellent sensitive measurement of thiocholine, butyrylcholinesterase and its inhibitors fortheirfurtherapplicationinenvironmental and clinical monitoring.

1 Corresponding author: Arkadiy Eremenko, Kosygina str. 4, 119334Moscow, Russia; E-mail: [email protected]

Page 85: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

83

1. Materials and methods

2. Manganese dioxide synthesis and thiocholine productionManganese dioxide was obtained in a comproportionation reaction with manganese

acetate by addition of 0.5 ml of 0.25 mM KMnO4 aqueous solution to 0.5 ml of0.375 mMMn(CH3COO)2·4H2O at room temperature yielding a stable hydrosol [5].

Thiocholine was obtained according to the method described in [6] modifi ed at the stage of enzymatic hydrolysis of propionylthiocholine. Propionylthiocholine chloride (21.2 mg) was added to a 1-ml aliquot of 0.4 mg/ml BChE solution in phosphate buffer (pH 8) and incubated during 1 h. Under these conditions, thiocholine fi nal concentration was 0.1 M. The Ellman method was used to control thiocholine concentration in a parallel experiment. An aliquot of thiocholine solution was diluted 400 times and 100 μl of the diluted solution was introduced into a well containing 100 μl Ellman reagent (6 mM DTNB). Optical density was read at 405 nm. Thiocholine concentration was calculated using coeffi cient of extinction of 5-thionitrobenzoic acid at 405 nm (ε405 = 13.3 ml.μmol-1cm-1).

3. Fabrication of thick fi lm graphite sensors and formation of the mediator layer Screen-printed electrodes were fabricated using a WSC160B semi-automated screen-

printing apparatus (Winon, Taiwan) and graphite paste (Coates Screen, Germany) on a 0.2-mm thick polyvinylchloride support. Then, the electrodes were heated at 60єС during 2 h. Diameter of the electrode working area was 2.5 mm.

A 10-μl drop of freshly prepared manganese dioxide hydrosol was applied on the surface of the screen-printed graphite electrode. The electrode was dried thoroughly at room temperature. The procedure was repeated several times when necessary, then the electrodes were washed with water, excess water was removed, and the electrode was dried again. Then, the electrodes were heated for 1 h at 60єС in a thermostat and stored at room temperature before use.

4. Measurements

5. VoltammetryCyclic voltammograms of the screen-printed electrodes modifi ed with manganese

dioxide were registered in the range of +250 to +750 mV in 50 mM HEPES buffer containing 30 mM KCl, pH 7.5, at the rate of 30 mV/s on an IPC-2000 potentiostat (Khronas, Russia) in a two-electrode cell (with Ag/AgCl reference electrode).

6. Amperometric measurementsMeasurements were performed in a 1-ml electrochemical cell with magnetic stirrer

in 50 mM HEPES buffer containing 30 mM KCl, pH 7.5. The cell contained an Ag/AgCl reference electrode and a working electrode. The working screen-printed electrode modifi ed with manganese dioxide was placed into the cell and the working potential of +450 mV (vs. Ag/AgCl) was applied to the electrode.

7. Butyrylcholinesterase (BChE) assayMeasurement of the BchE activity carried out in two ways. According to the fi rst

method, the activity was determined through electrochemical measurement of the

Page 86: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

84

initial rate (kinetic method) of thiocholine accumulation upon enzymatic hydrolysis of propionylthiocholine. Buffer solution (880 μl) together with 50 mM substrate solution (20 μl) were placed into the electrochemical cell and a chronoamperometric curve was recorded during 200 s. Spontaneous hydrolysis rate was determined by the slope of the curve at the starting point (tg α1). Then, 100 μl of the BChE solution were introduced in the cell and the linear current increase was recorded during 200 s. Total rate of spontaneous and enzymatic hydrolysis of the substrate was determined as theslope of the curve upon introduction of the enzyme to the cell (tgα2). Enzyme activity was calculated as difference between tgα2 and tg α1.

The second method consisted in measuring thiocholine accumulation for 10 min (end point method) enzymatic hydrolysis propionylthiocholine. From the incubation mixture aliquot was added to the electrochemical cell containing a thiol-sensitive sensor and steady-state current was measured.Analytical characteristics in this case considered the value of steady-state current, which is proportional to the concentration of accumulated thiocholine.

Inhibition analysis was demonstrated by the example of the organophosphorus pesticide diazinon. Diazinon, preliminarily oxidized with bromine water to its oxone form which is a more effi cient inhibitor of BChE, was used as an inhibitor. Bromine water was prepared by the addition of 4 μl bromine to 12.5 μl of0.5 M KBr. Diazinon solutions were oxidized during 3 min and then diluted with buffer solution so that the concentration was 10–8–10–10 M. Incubation mixture contained 180 μl of the inhibitor at the defi ned concentration and 20 μl of the 1 nM enzyme solution (10–10 М fi nal concentration). After a 10 min incubation of BChE with the inhibitor, enzyme activity was measured as described above two methods.

8. Results and DiscussionFigure 1 demonstrates the scheme of electrochemical determination of thiol compounds

using thick fi lm graphite sensor modifi ed with manganese dioxide by the example of thiocholine. Analytical response of the sensor is formed by the reduction of Mn(IV) to Mn(II/III) on the sensor in the presence of thiocholine followed by electrochemical oxidation of the mediator to Mn(IV) under positive potential.

Figure 1. Scheme of electrochemical analysis of thiocholine on thick fi lm graphite sensor modifi ed with MnO2

Figure 2 presents the cyclic voltammetric characteristics obtained using thick fi lm graphite sensors modifi ed with MnO2 in the absence (a) and in the presence (b) of 0.4 mM thiocholine. According to the cyclic voltammograms (Fig. 2a) γ-MnO2 was capable to high-rate reduction/oxidation processes on the electrode which is evidenced by the lower reduction

Page 87: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

85

and higher oxidation current values.The addition of thiocholine resulted in the sharp increase in catalytic current starting from the working potential values of +300–350 mV vs Ag/AgCl (Fig. 2b). The experiment demonstrates the cyclic electrocatalytic conversion of manganese dioxide in the presence of thiocholine described in the scheme of Figure 1. On the basis of the cyclic voltamogramms, the working potential value for thiocholine determination on electrodes modifi ed with manganese dioxide was chosen as +450 mV.

Figure 2. Cyclic voltammograms of the sensors modifi ed with γ-MnO2, obtained (a) in the absence and (b) in the presence of 0.4 mM thiocholine in 50 mM HEPES buffer containing 30 mM KCl, pH 7.5.

Scan rate 30 mV/s.

Stable hydrosol of γ-MnO2 was used further on to study analytical characteristics of the sensors for thiols determination. The values of detection limits, linear range, and sensitivity of the screen-printed electrodes modifi ed with γ-MnO2 with respect to thiocholine were studied. Sensitivity to thiocholine was 345 mA·cm2/M, the linear range for thiocholine was 5 · 10–7–1 · 10–5 М and the limit of detection, 6 · 10–8 М. The analytical characteristics of the sensors elaborated allow for the highly sensitive analysis of BChE and its inhibitors.

Determination of BChE activity was performed by “kinetic” and “end point” modes of measurement in an electrochemical cell with stirring. The values of detection limits, linear range, and sensitivity of the screen-printed electrodes modifi ed with γ-MnO2 with respect to thiocholine, cysteine, and glutathione are presented in Table 1.

The studied range of the enzyme concentrations was 1 · 10–12 – 5 · 10–11 М. Should be note, both modes of BchE assay show low detection limits for the enzyme. For comparison, a spectrophotometric Ellman’s method to measure enzyme concentration is more than two orders of magnitude 10-10M.

Table 1. Analytical characteristics thick fi lm sensors for BChE assay

Measurement mode Detection limit (M) Linear Range (M) Sensitivity

kinetic 1 · 10-12 1 · 10-12 – 5 · 10–11 8,16 nА/(nМ · s)

end point 2 · 10-12 1 · 10-12 – 5 · 10–11 630 nА/nМ

Page 88: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

86

It should be noted that both modes of BchE assay show low detection limits for the enzyme. For comparison, a spectrophotometric Ellman’s method to measure enzyme concentration is more than two orders of magnitude 10-10M.

Figure 4. Relative BChE activities in function of diazinon concentration for “kinetic” (A) and “end point” methods.

Data on diazinon determination using the developed thick fi lm thiol sensor are presented in Figure 4. Figure 4A and 4B demonstrate relative BChE activities in function of diazinon concentration obtained by “kinetic” and “end point” methods. Since confi dentialinterval value of BChE activity determination is 15% (as calculatedunder assumptionof 95%level of confi dencefor5measurements), diazinon limit of detection for both methods may be estimated as 6 · 10–10 М. Therefore, the developed thiol sensors allow for determination of cholinesterase irreversible inhibitors at the nanomolar range. Importantly, the analysis duration is less than 15-20 min. Also, the sensors should be noted for the fair operational stability. The response to thiocholine decreased by less than 0.2% per measurement allowing for multiple use of the developed electrodes in determination of thiols, BChE, and inhibitors in contrast to the electrodes modifi ed with platinum or other metal oxide mediators which are easily poisoned by sulfur in the course of the analysis and thus are mainly nonreusable.

0 2 4 6 8 100,0

0,2

0,4

0,6

0,8

1,0

E/E

0

Diazinon (nM)

A

0 2 4 6 8 100,0

0,2

0,4

0,6

0,8

1,0

E/E 0

Diazinon (nM)

B

Page 89: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

87

9. Conclusion

We have demonstrated the application of manganese dioxide as an electrochemical mediator in thiol determination. High catalytic activity in thiol detection was exhibited by the thick fi lm graphite sensors modifi ed with γ-MnO2. Low limit of detection of thiocholine (6 · 10–8 М) obtained with the elaborated sensors allowed for the development of a highly sensitive technique to determine BChE and its inhibitor diazinon which may be used in environmental control procedures and clinical diagnostics.

10. AcknowledgementsThe fi nancial support for this work from Russian Foundation for Basic Research

(project no. 10-08-00895-a, 11-08-01306-a and 13-08-01078) and NATO grant NRSFP-#984082 are gratefully acknowledged.

References

[1] T.L. Rosenberry, Acetylcholinesterase in M. Alton (Ed.), Advances in Enzymology and Related Areas of Molecular Biology (2006), 103–218.

[2] T.C. Marrs, Toxicology of organophosphate nerve agents, chemical warfare agents , in: T.C. Marrs, R.L. Maynard, F.R. Sidell (Eds.), Toxicology and Treatment, Wiley, New York, (2007), 191–221.

[3] J. Bajgar, Biological monitoring of exposure to nerve agents ,Br. J. Ind. Med.49 (1992) 648–653.[4] B.W. Wilson, J.D. Henderson, Blood esterase determinations as markers of exposure, Rev. Environ. Contam.

Toxicol.128 (1992) 55–69.[5] E.A.Dontsova,Y.S.Zeifman,I.A.Budashov,A.V.Eremenko,S.L.Kalnov,I.N.Kurochkin, Screen-printed carbon

electrode for choline based on MnO2 nanoparticles and choline oxidase/polyelectrolyte layers,SensorsandActuatorsB:Chemical,159 (2011) 261-270.

[6] A. V. Eremenko, E. A. Dontsova, A. P. Nazarov, E. G. Evtushenko, S. V. Amitonov, S. V. Savilov, L. F. Martynova, V. V. Lunin, I. N. Kurochkin,ManganeseDioxideNanostructuresasaNovelElectrochemicalMediatorforThiolSensors, Electroanalysis, 3 (2012), 573-580.

Page 90: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

88

Chapter 7

Genotyping Of Esterase Genes (Ache, Pnpla6, BcheAnd Ces1) And Paraoxonase 1 Gene (Pon1)

Valery V. Nosikova 1, Vladimir V. Bochkarevb, Alexey G. Nikitina

a Laboratory of post genomic molecular genetic studies, Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, Russia

b Laboratory of functional genomics and transcriptomics, Institute of General Pathology and Pathophysiology of RussianAcademy of Medical Science,Moscow, Russia

Abstract.Some toxic organophosphorus compounds ( OPCs) were produced and used in several countries as insecticides, therapeutic agents and chemical warfare agents. We suppose that individual genetic differences could determine the susceptibility to OPC action. Four esterase genes, which products act as stoichiometric scavengers of OP compounds, have been used in our study: acetylcholinesterase (ACHE), neuropathy target esterase (PNPLA6), butyrylcholinesterase (BCHE) and carboxylesterase (CES1). Paraoxonase 1 gene ( PON1), which product can hydrolyze and detoxify OP compounds, has been also included to our study. To identify the polymorphic marker genotypes we developed PCR technique in combination with restriction endonuclease cleavage. In case of ACHE gene we used intronic polymorphism C/T (rs2571598) and restriction endonuclease HaeIII. In case of PNPLA6 gene we used two polymorphisms: G221A (rs2303178) in promoter region and Ala412Pro (rs17854645) in coding region (restriction endonucleases HaeIII and PspN4I, correspondingly). In case of BCHE gene we used two polymorphism in coding region: Asp70Gly (rs1799807) and Ala539Thr(rs1803274) with restriction endonucleases Kzo9I and AluI, correspondingly. In case of CES1 gene we used C/T polymorphism (rs2287194) in promoter region and restriction endonuclease TaiI. In case of PON1gene we used three polymorphisms: Gln192Arg (rs662) and Leu55Met (rs854560) in coding region (restriction endonucleases PctI and FaeI, correspondingly) and C(-108)T (rs705379) in promoter region with restriction endonuclease Fsp4HI.

Key words. esterase genes, paraoxonase 1 gene, PCR technique, restriction endonuclease cleavage, identifi cation of polymorphism genotypes

Introduction Organophosphorus compounds ( OPCs) with anticholinesterase properties (anti-ChE

OPCs) are widely used as insecticides; to a less extent they are used as therapeutic agents: for treatment of schistosomiasis, glaucoma and Alzheimer’s disease (AD).In addition, a wide agricultural and domestic use of OPCs as insecticides makes these compounds accessible to various strata of society and consequently widens the possibility of OPCs

1 Corresponding author: Valery V. Nosikov, Kosygin street 4, Moscow 119991, Russian Federation; E-mail: [email protected]

Page 91: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

89

being used for chemical terrorism. OPC of both known and unknown structure may also arise as a result of various chemical accidents.

Genetic polymorphisms of esterases involved in OPC interactions may lead to enzyme variants with a higher or lower catalytic activity and/or with different levels of expression. It allows us to suggest that polymorphisms of esterases can play essential role in sensitivity to OPC and intoxications development. Thus we suppose that individual genetic differences could determine the susceptibility to OPC action. All polymorphisms of esterase genes and paraoxonase 1 gene, which were developed in our study, and data about chromosome localization, numbers in SNP database (RS) and polymorphism types are listed in table 1.

The aim of present study was to develop the methods for identifi cation of the genotypes of several polymorphisms located in four esterase genes (ACHE, PNPLA6, BCHE and CES1) and paraoxonase 1 gene ( PON1).

Table 1. Polymorphic markers of esterase genes and paraoxonase 1 gene.

GENE Chromosome localization RS Polymorphism

Butyrylcholinesterase (BCHE) 3q26.1 rs1799807rs1803274

Asp70GlyAla539Thr

Acetylcholinesterase (ACHE) 3p25 rs2571598 316C>T in intron 3

Carboxylesterase 1 (CES1) 16q13-q22.1 rs2287194 269C>T in 3’-NTS

Neuropathy target esterase(PNPLA6) 19p13.3 rs2303178

rs17854645 221A>G in 3’-NTS Ala412Pro

Paraoxonase 1 ( PON1) 7q21.3rs662

rs854560rs705379

Gln192ArgLeu55MetC(–108)T

1. Materials and methods

Total DNA was isolated from whole- blood samples pretreated with proteinase K using a standard protocol for extraction with phenol-chloroform [1]. For each gene studied, polymorphic regions were amplifi ed by a polymerase chain reaction ( PCR) in a total volume of 25 μl using PCR reagents and Taq polymerase from “Evrogen” followed by digestion of a PCR product with a corresponding restriction endonuclease. The primer sequences and the corresponding restriction endonucleases are summarized in Table 2. Polymorphic regions of genes were amplifi ed in a PCR buffer containing 10 mmol/l Tris-HCl, pH 8.8, 50 mmol/l KCl, 0.08% Nonidet P40, 1.5 mmol/l MgCl2, 0.2 mmol/l of each dNTPs, 100 ng of genomic DNA, 1 U Taq polymerase, and 10 pmol of each primer. The PCR cycling was performed in a DNA Engine Dyad Peltier Thermal Cycler (Bio-Rad Laboratories, Hercules, CA, USA) as follows: initial denaturation step at 96°C for 3 min, followed by 30 cycles of denaturation at 94°C for 30 s, annealing at 62 – 65°C for 30 s and extension at 72°C for 30 s, with a fi nal extension of 7 min at 72°C. About 5 μl of the PCR product was then digested with 5U of corresponding restriction endonucleases (Sibenzyme Ltd, Novosibirsk, Russia) for 3 h in 10 μl of the reaction mixture, containing corresponding buffers (Sibenzyme) and 0.1 mg/ml BSA.

Page 92: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

90

Table 2. The primer sequences and the corresponding restriction endonucleases.

Gene Polymorphism Primers Restriction endonuclease

BCHErs1799807

F1, ggtctgatatttggaatgR1, tgacactacaataactct

Kzo9I

rs1803274F, atgctgtactgtgtagttagagaaR, ctgctttccactcccattcag

AluI

ACHE rs2571598F1, tcttgttatgttgtccagR1, atatgctcacaaagtaga

HaeIII

CES1 rs2287194F, gggggcaggggacagagR, aaaggtgcatcaggcccag

TaiI

PNPLA6rs2303178

F, ctgacctgccctgagcggR, cagaaggtgggtccccaggc

HaeIII

rs17854645F, acatccctggaaaccccctcgR, catggccctccccgcagacc

PspN4I

PON1

rs662F, ggaatagacagtgaggaatR, ttccattagcaaaatcaaatc

PctI

rs854560F1, cagtccattaggcagtatR1, ttcaagtgaggtgtgataa

FaeI

rs705379F1, ccgattggcccgcgccR1, ggacttttggctgaaagtg

Fsp4HI

2. Results and DiscussionIn case of butyrylcholinesterase gene we developed the methods for allele and

genotype identifi cation for two polymorphisms: Asp/Gly in position 70 and Ala/Thr in position 539 of aminoacid sequence of this enzyme. In case of Asp70Gly polymorphism it was shown that BuChE with Gly in position 70 has 30% lower enzymatic activity than Asp variant [2]. Homogygous carriers (G/G) display extreme anxiety after exposure to cholinesterase inhibitors (CIs). These variations can result in atypical reactions to CIs and altered behavior/ toxicity of pharmaceuticals. In case of Ala539Thr it was shown that BuChE with Thr in position 539 (K variant) has 30% lower enzymatic activity than Ala variant [3]. Thr359 allele often co-inherited with “atypical” BuChE (Gly70).

In case of Asp70Gly polymorphism after PCR we have obtained DNA fragment with length 275 bp. The digestion of this DNA fragment with restriction endonuclease Kzo9Iresulted in three DNA fragments (170, 75 and 30 bp) for Asp allele and two DNA fragments (170 and 105 bp) for Gly allele.

In case of Ala539Thr polymorphism after PCR we have obtained DNA fragment with length 144 bp. The digestion of this DNA fragment with restriction endonuclease AluIresulted in two DNA fragments (120 and 24 bp) for Ala allele, while the Thr allele was not digested.

In case of acetylcholinesterase gene we developed the methods for allele and genotype identifi cation for C/Tpolymorphism locatedin intron 3 (position 316) of ACHE gene. This polymorphic marker associated with the late-onset form of Alzheimer’s disease and with response to treatment with donepezil and rivastigmine [4]. In case of 316C/T polymorphism after PCR we have obtained DNA fragment with length 244 bp. The digestion of this DNA

Page 93: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

91

fragment with restriction endonuclease HaeIIIresulted in three DNA fragments (134, 78 and 30 bp) for G allele and two DNA fragments (134 and 108 bp) for C allele.

In case of carboxylesterase 1 gene we developed the methods for allele and genotype identifi cation for C/Tpolymorphism locatedin 3’-NTS (position 269) of CES1 gene. This polymorphic marker associated with transcriptional activity and with response to treatment with clopidogrel [5]. In case of 269C/T polymorphism after PCR we have obtained DNA fragment with length 266 bp. The digestion of this DNA fragment with restriction endonuclease TaiIresulted in two DNA fragments (156 and 110 bp) for T allele, while the C allele was not digested.

In case of neuropathy target esterase (in OMIM patatin-like phospholipase domain-containing protein 6) we developed the methods for allele and genotype identifi cation for two polymorphisms: A/G polymorphism locatedin 3’-NTS in position 221 and Ala/Pro in position 412 of aminoacid sequence of this enzyme. Polymorphismsof PNPLA6 gene associated with transcriptional activity and sick building syndrome[6].

In case of 221A/G polymorphism after PCR we have obtained DNA fragment with length 119 bp. The digestion of this DNA fragment with restriction endonuclease HaeIII resulted in two DNA fragments (100 and 19 bp) for G allele, while the A allele was not digested. In case of Ala412Pro polymorphism after PCR we have obtained DNA fragment with length 100 bp. The digestion of this DNA fragment with restriction endonuclease HaeIII resulted in two DNA fragments (77 and 23 bp) for Ala allele, while the Pro allele was not digested.

In case of paraoxonase 1 gene we developed the methods for allele and genotype identifi cation for three polymorphisms: Gln/Arg in position 192 and Leu/Met in position 55 of aminoacid sequence of this enzymeandalso C(–108)T in promoter region of PON1 gene. PON1 exhibits a substrate dependent polymorphism. The PON1 isozyme (glutamine at position 192) can be several times less effi cient than isozyme with arginine at position 192 in hydrolyzing paraoxon. On the other hand diazoxon, soman and sarin are hydrolyzed appreciably better by the Arg192 variant than the Glu192 isozyme [7, 8]. A second polymorphism Leu55Met affect the PON1 activity lesser than polymorphism at position 192 [9, 10]. Variants of Gln192Arg polymorphism are correlated with heart diseases and trait-anxiety scores. Variants of Leu55Met polymorphism are correlated with heart diseases, diabetic retinopathy and trait-anxiety scores. In case of C(–108)T polymorphism the PON1 enzyme activity has been reported to primarily involve this SNP, with the following average values of arylesterase activity reported per genotype: CC – 126, CT – 103, TT – 68 [11].

In case of Gln192Arg polymorphism after PCR we have obtained DNA fragment with length 267 bp. The digestion of this DNA fragment with restriction endonuclease PctI resulted in two DNA fragments (166 and 101 bp) for Arg allele, while the Gln allele was not digested. In case of Leu55Met polymorphism after PCR we have obtained DNA fragment with length 125 bp. The digestion of this DNA fragment with restriction endonuclease FaeI resulted in two DNA fragments (95 and 30 bp) for Met allele, while the Leu allele was not digested. In case of C(–108)T polymorphism after PCR we have obtained DNA fragment with length 124 bp. The digestion of this DNA fragment with restriction endonuclease Fsp4HI resulted in two DNA fragments (107 and 17 bp) for C allele, while the T allele was not digested.

Page 94: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

92

3. ConclusionThe methods for identifi cation of the genotypes of several polymorphisms located in

four esterase genes (ACHE, PNPLA6, BCHE and CES1) and paraoxonase 1 gene ( PON1) have been developed.

4. AcknowledgementsThis work is supported by NATO Science for Peace and Security Program – SfP No

984082

Rferences

[1] B. Budowle, F.S. Baechtel. Modifi cations to improve the effectiveness of restriction fragment length polymorphism. Appl. Electrophor. 1 (1990) 181 – 187.

[2] M.C. McGuire, C.P. Nogueira, C.F. Bartels, H. Lightstone, A. Hajra, A.F. Van der Spek, O. Lockridge, B.N. La Du. Identifi cation of the structural mutation responsible for the dibucaine-resistant (atypical) variant form of human serum cholinesterase. Proc. Natl. Acad. Sci. USA. 86(3) (1989), 953 – 957.

[3] C.V. Altamirano, C.F. Bartels, O. Lockridge. The Butyrylcholinesterase K-Variant Shows Similar Cellular Protein Turnover and Quaternary Interaction to the Wild-Type Enzyme. Journal of Neurochemistry74(2) (2000) 869 – 877.

[4] R. Scacchi, G. Gambina, G. Moretto, R.M. Corbo. Variability of AChE, BChE, and ChAT genes in the late-onset form of Alzheimer’s disease and relationships with response to treatment with Donepezil and Rivastigmine. Am. J. Med. Genet. Part B. Neuropsychiatric Genetics.150B(4) (2009) 502 – 507 .

[5] D. Xiao, Y.T. Chen, D. Yang, B. Yan. Age-related inducibility of carboxylesterases by the antiepileptic agent phenobarbital and implications in drug metabolism and lipid accumulation. Biochem Pharmacol.84(2) (2012) 232 – 239.

[6] Y. Matsuzaka, T. Ohkubo, Y.Y. Kikuti, et al. Association of sick building syndrome with neuropathy target esterase ( NTE) activity in Japanese. Environ Toxicol. (2013) Feb 18. [Epub ahead of print].

[7] B.N. LaDu, E.C. Furlong, E. Reiner. Recommended nomenclature system for the paraoxonases, Chem.-Biol. Interact. 119–120 (1999) 599–601.

[8] D.M. Shih, A.J. Lusis, L.G. Costa. The PON1 gene and detoxication, NeuroToxicology. 21(4) (2000) 581–588.

[9] B. Mackness, M.I. Mackness, S. Arrol, W. Turkie, P.N. Durington. Effect of the molecular polymorphisms of human paraoxonase ( PON1) on the rate of hydrolysis of paraoxon. Br. J. Pharmacol.122 (1997) 265–268.

[10] C.E. Furlong,W.-F. Li, R.J. Richter, D.M. Shih, A.J. Lusis, E. Alleva, L.G. Costa. Genetic and temporal determinations of pesticide sensitivity: role of paraoxonase ( PON1). NeuroToxicology 21(1–2) (2000) 91–100.

[11] V.H. Brophy, R.L. Jampsa, J.B. Clendenning, L.A. McKinstry, G.P. Jarvik, C.E. Furlong. Effects of 5’ regulatory-region polymorphisms on paraoxonase-gene ( PON1) expression. Am. J. Hum. Genet.68(6) (2001) 1428-1436.

Page 95: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

93

Chapter 8

Esterase Status as Biomarker of OPC Exposure and Treatment with Reactivator of CHE

Christophor DISHOVSKY, Tifon IVANOV, Iskra PETROVA

Department of Medicine of Disasters and Toxicology, Department of Toxicology, Military Medival Academy, Sofi a, Bulgaria

Abstract. Some highly toxic OPCs were produced and used in several countries as chemical warfare agents. Defending against such agents requires rapid, sensitive and specifi c detection of them and their biological effects. Thus, the development of biomarkers of human exposures to OPCs and their quantifi cation is a vital component of a system of prediction and early diagnosis of induced diseases. The esterase status was defi ned as a total activity of different esterases which could be used as potential biomarker for OPCs exposure. The main enzymes which determine the esterase status are acethylcholinesterase (AChE), buthyrylcholinesterase (BChE), carboxylesterase ( CaE), neurotoxic esterase and paraoxonase 1 ( PON1).In the present study male Wistar rats (180-220 g) were used in the experiments. The paraoxon was administrated at dose 1.5 LD50 (i.m.), 20 min after i.m. application of atropine (20mg/kg b.w.). The therapy was done 1 min after poisoning using the tested reactivator Toxidin ( HI-6) (20mg/kg b.w.), i.m. The blood samples were taken in 60 min after antidote application. The methods of blood sampling and preparation as well as spectrophotometric methods for determination of activities of AChE, BChE, CaE and PON1 were adopted by the IPAC RAS team. The blood hemolysate and plasma were prepared. In the AChE assay ethopropazine was used as a specifi c inhibitor of BChE.The obtained results showed that AChE activity in rat blood was decreased after intoxication with paraoxon. When reactivator was applied, the activity was notability recovered. CaE was signifi cantly affected by the OPC and it was not reactivated by the tested oxime. Plasma BChE as well as PON1 were not affected in all cases compared to the control group.

Key words. Cholinesterase reactivators, esterase status, HI-6, Paraoxon, Toxidine

Introduction

Organophosphorus compounds ( OPCs) with anticholinesterase properties are widely used as insecticides; to a less extent they are used as therapeutic agents. Some highly toxic OPCs were produced and used in several countries as chemical warfare agents. The growing threat of international terrorism brings new scenarios for disaster inwhich known organophosphorus agents can be used or OPCs of an unknown structure may arise as a result of attacks on chemical plants or stockpiles of pesticides and other chemicals.

Defending against such agents requires rapid, sensitive and specifi c detection of them and

Page 96: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

94

their biological effects. Thus, the development of biomarkers of human exposures to OPCs and their quantifi cation is a vital component of a system of prediction and early diagnosis of induced diseases and analysis of effectiveness and prognosis of antidote treatment. The esterase status was defi ned as a total activity of different esterases which could be used as potential biomarkers for OPCs exposure [1,2]. The main enzymes which determine the esterase status are: acetylcholinesterase (EC 3.1.1.7, AChE), butyrylcholinesterase (EC 3.1.1.8, BChE), carboxylesterase (EC 3.1.1.1, CaE), neuropathy target esterase (EC 3.1.1.5, NTE) and paraoxonase (EC 3.1.8.1, PON1).

Determination of this set of esterase activities allows one to improve the possibilities of diagnosis and prognosis of intoxications development.

The drug therapy on intoxication with organophosphorus compound (OPC) included mainly com bination of cholinesterase reactivators and cholinolytics [3]. There is no single AChE reactivator having the ability to suffi ciently reactivate inhibited enzyme due to the high variability of chemical structure of the inhibitors. Antidote activity of reactivators of ChE is different against the different OPC. Up to now, drugs effective against all the neuroparalitic OPC [4] have not been found.

The most well-known among of new reactivators of ChE is HI-6 ((1-(4-imino-carbonylpyridinium) 1-(2-hydroxyiminomethyl-pyridinium) dimethylether dichloride), because the research so far shows that at the moment it is one of the best reactivators of the inhibited from Soman acetyl cholinesterase ( AChE )[5]. This reactivator, including synthesized in our Department Toxidine, has an effect against intoxications with sarin, soman and Vx, and to a lesser degree against tabun [6,7 ] .

In clinical cases [8 ] after intoxications with chlorpirofos and diasinon treated with reactivator of ChE toxogonin, the recovery of ChE activity was not correlated with the improvement of the health restoration. In this study our aim was to examine the effects of a Paraoxon caused intoxication and treatment with reactivator of ChE HI-6, on esterase status as a model of complex biomarker of the OPC-caused intoxications and its prognoses.

Materials and Methods.

Chemicals. Paraoxon (O,O-diethyl-4-nitrophenyl phosphate) was from Sigma Chemical Co (St. Louis, MO). Toxidin ( HI-6,(1-(4-imino-carbonylpyridinium) 1-(2hydroxyimino-methyl-pyridinium) dimethylether dichloride) was synthesized and characterized in the Department of Toxicology, Military Medical Academy, Sofi a. All other chemicals were analytical grade.

Animals and experimental model. Male Wistar rats (180-220 g) were used in the experiments. The animals had access to food and water ad libitum and were maintained at 24 ± 2 °C with a 12 h light/dark cycle. All experiments and procedures with animals were approved and carried out according to the guidelines of the Ethical Committee on Military Medical Academy. Four experimental groups of animals (N=6) were used: control group of untreated animals (Control); control group with antidote treatment – reactivator of ChE ( HI-6), poisoned group ( paraoxon only) (Intox), poisoned group with antidote treatment (Intox+treat).

Poisoning and treatment.The paraoxon was administrated at dose 1.5 LD50 (i.m.), 20 min after i.m. application of atropine (20mg/kg b.w.). The therapy was done 1 min after poisoning using the tested reactivator Toxidin (20mg/kg b.w.), i.m. The blood samples

Page 97: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

95

were taken in 60 min after antidote application. The studied reactivator was synthesized in Military Medical Academy, Dept. Toxicology, Sofi a.

Enzyme assays. The blood hemolysate and plasma were prepared. The enzyme activity assays (spectrophotometric methods for determination of activities of AChE, BChE, CaE and PON1), were performed according to Rudakova et al. [2] using Hospitex Diagnostic clinical chemistry analyzer (Hospitex) and S-22 UV-Vis spectrophotometer (Boeco). In the AChE assay ethopropazine was used as a specifi c inhibitor of BChE.

Results and discussion

At fi gures 1 and 2 are presented the structural formulas of HI-6 and paraoxon.

Fig. 1. HI-6, Toxidin ,,(1-(4-imino-carbonylpyridinium) 1- Fig. 2. Paraoxon(2 hydroxyimino-methyl-pyridinium) dimethylether dichloride)

The results on activity of AChE, BChE, CaE (substrate – Naphtyl acetate) and PON1 (substrates – Paraoxon or Phenylacetat ) in blood hemolysate or plasma after intoxication with 1.5 LD50 of paraoxon and treatment with reactivator of ChE are presented in Fig. 3, 4, 5, 6, 7 8.

Fig. 3. Enzyme activity of AChE and BuChE in hemolyzed blood after Paraoxon intoxication and treatment with HI-6.

N N CONH 2CH 2OCH 2

2X

CHNOH

Page 98: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

96

The obtained results showed that in hemolyzed blood AChE activity was decreased in the poisoned group of animals, but when reactivator was applied the activity was recovered (Fig. 3).

Plasma’s AChE and BChE are inhibited from this dose of paraoxon and reactivated from HI-6 in different extent (Fig. 4).

The most affected plasma enzyme was CaE which had been practically inhibited by the poison (up to 90% inhibition) and the enzyme activity was not recovered by the reactivator (Fig.5 and 6).The reactivator itself displayed slight activation effect on the enzyme activity.

PON1-PO and PON1-PhA are not affected in all cases compared to the control group (Fig. 7 and 8).

Fig. 4. Enzyme activity of AChE and BuChE in plasma after Paraoxon intoxication and treatment with HI-6

Fig. 5. Enzyme activity of CaE in hemolyzed Blood after Paraoxon intoxication

and treatment with HI-6.

Page 99: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

97

Fig. 6. Enzyme activity of CaE in plasma after Paraoxon intoxication

and treatment with HI-6

Fig. 7. Enzyme activity of PON 1 in plasma (substrate PO)

after Paraoxon intoxication and treatment with HI-6

Fig. 8. Enzyme activity of PON 1 in plasma (substrate PhA)

after Paraoxon intoxication and treatment with HI-6

Page 100: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

98

Conclusion

In this experiment, esterase status (excluding neurotoxic esterase) can be considered as the effective, sensitive and informative complex biomarker of exposure to OPCs.

AChE, BChE and CaE were inhibited by Paraoxon in different extend.Therapeutic treatment with reactivator of ChE – HI-6, showed that AChE and BChE

were reactivated in different extend. CaE activity was not recovered by reactivator of ChE HI-6.

There are differences in activity of enzymes included in esterase status in rats and human blood [2,9]. That must be discussed if esterase status will be used for the control and prognosis of OPC intoxications and optimization of therapy in human clinical practice.

This work is supported by NATO Science for Peace and Security Program - SfP No 984082.

References

1. G. F. Makhaeva, E.V. Rudakova, N.P. Boltneva, L.V. Sigolaeva, A.V. Eremenko, I.N. Kurochkin, R.J. Richardson, Blood Esterases as a Complex Biomarker for Exposure to Organophosphorus Compounds. In: “Counteraction to Chemical and Biological Terrorism in the East Europe Countries”, Eds C. Dishovsky, A. Pivovarov, NATO Security through Science Series A, Springer, (2009,) 177-194.

2. E. V. Rudakova, N.P. Boltneva, G.F. Makhaeva, Comparative analysis of esterase status of human and rat blood, Bull Exp Biol Med 152(1) (2011), 73-75 [Translated from Byulleten’ Eksperimental’noi Biologii i Meditsiny, 152 (7) (2011), 80-82.]

3. Dishovsky, C., Chlinestrase reactivators, SA, Sofi a, 1990, (in Bulgarian).4. Briggs, C., and Simons, K., Personal protection against chemical agent: development of antidotal treatment

for organophosphorus poisoning, Arch. Belg. Med. Soc., 20, 260-273, 1984.5. Oldiges, H., and Schoene, K., Pirydinium und Imidozolium-salze als antidote gegenuber soman und Paraoxon

vergiftungen bei Mausen, Arch. Toxicol.,26,293,1970.6. Dishovsky, C., Doctor of Sciences work, 1989, MMA, Sofi a, (in Bulgarian).7. Dishovsky, C., Chlinestrase reactivators, SA, Sofi a, 1990, (in Bulgarian).8. Dishovsky C., Popov T., Petrova I., Kanev K., Hubenova A., Samnaliev I., Biomarkers of Nerve Agents

Exposure, Dishovsky C., Pivovarov A., Editors, Counteraction to Chemical and Biological Terrorism in the East Europe Countries, Springer, 2009, 155-165.(J.Med.CBRDefense).

9. B. Li, M. Sedlacek, I. Manoharan, R. Boopathy, E.G. Duysen, P. Masson, O. Lockridge, Butyrylcholinesterase, paraoxonase, and albumin esterase, but not carboxylesterase, are present in human plasma, Biochem Pharmacol 70 (2005), 1673-1684.

Page 101: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

99

Part 2

CONTEMPORARY ASPECTS OFCLINICAL TOXICOLOGY

Page 102: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

100

Page 103: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

101

Chapter 9

Multi-organ Dysfunction Syndrome – a Result of Prolonged Hypoxia from an Overdose

of Methadone

GESHEVA M., M. PETKOVA, J. RADENKOVA-SAEVA, A. LOUKOVAToxicology Clinic, UMHATEM “N. I. Pirogov”, Sofi a, Bulgaria

Absract: Methadone is a synthetic opioid, used for treatment of opioid dependence. Methadone is characterized by slow metabolism. This peculiarity deremines a long-term depression of consciousness and breathing with prolonged hypoxia. We present three cases of severe methadone poisoning, treated in the Clinic of toxicology with multi-organ dysfunction syndrome. Clinical symptoms are characterized by coma, cyanosis and respiratory depression. We observed the laboratory abnormalities and functional performance, course and outcome of the disease. Two of the patients were discharged from the hospital with severe multi-organ system changes, considered as “ persistent vegetative state”.We consider that the regime of supply with methadone should be revised. Adoption without control creates a risk of fatal poisoning or temporary / permanent disability.

Key words: methadone, hypoxia, “ persistent vegetative state”

Introduction

Methadone is a synthetic opioid, similar to morphine, but it doesn`t cause euphoric effect. It was developed in Germany in 1937 to deal with acute and chronic pain.[1] It was fi rst used in the U.S.A. in 1947. Methadone is nowadays administered in the treatment of opioid addiction and 500 000 people in Europe are on Methadone treatment in 2013. The fi rst Methadone treatment program in Bulgaria started in 1995. The majority of heroin addicts are currently in public and private Methadone treatment programs. With the increasing number of patients included in these programs appears the black market of methadone, maintained by illegal import and addicts in public Methadone programs who receive doses for a few days at once and sell their methadone. The black market is a prerequisite for the occurrence of methadone addiction without background for heroin addiction. We observe uncontrolled abuse of methadone, including intravenous use.

There are many similarities between heroin and methadone but on the other hand there are also differences in their action. Similarities: opiate receptor agonists. Differences: methadone has a slow metabolism and has a long plasma half-life, thus creating a plateau of methadone plasma concentration - 24-36 hours [2, 3, 4]. This is a desired therapeutic effect [2, 3]. It is administered once per day.

Page 104: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

102

Case presentationsWe present three clinical cases of young people, age between 21 and 26, who have been

treated in UMHATEM “N. I. Pirogov”-Clinic of Toxicology and ICU for a period of six months. In the three cases severe multi-organ dysfunction resulting from administration of toxic doses of methadone was observed.

Patients were not included in Methadone treatment programs and had no history of heroin dependence.

We present each clinical case, describing the pathological focus of clinic presentation, laboratory tests, imaging, toxicochemical analysis, treatment and outcome.

Case I A 22-year-old woman with history of alcohol, marijuana, amphetamine and

benzodiazepine intake the previous day, who spent eight hours in “sleep” and could not be awakened.She was transported to the Emergency Room of UMHATEM “N. I. Pirogov”. Her status upon admission: coma, moderate miosis, mild cyanosis, breathing - 6 / min, exotoxic shock, blood pressure of 70/30mmHg, heart rate of 100bpm, serum glucose 2.0 mmol / l. Chemicotoxicologicalanalysis detectedMethadone, Rivotril , cannabinoid, alcohol - 0,51 ‰. Radiogram of the lungs - on admission: evidence of aspiration in the right; on the sixth day – pneumothorax in the right. Abdominal ultrasound - on admission: normal; on the sixth day - acute renal parenchymal process.

Table1: Laboratory results

Test

Day

Creat.

μmol/l

Urea

mmol/l

АSАТ

U/l

АLАТ

U/l

Amylase

U/l

PT

%INR

1 130 6,8 8832 6215 202 34 2,38

3 348 13,3 3736 4836 - 25,3 3,01

4 475 31,3 82 66 50 47,8 1,81

16 80 3,9 41 100 - 75 1,26

CBCandblood gases – normal.

Treatment and course of the diseaseTreatment - resuscitation, antidote, detoxication, antibiotic,cerebroprotective,

hepatoprotective and symptomatic treatment, transfusion of bioproducts.Course of the disease - the patient developed transition hepatorenal syndrome, a complication of aspiration pneumonia with pneumothorax, which required treatment in ICU - 7 days. She was discharged without signs of intoxication and with recovered hepatic, renal, pulmonary and pancreatic functions on day 19 of hospitalization.

Neuropsychiatric status at discharge: comprehensively orientated, slowand poor associative thought process, desire for continuous movement of the lower extremities, retrograde and congrade amnesia. The patient was discharged with posthypoxic exotoxic encephalopathy and encephalopathy related to abuse of psychoactive drugs.

Page 105: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

103

Case I IA 26-year-old man with a history of methadone abuse in the last 3-4 months and alcohol

ingestion /beer and concentrate/ the previous day. He spent 13 hours in “sleep”, could not be awakened and was transported to the Emergency Room of UMHATEM “N. I. Pirogov”.His status upon admission: coma, cyanosis, pinpoint pupils, breathing -2-3/ min, blood pressure of 112/70, heart rate 120/min.Chemicotoxicological analysis detectedmethadone and ethanol 0,66 ‰ . Radiogram of the lungs and abdomen and abdominal ultrasonography -normal. CT of the brain - on admission: leucodystrophic changes in the white matter of the cerebellar hemispheres and symmetrical dystrophic changes in the basal ganglia.

Table2: Laboratory results

Test

Day

Creat.

μmol/l

Urea

mmol/l

АSАТ

U/l

АLАТ

U/l

Amylase

U/l

1 195 9,6 936 266 408

2 111 7,1 602 234 302

3 76 4,3 412 207 222

14 51 5,2 25 43 122

Othertests - CBC, coagulation, metabolic status and blood gases - inthenormalrange.

Treatment and course of the disease:Treatment - resuscitation, antidote, detoxication, cerebroprotective, hepatoprotective,

antibiotic and symptomatic treatment. No sedative agents were required for the conduction of mechanical ventilation in the ICU. Course of the disease - the patient was transferred from the ICU to the Clinic of Toxicology on day 14 and was discharged with overcome intoxication and recovered hepatic, renal and pancreatic functions on day 26.

Neuropsychiatric status after transfer from CIU and at discharge: responded to painful stimuli, answered simple questions / name /, preserved swallowing refl ex, muscle tone –abnormally low muscle tone of the limbs (especially upper limbs), hypoactive tendon refl exes, no pathological refl exes. The patient was discharged with posthypoxic exotoxic encephalopathy.

Diagnosis: Persistent vegetative state.Case I II A 21-year-old male went back home sleepy and with slurred speech after party in a

disco. After 8 hours ‘ sleep ‘ he couldn`t be waken up by his parents and was transported to the Emergency Room of UMHATEM “N. I. Pirogov”. His status upon admission: coma, cyanosis, pinpoint pupils, respiratory rate - 2-4 / min, BP 90/56, heart rate 144/min. Chemicotoxicologic alanalysis detected Methadone, Akineton, ethanol - 0,9 ‰. Radiogram of the lung - on admission: normal, on day 7 of hospitalization: pleural effusion. Ultrasonography of the abdomen and heart – normal. CT of the brain – on admission:

Page 106: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

104

leucodysthrophic / ischemic areas with lower density/ white matter lesions of the brain located periventriculary. EEG – on day 21: diffuse slow wave activity with bilateral temporal bisynchronous epileptiform focus; findings corresponding to encephalopathy with complex partial seizures.

Table 3: Laboratory results

Test

Day

Creat.

μmol/l

Urea

mmol/l

АSАТ

U/l

АLАТ

U/l

Amylase

U/l

1 208 8,3 191 137 219

2 199 6,2 88 125 308

3 114 4,7 60 48 260

58 37 3,3 29 34 88

CBC, coagulation, metabolic status and blood gases - in normal range.

Treatment and course of the diseaseTreatment - resuscitation, antidote, detoxication, cerebroprotective, hepatoprotective, antibiotic, anticonvulsant and symptomatic treatment, parenteral nutrition. No sedative agents were required for the conduction of mechanical ventilation in the ICU. Course of the disease - the patient was discharged without signs of intoxication and with recovered hepatic, renal and pancreatic functions on day 60 of hospitalization.

Neuropsychiatric status: awake but unresponsive, lying with open eyes, reacted to painful stimuli with a grimace, decerebrate rigidity, preserved swallowing refl ex, clonic movements of the masseter muscles, whitout symptoms of meningoradicular irritation, deviated eyes to the left, quadripyramidal syndrom with contractures of the upper and lower extremities.The patient was discharged with posthypoxic exotoxic encephalopathy.Diagnosis: Persistent vegetative state.

DiscussionIn the three cases impress the following similarities: a lack of a history of heroin abuse;

the patients were not in Methadone treatment programs; presence of methadone and alcohol ingestion; long period between the Methadone ingestion and the medical assistance; on admission the patients presented with coma, miosis, cyanosis and respiratory depression; adequate response to antidote-Naloxone; severe multi-organ damages from hypoxic type; two of the patients developed an organic brain psychosyndrome as a result of prolonged exotoxic hypoxia.

ConclusionThe slow metabolism of methadone and the long plasma half-life are prerequisite for

a prolonged toxic concentration after overdose [3, 4]. The late hours of the day when the physiological sleep passes into exotoxic depression of consciousness and breathing do

Page 107: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

105

not worry the others are especially dangerous. The prolonged hypoxia is the cause for the development of a severe multi-organ failure [4, 5, 6]. It could be overcome if adequate treatment is administered but the damages of the CNS are permanent or defi nitive and lead to disability.

We consider that the regime of methadone supply should be revised and personally differentiated, based on the building of therapeutic physician-patient alliance.

We appeal for the establishment of administrative health and social normative docu-ments to restrict and repress the black market of methadone and its illegal distribution.

References

[1]. Wechsberg, Wendee M., and Jennifer J. Kasten. Methadone Maintenance Treatment in the U.S.: A Practical Question and Answer Guide. New York, NY: Springer Publishing Company; 1 Edition, 2007.

[2]. Inturrisi CE, Verebely K: The levels of methadone in the plasma in methadone maintenance. Clin Pharmacol Ther 1972; 13:633-637

[3]. Holmstrand J, Anggard E, Gunne LM: Methadone maintenance: Plasma levels and therapeutic outcome. Clin Pharmacol Ther 1978; 23:175-180

[4]. Mathew J. Ellenhorn and Donald G. Barceloux, Medical Toxicology, 2 ed, 1988, Elsevier Science Publishing Company, Inc, pp 714-718

[5]. Garriott JC, Sterner WQ, Mason MF: Toxicology fi ndings in six fatalities involving methadone. Clin Toxicol 1973; 6:163-173

[6]. Baselt R: Disposition of Toxic Drugs and Chemicals in Man, vol 1. Canton, Conn, Biomedical Publications, 1978, pp 21-24

Page 108: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

106

Chapter 10

Serotonin Syndrome in Acute Amphetamine Intoxication

E. KIROVAClinic of Toxicology, UMHATEM “N. I. Pirogov”, Sofi a, Bulgaria

Abstract: Amphetamines are synthetic psychoactive drugs called central nervous system (CNS) stimulants. They are simple synthetic derivatives of phenylethylamine which differ only in possessing a methyl group attached to the side chain. The phenylethylamine structure of amphetamines is similar to catecholaminergic and serotonergic agonists (biogenic amines), which explain their actions. Overdose or chronic excessive use causes a sympathomimetic toxidrome (eg, tachycardia, hypertension, mydriasis, diaphoresis, hyperrefl exia, agitation). According to some authors serotonin syndrome may occurwith CNS effects (coma or agitation), autonomic instability (hyperthermia)and increasedneuromuscularexcitability (seizures).We present a caseof acute amphetamine intoxication with the development ofl ife-threatening serotonin syndrome, which caused differential diagnostic problems. The administration of adequateresuscitation, detoxicating-depuration and symptomatic treatment resulted inrapid correctionandresolution of the syndrome caused by theintoxication within 24 hours. Serotonin syndrome may be a seriouslife-threatening conditionof acuteamphetamineintoxication and in differential diagnostic plan itshould be considered as a possible cause for the development of similar clinical features.

Key words: serotonin syndrome, life-threatening, amphetamine intoxication

IntroductionAmphetamines are synthetic psychoactive drugs called centralnervoussystem (CNS)

stimulants. They are simple synthetic derivatives of phenylethylamine which differ only in possessing a methyl group attached to the side chain.The phenylethylamine structure of amphetamines is similar to catecholaminergic and serotonergic agonists (biogenic amines), which explain their actions[1]. Overdose or chronic excessive use causes a sympathomimetic toxidrome (eg, tachycardia, hypertension, mydriasis, diaphoresis, hyperrefl exia, agitation). According to some authors serotonin syndrome may occur with CNS effects (coma or agitation), autonomic instability (hyperthermia) and increased neuromuscular excitability (seizures)[2, 3, 4].

Serotonin syndrome is a potentially life-threatening condition caused by excessive serotonergic activity in the nervous system. It is characterized by the presence of a triad [4, 5] of:

1. Mental-status changes:confusion, agitation

Page 109: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

107

2. Autonomic hyperactivity: hyperthermia, sweating, hypertension, tachycardia, hyperactive bowel sounds, mydriasis, fl ushing, shievering

3. Neuromuscular abnormality: clonus (spontaneous/inducible/ocular) , muscular hypertonicity, hyperrefl exia, tremor

The clinical manifestations of serotonin syndrome are highly variable. The presence of muscular hypertonicity, sustained clonus and hyperthermia (which may rise as high as 41°C), indicate severe disease.

The syndrome is the consequence of excessive stimulation of the central nervous system and peripheral serotonin receptors by serotonergic agents, ie 5-hydroxytryptamine (5-HT) agonists.

Most cases due to a combination of two or more “serotonergic” drugs but it is possible to be caused by an overdose of a single “serotonergic” drug. Medications that affect any of the steps in serotonin metabolism or regulation can provoke toxicity[6].

Table1: Medications causing serotonin syndrome

MECHANISM DRUGS CAUSING SEROTONIN TOXICITY WITHOUT DRUG INTERACTION

Increased production of serotonin L-tryptophan

Increased serotonin release from neurons Amphetamines, NMDA

5-HTagonism Buspirone, LSD

Decreased serotonin reuptake SSRIs, Venlafaxine, Clomipramine, imipramine,Tramadol, meperidine, methadone, fentanyl, Dextromethorphan

MAO inhibition MAOIs, Selegiline, Linezolid

Uncertain Lithium

5-HT1a - serotonin 1Areceptor, LSD - lysergicaciddiethylamide, MAO - monoamineoxidase, MAOI - monoamine oxidase inhibitor, NMDA - N-methyl-D-aspartate, SSRI - selective serotonin reuptake inhibitor.

Pathophysiology: Serotonin (5-hydroxytryptamine, 5-HT) is synthesised from the amino acid tryptophan. It has central and peripheral effects and there are at least seven different types of serotonin receptors. Centrally, serotonin acts as a neurotransmitter with infl uences on mood, sleep, vomiting and pain perception. Depression is often associated with low concentrations of serotonin. Peripherally, the primary effect of serotonin is on muscles and nerves. The majority of serotonin is synthesised and stored in the enterochromaffi n cells of the gut where it causes contraction of gastrointestinal smooth muscle. Serotonin is also stored in platelets and promotes platelet aggregation. It also acts as an infl ammatory mediator.

Potential mechanisms of serotonin syndrome include increased serotonin synthesis or release; reduced serotonin uptake or metabolism; and direct serotonin receptor activation. Amphetamines cause serotonin syndrome by increasing the release of serotonin and decreasing its reuptake.

There are 7 serotonin receptor families (5-HT1 to 5-HT7), which are further subdivided into groups based on different activities in neural and peripheral organ systems[4, 5].

Page 110: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

108

Table 2: A summary of the effects of serotonin receptor subtypes in relation to serotonin toxicity

5-HT RECEPTOR MAIN ACT ION RELATED TO SEROTONIN TOXICITY

5-HT1A Neuronal inhibition, regulation of sleep, feeding, thermoregulation, hyperactivity associated with anxiety, hypoactivity associated with depression

5-HT1B Locomotion, muscle tone

5-HT1D Neuronal excitation, learning, peripheral vasoconstriction, platelet aggregation

5-HT2B Stomach contraction

5-HT3 Nausea and vomiting, anxiety

5-HT4 Gastrointestinal motility

5-HT-serotonin

There are no specifi c laboratory tests to diagnose serotonin syndrome; A history of current and recent medication use is important, as is ruling out the use of illicit drugs; laboratory and other diagnostic testing are used to rule out alternative explanations of symptoms (neuroleptic malignant syndrome, anticholinergic syndrome, malignant hyperthermia, sepsis, encephalitis, delirium tremens).

Fig. 1: Hunter’s Decision Rules for Diagnosis of Serotonin Toxicity(in patients who are known to have taken a serotonergic agent)

Page 111: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

109

Sternbach’s criteria require three of 10 clinical features coincident with an addition or recent increase of known serotonergic drugs to an established medication regimen:Agitation (restlessness), Diaphoresis, Diarrhea, Disseminated intravascular coagulation, Fever above 100.4° F (38° C), Hyperrefl exia, Incoordination (ataxia), Mental status changes(Confusion, Hypomania), Multi-organ failure, Myoclonus, Ocular clonus, Rhabdomyolysis, Shivering, Tonic-clonic seizures, Tremor.

Case reportWe present a case of a 32-year-old man who was found unconscious and shivering on

the street nearby his home. In the Emergency Medicine Hospital upon admission he was in coma, shievering and sweating, with series of clonic seizures, muscle hypertonicity, hyperthermia up to 42,1 C, dilateded pupils, tachycardia (120-170/min), tachypnea(40/min) and frothing at the mouth. A packet with white powder was found in his pocket and it was unraveld that the patient was addicted to psychoactive drugs and was in Methadone treatment program.

Table 3: Diagnostic tests

Toxicology screening test + methadone and amphetamine in urine sample

Blood panel WBC 14,1 G/L (normal ranges 4,1-11G/L)

CPK 3560 U/L (normal ranges 0-171 U/L)

Other laboratory results in referent ranges

Cranial computer tomography scan normal

Abdominal ultrasound normal

Chest x-ray normal

Blood culture negative

Immediate supportive treatment, including intravenous fl uids, was started. The neurological symptoms were treated with benzodiazepines and Phenobarbital. The hyperthermia was aggressively managed with external cooling, diazepam, hydration and antipyretics (Analgin and Perfalgan), though there is a limited role for traditional antipyretics, as its mechanism in serotonin syndrome is due to muscle tone rather than central thermoregulation [5, 6]. The patient was intubated with induced neuromuscular paralysis. Antibiotic was administered.

The administration of adequateresuscitation, detoxicating-depuration and symp-tomatic treatment resulted in rapid correction and resolution of the syndrome caused by the intoxication within 24 hours.The clonic seizures resolved in an hour after the initiation of treatment, the hyperthermia resolved in 6 hours, in 15 hours the patient became conscious when left without sedation, in 24 hours all vital signs were stabilized and in 36 hours he was extubated. He was discharged on day 4 in good condition.

DiscussionMany cases of serotonin syndrome go unrecognized. In the reported case we observed

the following clue moments, on which depended the accurance of the diagnosis and treatment: critical condition of the patient; presence of a triad (coma, hyperthermia,

Page 112: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

110

seisures); history of amphetamine abuse in combination with methadone ingestion(detected in urine sample); normal diagnostic tests except for the chemicotoxicological analysis; rapid resolution of the syndrome after treatment was started.

Conclusion Serotonin syndrome may be a seriouslife-threatening conditionof acute amphetamine

intoxication, which can cause differential diagnostic problems. Physicians should consider the possibility of serotonin syndrome in patients who use serotonergic agents and present with autonomic changes, mental status changes, and neurological hyperexcitability [4,5]. The immediate adequate treatment leads to rapid resolution of the syndrome.

References

[1]. Leslie Iversen, Speed,ecstasy, ritalin, The science of amphetamines, ed 1, 2008, Oxford University Press Inc, pp 5-25.

[2]. Richard W. Carlson, MD, PhD , Michael A. Geheb, MD, Critical Care Clinics,1997, ElsevierScience Publishing Company Inc, Volume 13, Issue 4, pp 763-783.

[3]. Boyer, E. New England Journal of Medicine, March 17, 2005; vol 352: pp 1114-1120.[4]. ADRIENNE Z. ABLES, PharmD, and RAJU NAGUBILLI, MD, Spartanburg Family Medicine Residency

Program, Spartanburg, South Carolina, American Family Physician. 2010 May 1;81(9):1139-1142.[5]. Brown TM, Skop BP, Mareth TR. Pathophysiology and management of the serotonin syndrome. Ann

Pharmacother. 1996;30(5):527–533. [6]. Sternbach H. The serotonin syndrome. Am J Psychiatry. 1991;148(6):705–713.

Page 113: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

111

Chapter 11

Contemporary Profi le of Toxicological Morbidity at MMA in 2012, Sofi a

KONOV Valentin, NEYKOVALyudmila, KANEV KamenClinic of “Emergency Toxicology” MMA-Sofi a, Bulgaria

Abstract:The clinical toxicology deals with the problem of acute exogenous intoxications and toxoallergic reactions. Its relevance is increasing in the modern environment of social, environmental and demographic disharmony in terms of global recession and rising uncertainty.

The hospitalized patients in the clinic during 2012 were 1367 of whom 813 / 59.5% / were men and 554 / 40.5% / were women. 132 / 9.6% / were hospitalized more than once. 176 patients, including 112 men and 64 women were uninsured, of whom few were foreigners. A higher absolute number of Toxoallergic reactions among women in the age group between 31 and 35 years was noticed. For men, the leading toxic Knox was ethanol and 48 / 13.4% / of them had more than one hospitalization. The proportion of patients with Psycho Active Substances poisoning mainly among the young men is alarmingly increasing. The proportion of the toxic effects of benzodiazepines in young women was relatively high. A considerable number of the toxic effects was caused by noxious insects, animals, food and household products. The average duration of hospitalization was 3.7 days.

Working in conditions of emergency makes it necessary to make a diagnosis only based on scanty history, physical examination and minimum amount of clinical examinations to start specifi c and often life-saving therapeutic intervention. We therefore believe that the data on the toxicological morbidity will be useful for timely and quality treatment and prevention of acute exogenous intoxications and Toxoallergic reactions.

Key words: toxicology, acute intoxications, morbidity, distribution

Introduction

Clinical toxicology examines the problems of acute exogenous intoxications and toxoallergic reactions. It is becoming increasingly relevant in the modern environment of social, demographic and environmental disharmony.

The acute poisonings are characterized by acute onset of pathological abnormalities in the physiological functions of the human body under the impact of exogenous poisons. They show fast dynamics, sometimes nonspecifi c clinical manifestations and have unpredictable end, which makes it necessary to act with urgency.

This requires a timely diagnostic guided only on the basis of scarce medical history, physical examination and minimum amount of paraclinical examinations in order to start specifi c and often life-saving therapeutic intervention. These objective circumstances require toxicologists to have high professional competence and clinical thinking.

Page 114: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

112

AimThe aim of this study is to present the structure of morbidity in the clinic of “Emergency

Toxicology” at MMA in Sofi a for a one-year period.

Material and MethodsThe methods used are clinical observation, retrospective and graphical analysis.

ResultsThis review presents a retrospective morbidity study in “Emergency toxicology”clinic

at the Military Medical Academy in Sofi a, for 2012. A total of 1367 (100%) patients were hospitalized - 554 ( 40.5% female) and 813 (59.5% male). Of them 1028 (75.2%)used clinical pathway(C.P.) N 293- 370 (36%) women and 658 (64%) men. The remaining 339 (24.8%) patients used C.P.N 291. - 184 (54.3%) women and 155 (45.7%) men. The total number for 2012, with two or more hospitalizations was 132 (9.6%) patients- 45 (3.3%) women and 87 (6.3%) men. Uninsured were 176 (12.9%) - 64 (36.36%) women and 112 (63.64%) men. With C.P. 291 uninsured were 8 (4.54%) patients - 5 (62.5%) women and 3- (37.5%) men. Of the remaining 168 (95.46%) patients with C.P. 293 - 59 (35.12%) were women and 109 (64.88%) were men. With C.P. 291 there were no foreigners and patients with more than two hospitalizations, while by the C.P. 293 two women and six men were hospitalized more than twice, as well as 3 foreigners who were treated for a fee. The data is presented in Table 1.

Table 1. Patients distribution by sex, clinical pathways, number of hospitalizations and insurance status

Sex C.P. 291 C.P. 293 Two or morehospitalizations Uninsured Total

Women 184 /54,3 %/ 370 /36%/ 45 /3,3 %/ 64 /36,36 %/ 554 /40,5%/

Men 155 /45,7 %/ 658 /64%/ 87 /6,3 %/ 112/63,64 %/ 813 /59,5%/

Total 339 /24,8 %/ 1028 /75,2%/ 132 /9,6 %/ 176 /12,9 %/ 1367 /100%/

Table 2 Distribution by sex and age groups in clinical pathways 291 and 293

Age group C.P. 291 C.P. 293Women Men Total Women Men Total

Up to 20 13 4 17 14 18 3221 to 25 20 13 33 31 41 7226 to 30 13 13 26 44 65 10931 to 35 23 13 36 48 84 13236 to 40 10 16 26 43 91 13441 to 45 11 11 22 22 76 9846 to 50 13 17 30 31 80 11151 to 55 11 17 28 27 64 9156 to 60 15 18 33 30 56 8661 to 65 18 11 29 23 31 5466 to 70 6 8 14 19 20 39

Page 115: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

113

71 to 75 17 5 22 12 11 2376 to 80 8 7 15 8 8 1681 to 85 3 2 5 4 3 786 to 90 3 0 3 4 1 5Over 91 0 0 0 1 0 1Total 184 155 339 361 649 1010

With C.P. 291 the prevalent morbidity was among women with a peak at the age-group 31-35, which coincides with the highest level of overall morbidity. In men the morbidity pre-dominated in two age groups: 56-60 and 46-55.

With CP 293 among men predominated the morbidity, with a peak in the 36-40 age-group which coincided with the highest level of total morbidity for both sexes. In women predominant was the age group of 31 to 35 years.

Table 3 presents the distribution by sex and diagnostic sub-groups according to ICD 10 of patients with toxoallergic reactions treated in the clinic withC.P. 291.

Table 3. Distribution by sex and subgroups by C.P. 291

Sex Т 78.0 Т 78.1 Т 78.2 Т 78.3 Т 88.2 Т 88.6 Total by c.p. 291

Women 2 20 2 158 0 2 184

Men 1 30 3 119 1 1 155

Total 3 50 5 277 1 3 339

The largest number of patients from both sexes were with a diagnosis of “Angio edema”, with the prevalence of young women. On table 4 we present the distribution by sex and diagnostic sub-groups according to ICD 10 of the patients with acute poisonings treated in clinic by CP 293, with more than 10 hospitions for the year.

Table 4. Distribution by sex and diagnostic sub-groups according to ICD 10

Sex Т40.0чТ40.9

Т42.0чТ42.8

Т43.0чТ43.8

Т51.0ч Т51.8

Т54.0чТ55

Т59.0чТ59.8

Т60.0чТ 60.8

Т62.0чТ62.8

Т63.0чТ64

Т65.0чТ 65.8

Total by C.P. 293

Women 17 35 14 121 25 5 20 36 42 21 336

Men 72 21 10 258 11 19 23 47 55 19 635

Total 89 56 24 479 36 24 43 83 97 40 971

The largest number of patients were with alcoholic poisoning, followed by toxic ef-fects caused by :poisonous animals, poisonous drugs and psycho-dyspeptic substances, food poisonings, antiepileptics, sedatives, hypnotics and anti-Parkinson medicines. Among the men, the leading toxic noxa was ethanol followed by psychoactive substances, poisonous animals and food poisoning. Alarmingly high was the proportion of patients with alcohol poisoning with more than one hospitalization: of total 57 patients 48 were men. The psychoactive substances in young men predominated. Relatively high was the

Page 116: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

114

proportion of benzodiazepines toxic effects in young women, followed by intoxication by household cleaning substances. The average duration of treatment by C.P. 291 was 3.5 days, and by C.P. 293 was 3.9 days. The overall median treatment duration in both clinical pathways was 3.7 days.The analysis of the data from our study leads to the following main.

Conclusions:

1. In the allergic reactions predominant were women in younger age.2. Prevalent morbidity among men was the alcohol intoxication, while multiplicity of

hospitalizations and lack of health insurance was observed.3. Most common in both sexes were the acute alcohol intoxication, observed more

often among men.4. We believe that the data on the trend of contemporary toxicological morbidity will

be useful for the timely, adequate and qualitatively treatment but also of acute exogenous intoxications and toxoallergic reactions prevention.

Page 117: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

115

Chapter 12

Clinical Case of a Welder Chronic Intoxication with Metal Aerosols

Tania KUNEVA, Diana APOSTOLOVA, Tsvetanka DIMITROVA, Vladimira BOYADZHIEVA, Vera PETKOVA

UMHAT “St. Iv. Rilski”Medical University-Sofi a, BULGARIA

Abstract.Relates to a woman, who worked as a welder in a factory for heavy machinery for 27 years. During the period she had a direct contact with welding gases such as NO2, CO, manganese aerosols and iron dust, all of which exceeding above-norm concentration. The patient is diagnosed with chronic manganese intoxication exhibited with extrapyramidal syndrome in tremor form, pneumoconiosis of the welder,and chronic asthma bronchitis. Acceptance of occupational etiology of the discovered diseases is done after a thorough examination of the factors exhibited at work site and the expert evaluation of Territorial Expert Medical Commission. The patient is under dynamic monitoring and is treated in the Department of Occupational Diseases – Sofi a for 10 years. The data from the tremorogram shows static postural and intentional tremor with frequency 7-8 Hz from badly grouped signals of muscular activity. Despite the exposition is terminated, a progressive development of symptoms is noted in regards to generalization of the tremor, development of shortness of breath and cor pulmonale.

Key words. metal aerosols, pneumoconiosis, manganese parkinsonism

IntroductionThe problem with pneumoconiosis from over- exposure to metal aerosols remains

unsolved to date. A number of publications published during the 80’s of XX century are too contradictory in terms of the clinical, X-ray and functional manifestations of siderosis [7, 12]. Until recently, the pulmonary siderosis was believed to be a benign condition unrelated to respiratory symptoms [6, 7, 12]. The review based on various literature proves this conclusion to be incorrect and fi nds siderosis to cause symptoms and functional changes [4, 5, 8, 13]. In recent years, there is information for increased sickness and mortality rate from lung cancer in workers exposed to metal aerosols [11].

Professional risk factorsThe patient is treated and dynamically monitored in the Department of Occupational

Diseases – Sofi a, for a period of 10 years. The patient is a woman who worked for 27 years as a welder in a factory for heavy machinery (protocol № 143/ 08.06.2006 NHI for conducted study of an occupational disease).

Corresponding Author: Tania Kuneva, Iv. Geshov 15, 1431 Sofi a, Bulgaria; E-mail: [email protected]

Page 118: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

116

The welder perform arc welding of complex parts made of different metals (ferrous high and low alloy steels and non-ferrous) and alloys with portable arc welding machine with different electrodes and welding equipment.

According to the production characteristics, a danger of exposure to chemicals on the work fi eld is identifi ed (manganese aerosols, nitrogen oxides and CO; iron powder) exceeding the normative values.

Protocol for control of the toxic substances № 01410/27.07.2004 and protocol for control of dust levels №01411/27.07.2004. The toxic substances and dust exceed the normative values when heated.

Risk factors on work fi eld Measured Norm

Ferrous dust 23,3 mg/mз 4 mg/mз

Manganese 0,48 mg/mз 0,3 mg/mз

NO 2,5 mg/mз; 2 mg/mз;

Physical loadForced working poses

Anamnesis: Complaints began ten years ago when she noticed shaking of the head and both hands , increasing with emotional tension and stress. Initially treated with antidepressants without signifi cant effect. A progress in the clinical symptoms was noted in the following years – tremors became frequent, gait – slower, movements – more diffi cult.At the same time cough appeared, diffi cult expectoration, and later - fatigue with minor physical exertion, tingling and wheezing, paroxysmal dyspnea, sweating and palpitations.

Past diseases: 1988 - hypertension, 1988 and 1996 – bronchitis, 2004 - attacks of bronchial asthma, 2006 - welder pneumoconiosis (siderosis). Chronic asthmatic bronchitis. Parkinson sindrom.2008 - CAD UA Hypertension II st

Does not smoke and does not drink, no anamnesis for family diseases. Status: poor general condition, contactable, hipersonoric percussionphenomenon,

decreased respiratory mobility, bilatteral weakened vesicular breathing, diffusely scattered dry wheezing, rhythmic heartbeat RR140/100, liver and lien -normal.

Neurological status: – active and passive motions in full range with reduces muscle power and slightly increased rigid muscle tonus. Limited dorsal refl exes of the left foot, coordiantion -static, postural and intentional tremor of the hands R>L; legs, head and torso.Gait – slowed down, with limited physiological synkineses. Slightly lowered Achilles refl ex on the left. Sensitivity – surface hyperesthesia in dermatome S1 left.

Tests: X-ray lungs and heart: pneumofi brotic bilateral changes, small nodular opacities bilateral in the base of lungs. Hillus Hilar lymph nodes with calcinosis; cardio vascular system – prolonged arc of the left ventricle

Page 119: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

117

Spirometry

date VC FEV1 /VC FEV1 FEF 50

06.04.2006 69 46 57

09.03.2007 56 67,6 48 27

13.06.2007 44 62,3 53 33

30.05.2008 66 56,3 47 34

23.02.2009 62 66,9 53 27

10.05.2013 47 78 47 21

Blood gases

date рН(7,36-7,44)

рСО2( 4,66-5,99) кРа

рО2(10,3-13,3) кРа % sat O2

06.04.2006 7,43 4,32 8,1 92

09.03.2007 7,39 5,05 8,5 92,1

13.06.2007 7,42 4,67 7,2 88,7

30.05.2008 7,44 4,35 8,5 93,2

23.02.2009 7,4 4,4 8,8 93,9

13.12.2012 7,42 4,2 8,3 87,7

10.05.2013 7,43 4,5 7,2 89,1

Tremo graph: static postural and intentional tremor with a frequency of 7-8 Hz poorly grouped bursts of muscle activity, sometimes with alternating pattern.

Hematological results, erythrocytes sedimentation rate, erythrocytes morphology, glucose, creatinine, uricacid, urea, cholesterol, triglycerides, liverenzymes – innormalrange.

Cutaneous-allergic tests – home dust, pollens, bacteria and others – negative.Coresilin /+++/; Acetone /++++/, white Alkyd paint / ++++/

Exposure biomarkers

datesMn blood

(0,22-0,90) μmol/l

Mnurine(0,0-0,30)

μmol/l

MnEDTA

Pbblood<1,19μmol/l

Pburine<-0,1

μmol/l

Pb μmol/lEDTA

06.04.2006 0,15 0,26 0,34 0,33 0,04 0,26

13.06.2007 0,26 0,20 0,39 1,26 0,10 0,85

10.05.2013 0,24 0,35 0,37

Page 120: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

118

Diagnosis:Welder pneumoconiosis (siderosis). Chronic obstructive bronchitis with frequent

exacerbations and chronic respiratory insuffi ciency. Chronic manganese intoxication moderate extrapyramidal syndrome manifested by tremor form.

Discussion:The patient is diagnosed with chronic intoxication with toxic aerosols and powders

(Mn, NO2, iron powder) that have occurred in the context of 27 years of service as welder. The intoxication is manifested by chronic nasopharyngitis, welder pneumoconiosis (bilateral pneumofi brosis – small nodular opacities), chronic obstructive bronchitis, expressed parkinsonian syndrome (extrapyramidal syndrome tremor form). So far we have not encountered this combination of syndromes in a single individual. In literature most often described pneumofi brosis changes are in welders[ 4,5,6 , 9,12 ]. Liubchenko and Vinnitskaia reported 6 professional lung diseases among surveyed welders as the most common is siderosis, followed by sensorineural deafness, toxic dust bronchitis and chronic manganese intoxication. In our presented clinical case prolonged exposure to manganese aerosols is the reason for the simultaneous monitoring of chronic nasopharyngitis, pulmopatiya and Parkinson’s syndrome. Determination of Mn in blood and urine has no intrinsic value for the diagnosis because of the short circulation of manganese in the blood and its minor elimination in the urine [2,3]. However, these indicators are used as biological indicators of exposure and support the diagnosis. The patient is also observed with although mild increase in biological fl uids and an increased excretion after antidotal therapy CaNa2 EDTA. Our results coincide with some studies in which it was found that the welding work is associated with obstructive airways disease .

References

[1]. DinkovaKr., Metalconioses (siderosis) in Labor Medicine ІІ, prof. D. Tsvetkov S, pb. „St. Cl. Ohridski” 2007, 577-578.

[2]. PetkovaV. ManganeseinLaborMedicine ІІ, prof. D. Tsvetkov S, pb. „St. Cl. Ohridski” 2007, 456-457.[3]. PetkovaV. ManganeseinOccupationalDiseases, V. Kostova, V. Petkova, S, pb RAL KOLOBYR, 2007,30-33.[4]. PetrovaE – WelderPneumoconiosisOccupationalDiseasesfromNonorganicDust, InvestpresAD. S, 2005,

137-144.[5]. PetrovaEWelderPneumoconiosis. Scriptaperiodica,2005, Vol.VIII, 2, 10-19.[6]. BillingsCGHowardPOccuppationalsiderosisandwelderslung: areview.MonaldiArchChestDis 1993, 48 (4)

304-14.[7]. GuidottiTL, etallArcWelderspneumoconiosis: application of advanced scanning electron microscopy. Arch

Environ Health 1978,33, (3)117-24.[8]. Kujawska A, Marek K Radiologikal picture of pneumoconiosis in welders Med Pr, 1979,30(1) 79-84.[9]. LaDouJ., Occupational МedicineAppleton&Lange, California,USA,1990.[10]. Liubchenko PN, Vinnitskaia TE – The structure of occupational morbidity in electric welders Med Tr Prom

Ekol 2000, (8): 7-10.[11]. Schneider WD et all – Functional signifi cance of lung siderosis in electrode weldwrs Z Gesamte Inn Med

1987, 42, (5)126-30.[12]. Stern RM. Process-dependent risk of delayed health effects for welders. Environ Health Perspect, 1981,

41, 235-53.[13]. Steurch F, Feyerabend R Sidero-fi brosis of the lung after decades of arc welding. Pneumologie, 1997, 51,

(6):545-9.

Page 121: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

119

Chapter 13

Health Risk Assessment of Exposure to Chemicals Among Miners

Karolina LYUBOMIROVAa1, Zaneta RATCHEVAb

a Department Occupational health, Faculty of Public Health, Medical University Sofi a, Sofi a, BULGARIA

bOccupational Health Service- Mini Maritza Iztok- LTD, St.Zagora, BULGARIA

Abstract: Mining and especially openmining is a traditional economicactivity in Bulgaria, which has been developed forcenturies. Though the main health risk factors at the work places are known, in the last twenty years there is a lack of information about the working conditions and the health status of miners in Bulgaria.Object and methods: A study on the health status of 7500 miners from the biggest open mine in Bulgaria for a 6 years period was carried out. The levels of exposure of the main risk factors – noise, vibrations, chemical substances, and microclimate – were assessed and analyzed.The health status of miners was analyzed though the data from the pre- and periodic check up examinations, work accidents and temporary disability data.Results:About 400 miners from the investigated group were found to be exposed to chemical substances above the Total Limit Values. The main occupations were vulcanizers and welders. The exposure to tricholoethylene ( up to 300 times TLV), Fe, CO, toluene, xylene, acetone, NO2, and benzene was registered.The non observance of the technology requirements, lack of effective ventilation and the bad working organization at some workplace resulted in the registered increased exposure to chemicals.Conclusions: The analysis of the exposure to chemical substances among open miners in Bulgaria has shown a trend to decrease in the observed period. However a combined exposure among some occupations should have in mind.

Key words: open coal mining, chemical substances, health risk assessment, health risk management.

Introduction:

Open coal mining is related to specifi c risks at the workplaces which infl uence the health of employees. From the ancient times mining activities were related to the so call “coal- miners’ disease” due to the exposure to dust. Later numerous large scale studies were performed for investigation of the correlation between the health effects among miners and

1 Corresponding author: Prof. KarolinaLyubomirova, Department Occupational health, Faculty of Public Health, Medical University Sofi a, 8 Bialo more str., 1527 Sofi a, Bulgaria, Tel: + 359 2 9432542, e-mail: [email protected]

Page 122: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

120

the magnitude of the exposure [1]. The improvement of the technology and the decrease of the dust concentration have lead to impairment of the lung function and pneumoconiosis’s prevalence in different countries [3,5,6].

Though the decrease of dust concentration, other workplace risk factors still exist which might infl uence the health of miners such as noise, vibration, chemical substances, unhealthy microclimate, exposure to ultraviolet radiation (for open coal mines).

In Bulgaria coal mining has been developed since the ancient times as well. One of the fi rst studies on the health of miners and the relationship with the working conditions was performed by Litchev in 1909 [4]. Later this study was extended by prof. Vesselin Borisov who has put the foundations and has implemented into the practice of the modern approach for health monitoring among the coal miners in Pernic region [2].

After that other researches were performed in coal mines in Bulgaria. Different models and methods for exposure reduction were developed in order to decrease the infl uence of the risk workplace factors on health of miners. After 2000 however, due to the transformations of the health system in Bulgaria and changes in the health and safety legislation, signifi cant changes has been done in health monitoring and health service of the employees. The complex monitoring of the health and work ability of workers has stopped and a lack of information about the health of coal miners appeared. This was the reason for a special planned study on a big representative group of open coal miner in Bulgaria.

Materials and methods

The object of the survey was a group of 7500 coal miners working in three mining areas and the administrative stuff of the company. The data of the health monitoring for a six years period (2006-2011) were presented.

The following risk factors at the working environment were assessed:1. Microclimate in hot and cold season- using a combined equipment “TESTO 445”2. Noise assessment through measurement of the peak noise values, mean daily noise

values and mean weekly noise values.3. Vibration control through measurement of the mean daily values to common and

local vibrations 4. Dust control through personal sampling and measurement of the concentration of

inhalable and respirable dust fraction. The concentration of free crystal SiO2 was also had in mind and compared to the limit values.

5. Measurement of the concentration of chemical substances in the ambient air through gas chromatographic and mass spectrometric analysis.

The health status of the employees was assessed by the results of the annual periodic check up examinations, personal health dossiers, work accidents.

The data were analysed by Alternativeanalysis - intensive and structural indicators; Variation analysis – averagevalues, comparison; Correlation analysis - correlation and dispersion coeffi cients; Graphical and table analysis– sorting the data in complex tables, pie charts, linear charts and collumn diagrams and fi gures.

Page 123: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

121

ResultsThe following health risk factors were identifi ed through the monitoring of the

working environment in the coal mine: noise and vibrations, exposure to dust and chemical substances, unhealthy microclimate. The noise control measurement showed that the noise level exceeded the limit values in the range 81-103 dB(A). The exceed of the daily exposure limit of 87 dB(A) was registered in 23% of the measurements.

The limit values of vibrations were exceeded in the cabins of bulldozers, excavators and autograders in 82% of the control measurements and in 77% of the measurements at the operation stations.

The exceed of the dust concentrations out of the cabins was from 5,4 to 70,1 mg/m3 inhalable dust (1.1 to 10.5 times the total limit value) and 1,4 to 9,4 mg/m3 respirable dust (1.2 to 4.8 times the TLV).

Due to the specifi ty of the open coal mining, the microclimate was strongly related to the atmospheric conditions. The monitoring showed inconformity with the microclimate parameters in 47% of the measurements due to high temperature. In the cold period of the year the inconformity was registered in 18% of the measurements, as well as in 15% of the measurements of the speed of wind.

The analysis of the working conditions revealed that 400 employees were exposed to chemical substances , many of them above the limit values. The main pollutants of the working environment are presented at Table 1.

It should be noted that at some workplace the employees were exposed to a combination of risky factors such as: microclimate and noise (mechanic of mining machines, electro mechanic), microclimate, noise and vibrations (machine operators of TTM-GLM, bulldozer operators), Microclimate, noise and dust (coal miners, mechanic of mining machines), microclimate and chemicals (welders, vulcanizers, accumulators’ workers),

The health status of the employees was assessed by the results of the periodic check up examinations performed by medical doctors and the temporary disability data.

The analysis of the demographic characteristics of the employees showed that 85% of them were men and 15%- women. The age distribution of the employees showed that the predominant part of the employees was below 45 years old, which presumed a better health. In 2006 63,64% of all were below 45 years old, in 2007 - 62,76%, in 2008 - 61,29%, in 2009 - 60,51%, in 2010 - 59, 89% and in 2011 - 59, 51%.

As a result of the periodic check up examinations, more than 60% of the examined employees were diagnosed with diseases. Health problems were registered among 64,4 to 79,03 per 100 employees which were higher than the mean accepted value in Bulgaria (62,0 per 100 employees).

The point prevalence for the investigated period reached 214,9 per 100 employees which meant that more than one disease was diagnosed among the examined people.

The analysis of the results of the periodic check up examinations showed that the diseases with the highest prevalence among the monitored coal miners were the diseases of the blood and circulation (20-30%), bones and muscles (8-16%), lungs (7-12%), eyes (21-41%).

Page 124: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

122

Table 1. Occupations exposed to chemical substances in the investigated coal-mining enterprise.

Occupation Chemical substance Concentration (Mg/m3 )

Vulcanizer Trichloroethylene 400 – 600 ( K*= 3 - 4,4)

CO 75- 150 (K=1,2- 3,8)

Electric welder Mn 0,35- 1,3 (1,1- 2,6)

Fe 5,4 - 8,3 (K= 1,1- 1,9)

Oxygen and gas welder Mn 0,35- 3,2 (K= 1,1- 10,7)

Electric fi tter Benzene 1117,2 (K=1,2)

Accumulator worker H2SO4 1,35 - 2,4 (K= 1,3- 2,4)

Diesel engine fi tter NO2 8- 9,5 (K=2,4)

Dyer engines Toluene 142,5

Xylene 287,5 (K=1,3)

Acetone 620

Turner Pb 0,17 (K=3,4)

Mn 0,55 ( K=0,55)

Milling-machine operator Pb 0,12 (K=2,4)

Blacksmith Pb 0,1 (K=2,0)

* K- exceed of the TLV

Discussionand ConclussionsA study of the health status of a representative group of 7500 coal miners was

performed in Bulgaria. The main risk factors at the workplaces were identifi ed such as noise, vibrations, dust exposure, exposure to unhealthy microclimate.

The analysis of the working conditions revealed that 400 employees were exposed to chemical substances above the limit values- mainly welders and vulcanizers.

The risky workplaces - sources of air pollution with chemical substances were identifi ed such as welding, vulcanizing, batteries’ fi lling, dying, diesel engines’ fi tter, coal mining. The

The non observance of the technology requirements, lack of effective ventilation and the bad working organization at some workplace resulted in the registered increased exposure to chemicals.

The analysis of the 6-years-period data revealed a trend of decrease of the concentrations of the chemical substances at the workplaces in the investigated coal mine. However there are still numerous workplaces and occupations where the concentrations of the substances are 2-3 times higher than the TLV.

The analysis of the point prevalence of the diseases showed that 41% of the coal miners suffered from eye disorders, 30%- from blood and circulatory diseases, 16%- from muscular-skeletal diseases, 10%- from diseases of the neuron system and 12%- from lung diseases. As more than 50% of the employees were below 45 years old, the percentage of the diagnosed diseases was higher than that of the common population in Bulgaria.

Page 125: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

123

Undoubtfully these results due to the infl uence of the long term exposure to the specifi c risk factors at the open coal mining. It is important that the registered point prevalence was above 100 per 100 employees which meant that more than one disease was diagnosed per coal miner. The analysis of the temporary disability data revealed prevalence of lung diseases ( up to 36%), infectious diseases ( up to 11%), neuron diseases (up to 12%), circulatory diseases ( up to 16,5%), muscular-skeletal diseases (11,3%), traumas ( up to 26%), etc. All these fi nding correlated positively with the increase of the age and the specifi c length of employment. These data confi rmed the infl uence of the identifi ed risk factors in the open coal mines on the health of the employees. Based on these results special risk reduction measures were implemented into the practice and health promotion programs were developed.

The health structures developed in the investigated coal mine functioned and followed the health and safety legislation. However their optimal capacity was not reached yet. There was a need of integration and analysis of the health information derived from all the structures. A complex study of the health status of workers should collect information from the periodic check exams, temporary disability data as well as from the common morbidity data.

A health monitoring model was developed as a result of the presented study. The implementation of the complex approach in health status research is a prerequisite for development of health risk management programs among groups of employees at risky workplaces. The results of these long term program will be monitored in a few years.

References

[1]. Attfi eld MD (2010). Kuempel ED. Erratum to “ mortality among U.S. underground coal miners: A 23-year follow-up”. American Journal of Industrial Medicine. 53(5):550

[2]. Borisov V (1971). PhD Thesis “Health status investigation among coal miners in Pernic region” (in bul), Sofi a.

[3]. Kimura K. Ohtsuka Y. Kaji H. Nakano I. Sakai I. Itabashi K. Igarashi T. Okamoto K (2010). [4]. Progression of pneumoconiosis in coal miners after cessation of dust exposure: a longitudinal study based

on periodic chest X-ray examinations in Hokkaido, Japan. Internal Medicine.49(18):1949-56. [5]. Litchev D(1905), Mining and pneumoconioses in Bulgaria ( in Bul), Sofi a.[6]. Suarthana E. Laney AS. Storey E. Hale JM. Attfi eld MD (2011). Coal workers’ pneumoconiosis in the

United States: regional differences 40 years after implementation of the 1969 Federal Coal Mine Health and Safety Act. Occupational & Environmental Medicine. 68(12):908-13.

[7]. Yang MD. Wang JD. Wang YL. Guo PS. Yao ZQ. Lu PL. Gu XY. Dong YL. Lu MX. Zhu P. et al.(1985) Changes in health conditions in the Huainan coal mine in the past three decades. Scandinavian Journal of Work, Environment & Health. 11 Suppl 4:64-7.

Page 126: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

124

Chapter 14

Methadone Poisoning – Clinical and Psychosocial Constellations

Julia RADENKOVA - SAEVA., Anelia LOUKOVAToxicology Clinic, UMHATEM “N.I.Pirogov”, Sofi a, Bulgaria

Abstract. Methadone is a long-acting synthetic opioid - a drug which is used for treatment of opioid dependence, and acute pain..However, methadone maintenance programmes are established in many countries as the treatment of choice in opiate addiction, methadone is now well recognized as signifi cant poison. Overdose of methadone may cause disturbance of consciousness, hypotension, respiratory depression, death.We performed a study of hospitalized in the Clinic of Toxicology - UMHATEM “N.I.Pirogov” patients for acute poisonings with methadone for the period 01.01. 2011- 31.12.2011.Patients were followed with regard to the severity of poisoning, level of consciousness, the cause of intoxication, complications, therapeutic approach, and various psychosocial aspects. Attention is paid to the patient’s motivation for committing suicide with methadone.Best results were achieved when treatment was reported by a multidisciplinary team with individualization for any particular patient.

Key words: methadone, acute poisonings, psychosocial aspects, multidisciplinary team

Introduction Methadone is a synthetic opioid, which is used for treatment of opioid dependence,

and acute pain [1, 11, 15]. As substitutive treatment for addicts, methadone was fi rst used in the late 50’s in Canada and in the 60’s in the USA. In Europe, the fi rst methadone programs introduced in the late 60’s - in Sweden , Britain , the Netherlands and Denmark, in the 70’s - Finland, Portugal, Italy and Luxembourg, to 80’s in Austria and Spain and in the 90’s - in Germany, Greece and France. Currently, about 500 000 people in Europe are being treated with methadone [4, 7, 9]. In Bulgaria, the fi rst methadone program was discovered in 1995 in Sofi a - National Addiction Centre.

Aims of the Methadone Programme: To reduce illegal or other drug use; to improve health and well-being; to reduce transmissions of blood borne disease spread by needle sharing, since Methadone is not given intravenously; to reduce deaths from Opiate use; to reduce drug-related crimes; to improve social functioning; to improve economic status, since Methadone (unlike heroin) is more cheaply. Methadone treatment makes it possible to stop drug use [8].

Page 127: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

125

However, мethadone maintenance programmes are established in many countries as the treatment of choice in opiate addiction, methadone is now well recognised as a signifi cant poison in both opiate addicts and in other subjects who are exposed to diverted methadone.Since the start of methadone programmes and the increase in prescriptions for methadone, reports have appeared detailing methadone deaths [2, 5].

Aim: To present the results of one - year study of characteristics and severity of acute methadone poisoning in Toxicology Clinic, Emergency University Hospital “Pirogov”.

MethodsThe study includes 10 patients with acute methadone poisoning, hospitalized in the Clinic,

for the period 01.01.2011- 31.12.2011. The patients were followed-up with regard to general condition, psychiatric state,comorbidity, and complications. Attention is paid to the patient’s motivation for committing suicide with methadone. The methods used include: clinical observation and examination, together with laboratory, imaging and psychiatric methods.

Results10 patients between the ages of 20 and 35 with acute methadone poisoning have been

observed. 7 were male and 3 female. Accidental poisonings occurred in 7 of the cases, while intentional self-poisonings were registered in 3 patients. Method of admission was oral in 8 patients and intravenous in 2 of them. In MMTP included six people.The intoxications were monotoxic in 9 persons, in 1 patient methadone were combined with Clozaril (Leponex) and Rivotril (see Table 1).

Table. 1. Patients – distribution by gender, age, method of admition, motivation

N Gender Age Method of Admission

Combination with other substances

MotiveIncluded in

the methadone program

10 7 male /3 female 20-35 Рer os – 8

I.V. - 2 Leponex andRivotril - 1

ТS - 3Overdose – 6Curiosity - 1

6

The severity of poisonings varied from moderate to extremely severe. The clinical presentation of methadone overdose is characterized by a triad ofcentral nervous system depression, respiratory depression, and miosis. Coma was identifi ed in 7 persons, soporin 2, and somnolence in 1 of them. Respiratory depression on admission was registeredin 8 patients. Miosis was registered in all patients. The hospital course was complicated by anaspiration in 2 persons, rhabdomyolysis, and subsequent acute renal failure – in 1, peripheral damage to n. femoralis dextra – in 1 person. Accompanying comorbidity occurred in 3 patients: affective disorder in1, depressive syndrome in 2 of them (see Table 2).

Table 2. Clinical characteristics

Consciousnes Respiratory failure on admission

Comorbidity Complication

Coma – 7 Sopor – 2Somnolence - 1

8 Affective disorder -1Depressive syndrome - 2

Aspiration-2 Rhabdomyolysis, ARF, Peripheral damage to n. femoralis dextra - 1

Page 128: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

126

TreatmentThe patients were managed with enhanced dieresis, antidotes – naloxone, notropil,

gastrointestinal decontamination with activated charcoal, symptomatic agents, antibiotics; vitamins.

OutputAll patients were discharged clinically healthy.

Discussion Methadone hydrochloride is a synthetic opiod, whichexhibits at least three different

mechanisms of action including potent opioid agonism, N-methyl-D-aspartate antagonism and monoaminergic effects.

This, along with methadone’s excellent oral absorption, high bioavailability, long duration of action and low cost, make it a very attractive option for the treatment

Peak blood levels after oral ingestion occur at 2 to 6 hours. Because of signifi cant protein binding (>90%), levels are constant over 24 hours.

In tolerant individuals, the half-life ranges from 13 – 47 hours (average 25 hours)The toxicity of methadone depends on the amount consumed and the tolerance of the

individual.In non-tolerant individuals, 10 mg is enough to kill a child and 50 mg is enough to

kill an adult [3].All patients with a signifi cant methadone overdose should be admitted to the hospital

for at least 24 hours and watch for the development of CNS or Respiratory depression, Non-cardiogenic pulmonary edema [2].

Monitoring: Check frequently for vital signs, respiratory rate and O2 sat, and hold a brief conversation to assess alertness.

ECG and cardiac monitoring are recommended to check for prolonged QT interval and ventricular arrhythmias ( methadone can cause torsades de pointes) [10, 12].

Clinical features1. The clinical presentation of methadone overdose is that of a gradual onset that

is prolonged.2. Symptoms begin up to 10 hours after the overdose!3. Early symptoms include nodding off, drowsiness, slurred speech and emotional

lability. 4. Respiratory depression occurs later!There is a triad of respiratory depression, central nervous system depression and

pin point pupils [2, 10, 13].

TreatmentIf above occurs, intubation is necessary followed by a Naloxone infusion (admission

to the ICU is necessary).

Page 129: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

127

Naloxone is a safe treatment in patients who are not physically dependent on opioids (e.g., a patient not in methadone therapy who took methadone at a party).

For methadone- or opioid-dependent patients, intubation avoids risks of naloxone-induced withdrawal.Intubation is necessary when hypotonia, hypercapnea are present and desaturation persists despite supplemental oxygen, or if the patient fails to respond to naloxone within two minutes [16].

Psychosocial supportSpecial cognitive-behavioural interventions might help to reduce additional

consumption of psychoactive substances. Psychosocial counseling can support patients with structuring their life again, based

on changed values, because the pressure to fi nd drugs is reduced substantially. Family involvement is crucial for the successful treatment while its dynamics might

only be understood and confronted with expert psychological support [6, 14].

ConclusionsFor more than 30 years, the synthetic narcotic methadone has been used to treat heroin

addiction. Methadone is a dangerous even fatal drug, binds to the same receptors as heroin but

without producing the euphoric rush. Methadone is a medication that is prescribed by a specialist. The overdose may result in fatal exit, or severe chronic disability of various organs

and systems.In acute intoxications best results are obtained when conducting treatment by a

multidisciplinary team and individualization for the individual patient.

References

[1]. Behrendt K, Chorzelski G, Meyer-Thompson HG: Gibt es einen „Goldstandard” der Substitutionsbehandung Methadon/Polamidon? (Is there a, gold standard’ for Maintenance Treament with Methdone / Levomethadone?) Suchttherapie, 2006, 7:78-81.

[2]. Corkery JM, F Schifano, A. Hamid Ghodse, A Oyefeso, The effects of methadone and its role in fatalities, Human Psychopharmacology: Clinical and Experimental, 2004. Vol 19, 8, 565–576,

[3]. Eap CB, Buclin T, Baumann P, Interindividual variability of the clinical pharmacokinetics of methadone: implications for the treatment of opioid dependence, Clinical pharmacokinetics, 2002, 41 (14): 1153–93.

[4]. European Monitoring Center for Drugs and Drug Addiction (EMCDDA):Annual Report 2006. The State of the drugs problem in Europe. Lisbon 2006.

[5]. Faggiano F, Vigna-Taglianti F, Versino E, Lemma P, “ Methadone maintenance at different dosages for opioid dependence”. In Faggiano, Fabrizio. Cochrane Database of Systematic Reviews, 2003 (3): CD002208.

[6]. Gerlach R., H.Stцver:Begleitende psychosoziale Unterstьtzung in der Substitutionsbehandlung(additional psychosocial support in substitution treatment). 2nd edition. Edited by Beubler E, Haltmayer H, Springer A. Opiatabhдngigkeit. Springer. Vienna; 2007:225-230.

[7]. Gerlach R:Drug Substitution Treatment in Germany: A Critical Overview of its History, Legislation and Current Practice. Journal on Drug Issues, 2002, 2:503-522.

[8]. Joseph H, Stancliff S, Langrod J, “ Methadone maintenance treatment (MMT): a review of historical and clinical issues”. Mt. Sinai J. Med. 2000, 67 (5–6): 347–64.

Page 130: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

128

[9]. Joseph R, Moselhy HF “National survey of methadone prescribing for maintenance treatment: ‘opiophobia’ among substance misuse services?” Psychiatric Bulletin 29, 2005, (12): 459–461.

[10]. Latowsky M, “ Methadone death, dosage and torsade de pointes: risk-benefi t policy implications”. Journal of Psychoactive Drugs, 2006, 38 (4): 513–9.

[11]. Leppert, W, “The role of methadone in cancer pain treatment--a review”. International journal of clinical practice, 2009, 63 (7): 1095–1109.

[12]. Maremmani I, Pacini M, Cesaroni C, Lovrecic M, Perugi G, Tagliamonte A, “QTc interval prolongation in patients on long-term methadone maintenance therapy”. European addiction research, 2005, 11 (1): 44–9.

[13]. Sadovsky R., Anderson IB, Kearney TE, Use of methadone, West J Med, January 2000; 172:43-6.[14]. Scherbaum N, Kluwig J, Specka M, Krause D, Merget B, Finkenbeiner T, Gaspar M:Group psychotherpy

for opiate addicts in methadone maintenance treatment – A controlled trial. Eur Ad Res, 2005, 11:163-171.

[15]. Toombs JD, Kral, LA,” Methadone treatment for pain states”. American family physician, 2005, 71 (7): 1353–1358.

[16]. Yip L, Megarbane B, Borron SW. Opioids. In: Shannon MW, Borron SW, Burns MJ, eds. Haddad and Winchester’s Clinical Management of Poisoning and Drug Overdose. 4th ed. Philadelphia, Pa: Saunders Elsevier; 2007: chap 33.

Page 131: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

129

Chapter 15

Mycotoxins – Adverse Human Health Effects

Julia RADENKOVA – SAEVAClinic of Toxicology, UMHATEM „ N.I.Pirogov”,

Totleben 21, 1606 Sofi a, Bulgaria, e-mail: [email protected]

Abstract: Mycotoxins are toxins, produced by some species of mold and are some of the most toxic substances in existence. They have adverse effects on humans, animals, and crops, that result in illnesses and economic losses. Afl atoxins, ochratoxins, trichothecenes, zearelenone, fumonisins and ergot alkaloids are the mycotoxins of greatest medico-agro-economic importance. Mycotoxicoses are diseases caused by mycotoxins. Exposure to mycotoxins is mostly by ingestion, but also occurs by the dermal and inhalation routes.The diagnosis of mycotoxicoses may prove to be diffi cult because of the similarity of signs of disease to those caused by other agents. Mycotoxicoses often remain unrecognized by medical professionals, except when large numbers of people are involved. The clinicalaspects of mycotoxicoses in humans, and the importance of this class of naturally occurring toxins have been discussed.

Key words: mycotoxins, mycotoxicoses, afl atoxins, ochratoxins, trichothecenes

Mycotoxins are toxic secondary metabolites of fungal phytopathogens - microscopic fungi or molds formed before and during harvesting, or improper storage of cereals. Until now there are established more than 300 different types of mycotoxins. The exposure of these substances on humans and animals is global, independent of geographical and climatic features, which largely determine the formation of mycotoxins. They can be found in peanuts, spices, cocoa, dried fruits, wine, beer, oatmeal, nectars, fruit juices, mousses, purees, baby foods [4, 8, 12, 16].

The adverse effect of moulds and fungi was known already in ancient times. There is historical evidence of mould’s existenceback as far as the time reported in

the Dead Sea Scrolls.The dangerous effect of mold is associated with the legend of the “curse of the pharaoh”. Due to the mysterious deaths of a few members of Howard Carter’s team, who opened the tomb of Tutankhamun, the belief in a curse was brought to many people’s attention. “Death shall come on swift wings to him who disturbs the peace of the King”.Recent studies of newly opened ancient Egyptian tombs found pathogenic bacteria of the Staphylococcus and Pseudomonas genera, and the moulds Aspergillus niger and Aspergillus fl avus [16, 45].

In the Middle Ages, ergotism caused by ergot alkaloids from Claviceps purpurea reached epidemic proportions, hamstring, and killing thousands of people in Europe.

Page 132: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

130

Ergotism was also known as “ignis sacer” (sacred fi re) or St. Anthony’s fi re, because at the time it was thought that a pilgrimage to the shrine of St. Anthony would bring relief from the intense burning sensation. The victims of ergotism were exposed to lysergic acid diethylamide (LSD), a hallucinogen, produced during the baking of bread made with ergot-contaminated wheat, as well as to other ergot toxins and hallucinogens [26].

The term “mycotoxin” was coined in 1962 in the result of an unusual veterinary crisis near London, England, during which approximately 100,000 turkey poults died. This mysterious “turkey X disease” was linked to a peanut meal, contaminated with secondary metabolites from Aspergillus fl avus [2]. Soon, the mycotoxin rubric was extended to include a number of previously known fungal toxins (e.g., the ergot alkaloids), some compounds that had originally been isolated as antibiotics (e.g., patulin), and a number of new secondary metabolites revealed in screens targeted at mycotoxin discovery (e.g., ochratoxin A) [45].

Certain moulds are used in the production of cheese and in the fermentation of beer and wine. Moulds are also used in the production of drugs – antibiotics (e.g. citrinin) [12].

Ergot alkaloids are still used in the treatment of parkinsonism, as prolactin inhibitors, in cerebrovascular insuffi ciency, migraine treatment, venous insuffi ciency, thrombosis and embolisms, for the stimulation of cerebral and peripheral metabolism, in uterine stimulation, as a dopaminergic agonist.

Most fungi are aerobic and are found almost everywhere in extremely small quantities due to the size of their spores. They consume organic matter wherever humidity and temperature are suffi cient. Where conditions are right, fungi proliferate into colonies and mycotoxin levels become high.The production of toxins depends on the surrounding intrinsic and extrinsic environments and the toxins vary greatly in their severity, depending on the organism infected and its susceptibility, metabolism, and defense mechanisms. The chemical structures of mycotoxins vary considerably, but they are all relatively low molecular mass organic compounds. Mycotoxins can appear in the food chain as a result of fungal infection of crops, either by being eaten directly by humans or by being used as livestock feed. Mycotoxins greatly resist decomposition or being broken down in digestion, so they remain in the food chain in meat and dairy products. Even temperature treatments, such as cooking and freezing, do not destroy some mycotoxins [21, 29, 30, 31].

Food-based mycotoxins were studied extensively worldwide throughout the 20th century. In Europe, statutory levels of a range of mycotoxins permitted in food and animal feed are set by a range of European directives and Commission regulations. The U.S. Food and Drug Administration has regulated and enforced limits on concentrations of mycotoxins in foods and feed industries since 1985 [9].

Mycotoxicosis is the term used for poisoning associated with exposures to mycotoxins.

The symptoms of mycotoxicosis depend on the type of mycotoxin; the concentration and length of exposure; as well as age, health, and sex of the exposed individual.[10] The synergistic effects associated with several other factors such as genetics, diet, and interactions with other toxins have been poorly studied. Therefore it is possible that vitamin defi ciency, caloric deprivation, alcohol abuse, and infectious disease status can all have compounded effects with mycotoxins.Chronic diseases are often the result of

Page 133: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

131

adopting minimum quantities mycotoxins for a long period [15, 21]. Mold metabolites may be irritants and may be involved in “ sick building syndrome” [22, 32].

From mycotoxicoses most important are the following: Afl atoxicoses; Fuzariotoxicoses; Mycotoxicoses caused by the Penicillium species of molds; Ergotism.

Afl atoxin mycotoxins are produced by the Aspergillus species of molds. Aspergillus molds grow mostly on crops, such as grains and nuts. Under the right conditions, Aspergillus often grows on grain before it is harvested. But it can also grow on harvested grain if the grain is stored damp.

Afl atoxicosis - afl atoxin poisoning usually occurs from eating food contaminated with afl atoxin mycotoxins. Afl atoxicosis is not contagious and drugs and antibiotics do little to help. Afl atoxicosis damages the liver more than any other organ. Afl atoxin mycotoxins also suppress the immune system [10].Currently 12 are isolated afl atoxins. Afl atoxin Types:There are three main types of afl atoxin mycotoxins: Afl atoxins B, Afl atoxins G, Afl atoxins M [35].

Afl atoxins B: This group includes afl atoxin B1 and B2. Afl atoxin B1 is the most common afl atoxin, as well as the most toxic and carcinogenic. Afl atoxin B1 is announced by the International Agency for Cancer Research (IARC) as a carcinogen in humans № 1 (Class 1 carcinogen) [33].

Afl atoxins G: This group includes afl atoxin G1 and afl atoxin G2.Afl atoxins M: This group includes afl atoxins M1 and M2. Afl atoxin M1 - so-called

“milk toxin” is found in the milk of animals receiving food contaminated with afl atoxin B1, B2, G1, G2.

There are two main ways people are usually exposed to afl atoxins. The fi rst is when someone takes in a high amount of afl atoxins in a very short time. This can cause: Liver damage; Liver cancer; Mental impairment; Abdominal Pain; Vomiting; Convulsions; Edema Pulmonary; Edema Hemorrhaging; Disruption of food digestion, absorption or metabolism; Coma; Death. During acute afl atoxicoses, which occurs rarely, a fat dystrophy and liver necrosis is happening, and in the chronic form - cirrhosis and primary liver carcinoma [20, 23, 46].

Ochratoxin A is produced by the Aspergillus and Penicillium species of molds. Ochratoxin A is probable carcinogen (group 2B) according to the International Agency for Cancer Research (IARC). It has renal toxicity, causes nephropathy and immune suppression, Balkan endemic nephropathy, cancer of the urinary tract [1, 6, 37, 39].In early 2012, the European Food Safety Association (EFSA) published a report on the modeling, predicting and mapping the emergence of afl atoxins in cereals in the European Union (EU) as a result of climate change. It was found that the main risk of afl atoxins is in Spain, Italy and Greece. Portugal, France, Bulgaria and Romania are also some of the risky countries.

The toxic mold Fusarium produces trichothecenes. Trichothecene is one of the most notorious mycotoxins because they are extremely toxic and because they are so diffi cult to destroy [7, 19, 34].

Trichothecenes are produced on many different grains like wheat, oats or maize by various Fusarium species such as F. graminearum, F. sporotrichioides, F. poae and F.

Page 134: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

132

equiseti. Some molds that produce trichothecene mycotoxins, such as Stachybotrys chartarum, can grow in damp indoor environments. It has been found that macrocyclic trichothecenes produced by Stachybotrys chartarum can become airborne and thus contribute to health problems among building occupants [32].

There are approximately 170 trichothecene mycotoxins identifi ed so far. The most common mycotoxins in this group are deoxynivalenol, nivalenol, 3 or 15-acetyl-deoxynivalenol, T-2 and HT-2 toxins. They are powerful inhibitors of protein synthesis and do this by reacting with components of the ribosomes [5, 14, 27].

Zearalenon is produced by the Fusarium species of molds and has strong estrogenic effect [18].

Fumonisinsare produced by the several Fusarium species of molds as Fusarium proliferatum and Fusarium nygamai,as wll as a Alternaria sp. The most common of them is fumonisin B1. Besides the typical hepatotoxicity and nephrotoxicity they affect the immune system [13, 24, 34].

Fuzariotoxicosis. This disease is associated with the use of wintered grain for food. This happens sometimes when cereals and root are stored in winter under the snow. In the spring people thresh grain from such plants and use them for food. Surveys suggest that the disease typically occur within 1-3 weeks after this grain is consumed.There have been registered cases when a single usage of such product has led to disease in severe toxicity form. The toxic property is acquired during winter under the snow by all of the common cereal grains (wheat, rye, barley). People working with such grain were recorded to acquire several pathological phenomena; sharply, irritation of the mucous membrane of the nasal cavity and upper respiratory tract,accompanied by epistaxis, sneezing and coughing. Since the late 19th century, over a long period of time in the Far East, in Russia annually has been registered population disease, associated with the use of grain with toxic properties. Since the climate of the Far East is characterized by higher humidity, the manifestation of the disease there is understandable phenomenon. A similar disease is known in other parts of the globe. Poisoning is characterized byheadaches, dizziness, general weakness, nausea and sometimes vomiting and indigestion [34].

Poisoning “drunken bread” - is caused by eating bread, prepared from grain, damaged by the Fusarium graminearum. The toxin has a neurotropic action. The clinical picture is similar to that of alcohol intoxication with euphoria, dizziness and abnormal and uncoordinated movements. Today the cases of poisoning “drunken bread” are rare.This is due to systematic application of control measures in the general cultivation complex, of which the most important are the following: 1) Phytopathological analysis in the seed- control laboratories, 2) Precise elimination by grain-cleaning machines of immature grain, as it is mostly contaminated; 3) Decontamination of grain, including thermal treatment; 4) Harvesting the grain in short periods [3].

Possibility to use T-2 mycotoxins as a biological agent has been demonstrated in the Russian army shortly after the World War II, when the grain, contaminated with Fusarium species, was used unconsciously for making bread for the civilian population. Some of the infected people developed protracted deadly illness - alimentary toxic aleukia- ATA [43]. It is serious illness caused by toxins produced by the Fusarium sporotrichoides. The disease occurs after consumption of products made from cereals, stored outdoors,

Page 135: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

133

especially bread.Toxins infect bone marrow and lymphoid tissue.ATA is characterized by the initial symptoms, such as abdominal pain, diarrhea, vomiting, prostration. Within a few days the patients developed a fever, myalgia, bone marrow suppression with granulocytopenia and secondary sepsis. In some patients was observed painful pharyngeal / laryngeal ulceration and diffuse bleeding of the skin (petechiae and ecchymoses), melena, hematuria, hematemesis, epistaxis, pancytopenia [3, 5, 34].

T-2 trichothecene mycotoxins are the only mycotoxins that have been used in biological weapons [8, 11, 17, 19, 25, 28, 36, 38, 40, 42, 43, 44]. These trichothecene mycotoxins have the advantages of being highly stable in the air, not degrading under ultraviolet light and being able to withstand heat. They are also used because they can be produced relatively easily and cheaply and they are extremely toxic with no antidote or vaccine available. T-2 mycotoxins are also the only substances used in biological warfare that can be absorbed through a person’s skin.

The United States military is reportedly doing 90% of its current biological weapons research in T-2 mycotoxins. The Yellow Rain biological attacks used in Vietnam and Afghanistan were concentrated T-2 mycotoxins and Gulf War syndrome is believed to be caused by American soldiers’ exposure to T-2 mycotoxins during biological attacks in Desert Storm .

Penicillin is still one of the most widely used antibiotic agents. It is obtained from the Penicillium mould. In 1928 Alexander Fleming noted that the growth of colonies of the bacterium Staphylococcus aureus was inhibited in those areas of a culture that had been contaminated by the mould Penicillium notatum. In 1940, research workers developed an injectable agent for therapeutic use.

Penicillium species are also known to produce mycotoxins. For example, P. verrucosum produces a mycotoxin, ochratoxin A, which is damaging to the kidney (nephrotoxic) and liver and could be cancer causing; there is also evidence that impairs the immune system. Other mycotoxins include patulin, citrinin [21,29, 45].

Patulin is believed to cause hemorrhaging in the brain and lungs and is usually associated with apple and grape spoilage. It can also cause bronchial asthma.

Citrinin can cause renal damage, vasodilatation, and bronchial constriction.P. camemberti has been responsible for inducing occupational allergies among those

who work with soft white cheeses on which the fungus grows.The production of the toxins usually occurs in cereal grains at cold climates but

they have been isolated also in buildings, contaminated with Penicillium. The common occurrence of Penicillium species in food is a particular problem. It is a good practice to discard foods showing the development of any mould.

On the other hand some species of Penicillium are benefi cial to humans. Cheeses such as Roquefort, Brie, Camembert, Stilton, etc. are ripened with species of Penicillium and are quite safe to eat [41, 45].

Ergotismis poisoning due to the ingestion of the alkaloids produced by the Claviceps purpurea fungus which infects rye and other cereals [21, 29, 41, 45]. Two toxic alkaloids involved, ergotamine and ertotaline, cause constriction of the smooth muscles, and ensuing restriction of peripheral blood supplies that can lead to gangrene and death. The symptoms can be roughly divided into convulsive and gangrenous.

Page 136: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

134

Convulsive symptoms include painful seizures, spasms, nausea, paresthesias, itching, nausea,vomiting and diarrhea, headaches, mental effects including mania or central nervous system psychosis. Usually the gastrointestinal effects precede central nervous system effects. People with this form of ergotism perform strange dancing with lots of jumping and screaming, usually ending with exhaustion.

The dry gangrene is a result of vasoconstriction, induced by the ergotamine-ergocristine alkaloids of the fungus. It distal affects the more poorly vascularized distal structures, such as the fi ngers and toes. Symptoms include desquamation or peeling, weak peripheral pulses, loss of peripheral sensation, edema and ultimately the death and loss of affected tissues.

Ergotism has been known for thousands of years, with reports as early as 857 BC. This mysterious disease struck the Spartans in 430 BC during their war with Athens. In the Middle Ages, the gangrenous poisoning was known as ignis sacer (“holy fi re”) or “Saint Anthony’s fi re”, named after monks of the Order of St. Anthony who were particularly successful at treating this ailment. In a 300-year period (1500-1800s), more than 65 epidemics were recorded throughout Europe and satellite countries. There is from 10 to 60% mortality, with many who recover having brain damage.

There is some evidence that ergotism may have been responsible for events surrounding the SalemWitchcrafts in the 17th century [26].

Claviceps gained great notoriety when Hoffman (Sandoz Labs, Switzerland) in 1938 discovered that the sclerotia also contained lysergic acid amide, a precursor of LSD, one of the most potent psychotropic drugs (100 times as potent as psilocybin). Five years later (1943) he discovered its hallucinogenic activity. LSD can result in temporary personality changes and severe “fl ashbacks”, often the latter requiring shock treatment and extensive therapy to eliminate.

Conclusion Mycotoxins are secondary metabolites of molds that have adverse effects on humans,

animals, and crops that result in illnesses and economic losses. Adverse effects of molds and mycotoxins have been recognized for centuries. The diseases – mycotoxicoses, caused by mycotoxins are quite varied. Most of these diseases occur after consumption of mycotoxin contaminated grain or products made from such grains but other routes of exposure exist. Occupational diseases are also recognized in association with inhalation exposure to fungi, usually in industrial or agricultural settings. Molds growing indoors cause building-related symptoms. The diagnosis of mycotoxicoses may prove to be diffi cult because of the similarity of signs of disease to those caused by other agents. The strict control of food quality is necessary to avoid outbreaks of mycotoxicoses.

References

[1]. Bayman, P., J. L. Baker, M. A. Doster, T. J. Michailides and, N. E. Mahoney. 2002. Ochratoxin production by the Aspergillus ochraceus group and Aspergillus alliaceus. Appl. Environ. Microbiol. 68:2326-2329.

[2]. Blout, W. P. 1961. Turkey “X” disease. Turkeys 9:52, 55-58, 61, 77.[3]. Brown, D., S. P. McCormick, N. A. Alexander, R. H. Proctor, and A. E. Desjardins. 2001. A genetic

and biochemical approach to study trichothecene diversity in Fusarium sporotrichioides and Fusarium

Page 137: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

135

graminearum. Fung. Genet. Biol. 32:121-133. [4]. Bunger J, Westphal G, Monnich A, Hinnendahl B, Hallier E, Muller M. Cytotoxicity of occupationally and

environmentally relevant mycotoxins. Toxicology. 2004 Oct 1; 202(3):199-211. (s)[5]. Bunner DL, Upshall DG, Bhatti AR. Toxicology data on T-2 toxin. In: Report of Focus Offi cers Meeting on

Mycotoxin Toxicity, September 23–24, 1985. Suffi eld, Alta, Canada: Defense Research Establishment at Suffi eld; 1985.

[6]. Castegnaro, M., H. Barsch and I. Chernozemsky. 1987. Endemic nephropathy and urinary tract tumors in the Balkans. Cancer Res. 47:3606-3609.

[7]. Chu, F. S. 1997. Trichothecene mycotoxicoses. Encyclopedia Hum. Biol. 8:511-522.[8]. Ciegler, A. 1986. Mycotoxins: a new class of chemical weapons. NBC Defense & Technol. Int., April 1986,

p. 52-57.[9]. Creppy EE. Update of survey, regulation and toxic effects of mycotoxins in Europe. Toxicol Lett. 2002 Feb

28;127(1-3):19-28 (s)[10]. Cullen, J. M., and P. N. Newberne. 1994. Acute hepatotoxicity of afl atoxins, p. 3-26. In D. L. Eaton and

J. J. Groopman (ed.). The toxicity of afl atoxins. Human health, veterinary, and agricultural signifi cance. Academic Press, San Diego.

[11]. Dashek WV, Mayfi eld JE, Llewellyn GC, O’Rear CE, Bata A. Trichothecenes and yellow rain: Possible biological warfare agents. Bioessays. 1986;4(1):27–30.

[12]. De Vries, J. W., M. W. Trucksess, and L. S. Jackson (ed.). 2002. Mycotoxins and food safety. Kluwer Academic/Plenum Publications, New York, N.Y.

[13]. Desjardins, A. E., and R. D. Plattner. 2000. Fumonisin B (1)-nonproducing strains of Fusarium verticillioides cause maize (Zea mays) ear infection and ear rot. J. Agric. Food Chem. 48:5773-5780.

[14]. Dohnal V., Jezkova A., Jun D., Kuca K. (2008). “Metabolic pathways of T-2 toxin”. Current Drug Metabolism9 (1): 77–82.

[15]. [15] Edmondson DA, Nordness ME, Zacharisen MC, Kurup VP, Fink JN. Allergy and “toxic mold syndrome”. Ann Allergy Asthma Immunol. 2005 Feb; 94(2):234-9. (s)

[16]. Etzel, R. A. 2002. Mycotoxins. J. Am. Med. Assoc. 287:425-427. [17]. Evans, G. 1983. The yellow rainmakers. Are chemical weapons being used in Southeast Asia? Verso/

Schocken, London, United Kingdom.[18]. Hagler, W. M., Jr., N. R. Towers, C. J. Mirocha, R. M. Eppley, and W. L. Bryden. 2001. Zearalenone:

mycotoxin or mycoestrogen?, p. 321-331. In B. A. Summerell, J. F. Leslie, D. Backhouse, W. L. Bryden, and L. W. Burgess (ed.), Fusarium. Paul E. Nelson Memorial Symposium. APS Press, St. Paul, Minn.

[19]. Henghold II, W. B. 2004. Other biologic toxin bioweapons: Ricin, staphylococcal enterotoxin B, and trichothecene mycotoxins. Dermatologic Clinics. 22: 257-262.

[20]. Henry, S. H., F. X. Bosch, and J. C. Bowers. 2002. Afl atoxin, hepatitis and worldwide liver cancer risks, p. 229-320. In J. W. DeVries, M. W. Trucksess, and L. S. Jackson (ed.), Mycotoxins and food safety. Kluwer Academic/Plenum Publications., New York, N.Y.

[21]. Hussein HS, Brasel JM. Toxicity, metabolism, and impact of mycotoxins on humans and animals. Toxicology. 2001 Oct 15; 167(2):101-34. (s)

[22]. Jarvis BB, Miller JD. Mycotoxins as harmful indoor air contaminants. Appl Microbiol Biotechnol. 2005 Jan; 66(4):367-72. E pub 2004 Nov 23. (s)

[23]. Li, F.-Q., T. Yoshizawa, S. Kawamura, S.-Y. Luo, and Y.-W. Li. 2001. Afl atoxins and fumonisins in corn from the high-incidence area for human hepatocellular carcinoma in Guangxi, China. J. Agric. Food Chem. 49:4122-4126.

[24]. Marasas, W. F. O., J. D. Miller, R. T. Riley, and A. Visconti. 2001. Fumonisins—occurrence, toxicology, metabolism and risk assessment, p, 332-359. In B. A. Summerell, J. F. Leslie, D. Backhouse, W. L. Bryden, and L. W. Burgess (ed.), Fusarium. Paul E. Nelson Memorial Symposium. APS Press, St. Paul, Minn.

[25]. Marshall, E. 1983. Yellow rain experts battle over corn mold. Science 221:526-529. [26]. Matossian, M. K. 1982. Ergot and the Salem witchcraft affair. Am. Sci. 70:355-357.

Page 138: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

136

[27]. Miller, J. D., J. W. ApSimon, B. A. Blackwell, R. Greenhalgh, and A. Taylor. 2001. Deoxynivalenol: a 25 year perspective on a trichothecene of agricultural importance, p. 310-319. In B. A. Summerell, J. F. Leslie, D. Backhouse, W. L. Bryden, and L. W. Burgess (ed.), Fusarium, Paul E. Nelson Memorial Symposium. APS Press, St. Paul, Minn.

[28]. Nowicke, J. W., and M. Meselson. 2000. Yellow rain—a palynological analysis. Nature 309:205-206. [29]. Peraica M, Domijan AM. Contamination of food with mycotoxins and human health. Arh Hig Rada

Toksikol. 2001 Mar;52(1):23-35 (s)[30]. Pitt JI. Toxigenic fungi and mycotoxins. Br Med Bull. 2000; 56 (1):184-92. (s)[31]. Pitt, J. I. 2000. Toxigenic fungi: which are important? Med. Mycol. 38(Suppl. 1):17-22. [32]. Portnoy JM, Kwak K, Dowling P, VanOsdol T, Barnes C. Health effects of indoor fungi. Ann Allergy

Asthma Immunol. 2005 Mar; 94 (3):313-9; quiz 319-22, 390. (s)[33]. Pozzi, C. R., B. Correa, J. G. Xavier, G. M. Direito, R. B. Orsi, and S. V. Matarazzo.2000. Effects of

prolonged oral administration of fumonisin B1 and afl atoxin B1 in rats. Mycopathologia 151:21-27. [34]. Proctor, R. H. 2000. Fusarium toxins: trichothecenes and fumonisins, p. 363-381. In J. W. Cary, J. E. Linz,

and D. Bhatnagar (ed.), Microbial food-borne diseases: mechanisms of pathogenesis and toxin synthesis. Technomic Publications, Lancaster, Pa.

[35]. Reijula K, Tuomi T. Mycotoxins of aspergilli: exposure and health effects. Front Biosci. 2003 May 1; 8: s232-5. (s)

[36]. Rothschild J.H., Tomorrow’s weapons[microform]: chemical and biological,New York : McGraw-Hill, 1964.

[37]. Schwartz, G. G. 2002. Hypothesis: does ochratoxin A cause testicular cancer? Cancer Causes Control 13:91-100.

[38]. Seagrave S. Yellow Rain: A Journey Through the Terror of Chemical Warfare. New York, NY: M Evans; 1981.

[39]. [Skaug, M. A., E. Eduard, and F. C. Stormer. 2000. Ochratoxin A in airborne dust and fungal conidia. Mycopathologia 151:93-98.

[40]. Stone, R. 2002. Peering into the shadows: Iraq’s bioweapons program. Science 297:1110-1112. [41]. Sweeney, M. J., S. White, and A. D. W. Dobson. 2000. Mycotoxins in agriculture and food safety. Irish J.

Agric. Food. Res. 39:235-244.[42]. Wannemacher RW, Bunner DL, Pace JG, Neufeld HA, Brennecke LH, Dinterman RE. Dermal toxicity

of T-2 toxin in guinea pigs, rats, and cynomolgus monkeys. In: Lacey J, ed. Trichothecenes and Other Mycotoxins. Chichester, England: John Wiley & Sons Ltd; 1985: 423–432.

[43]. Wannemacher, R. W., Acute Inhalation Toxicity of T-2 Mycotoxin in Mice and Guinea Pigs, 1988[44]. Watson, S. A., C. J. Mirocha and A. W. Hayes. 1984. Analysis for trichothecenes in samples from southeast

Asia associated with ‘yellow rain’. Fundamental Appl. Toxicol. 4: 700-717.[45]. Weidenborner, W. 2001. Encyclopedia of food mycotoxins. Springer, Berlin, Germany.[46]. Wilson, D. M., W. Mubatanhema, and Z. Jurjevic. 2002. Biology and ecology of mycotoxigenic Aspergillus

species as related to economic and health concerns, p. 3-17. In J. W. deVries, M. W. Trucksess, and L. S. Jackson (ed.), Mycotoxins and food safety. Kluwer Academic Plenum Publications, Dordrecht, The Netherlands.

[47]. Zilinskas, R. A. 1997. Iraq’s biological weapons. The past as future? J. Am. Med. Assoc. 276:418-424.

Page 139: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

137

Chapter 16

Blood Level of Histamine- Indicator for the Expression of Abstinence Syndrome

in the Phase of Alcohol Withdrawal

NEYKOVA L.1, KONOV V.1, KANEV K.,1 GALABOVA G.2

1Clinic “Emergency Toxicology” - MMA, Sofi a 2Radioisotope Laboratory, MMA, Sofi a

IntroductionThe endocrine and nervous systems are the two major systems involved in the control

and regulation of body functions. The level of hormonal secretion is often regulated on the principle of negative feedback. Histamine (2 - [4-imidazolyl] ethylamine) was discovered in 1910.

The properties of the so-called then β-iminazolylethylamine, were fi rst found by the British scientists Henry H. Dale and PP Laidlaw.[17] In 1932, it was described as a mediator of the anaphylactic reactions. It is a biogenic amine, and despite of its small molecule, as compared with other organic molecules (containing 17 atoms), it plays an important role in the organism. It is known to be involved in 23 different physiological functions.

The alcohol compromises the histamine action due to the common metabolizing enzymes - aldehyde dehydrogenase and aldehyde. The metabolic product of the ethanol oxidation - acetaldehyde, competes with the aldehydes obtained by histamine metabolism.

AimThe aim of this study was to investigate the histamine level in patients with alcohol

dependence and discuss its role as an indicator of the abstention syndrome in the phase of withdrawal of alcohol.

Material and methodsThe material includes 52 patients with alcohol dependence treated in Emergency

Toxicology Clinic (Military Medical Academy) in 2012. Of these 15 were women and 37 men. Their standard blood bio-chemical indicators were tested. The level of histamine was defi ned by radioimmunoassay quantifi cation of histamine in plasma and by in vitro diagnosis. The methods of clinical observation and introspection were also used.

ResultsThe normal levels of histamine in blood plasma are 0.3 to 1.0 ng / ml. The concentrations

of ethanol in the examined group were between 0.9 and 5.1 g / l; and in urine - between

Page 140: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

138

0.3-5.7 g / l (this data could be indicative of the phase: absorption or elimination). 10 patients (6 women: 4 men) were with no liver enzymes changes and steatosis (proven by ultrasound). The duration of the alcohol abuse was different as shown in Table 1.

Table 1 Duration of Abuse

Duration of Alcohol Abuse Number of Patients

Up to 5 years 21Up to 10 years 13Up to 20 years 11Over 21 years 7

In 18 of the examined 52 patients (34.61%) no deviations from the referent norms were found. The sex ratio was 17 men and 1 woman. The patients were within the age range of 34 and 63 years - the estimated mean age was 46.17. Eleven (61.11%) of these patients had no withdrawal symptoms, and the remaining 7 (38.89%) had the characteristic withdrawal symptoms. Four or 22.22% are without steatosis or abnormalities in laboratory liver function tests. In the remaining 14 patients (77.78%) by ultrasound were proven changes or deviations from the reference levels for ASAT, ALAT, GGT.

Of the 52 patients tested 34 (65.38%) had high level of histamine. In 13 of 34 patients (38.24%) or 25% of the total sample, the levels were 3-4 times higher than the normal range (average of 3.615 ng/ml). The remaining 21 patients were with deviations from the normal level of histamine, i.e. 61.76% were towards a higher level (between 1.1 - 2.4 ng/ml, an average of 1.88 ng/ml) from the reference range. The age rangewas 25-65 years, with mean age 41.45. The sex ratio was 20 men and 14 women. The concentration of ethanol in blood was between 0.9 and 5.1 g / l; in urine - between 0.3 - 5.7 g / l (this data could be considered as indicators of the phase: absorption or elimination). Six of the patients had no liver changes. Eighteen patients (52.94%) had no withdrawal symptoms, the remaining 16 (47.06%) were abstinently, two of them were with alcoholic delirium (5.88%). Ten patients (35.71%) had different forms of depressive disorders- 6 men: 4 women. Of the total tested group, 38 patients (73.07%) were admitted after heavy episodic drinking. The other 14 (26.93%) were consuming alcohol on a daily basis.

In individuals from the latter group 6 patients (11.54% of the total group, or 42.86% of the sub-group) had normal levels of histamine (N-Histamine). The remaining 8 patients (15.38% of the total number or 57.14% in the subset) had levels of histamine between 1.3-3.4 ng / ml, average 2.02 ng / ml. With the withdrawal symptoms were 3 patients with normal histamine and 4 of them were with higher levels (between 2.1 and 3.9 ng / ml). In the same group 4 were with depressive symptoms (at 2 in the sub-groups), one with Panic disorder and normal histamine.

In the subgroup “heavy episodic drinking” 13 individuals (25 % of the total number or 34.21% of the subgroup ) were with normal levels of histamine. Five of them ( 38.46%) were in a withdrawal state ; 2 patients had depression ( 15.38% ) . The remaining 25 patients were with a deviation of the reference ranges between 1.3 and 4.9 ng/ml. Of these, 11 patients (73.33%) expressed the withdrawal phenomena. Four patients were with depressive disorders, 3 had epilepsy, 2 were with bipolar affective disorder (BAD)

Page 141: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

139

(one woman suffered from bulimia), and one female patient was with anorexia. They all had high histamine levels: between 1.9-4.9 ng/ml. Three patients - 2 men and 1 woman had combined (benzodiazepine) dependency. In 13 of the patients with elevated levels of histamine (37.14%) high plasma concentrations of prolactin was found. Of the 23 patients with withdrawal symptoms (7 subgroup of “daily abuse” of alcohol and 14 - “ heavy episodic drinking “), 65.22% or 28.85% of the whole group were patients with pre-delirious predisposition of them 11 men and 4women.

DiscussionIn the organism, histamine is synthesized by decarboxylation of the amino acid

histidine, a reaction catalyzed by the enzyme L-histidine decarboxylase. Once synthesized, it is stored in the form of granules in mast cells and basophiles, and is rapidly deactivated by the main enzymes that degrade it – the histamine-N-methyltransferase transferase (HNMT) or the diamine oxidase (DAO).

The mastocytes, whicharethe main reservoirs of histamine in the body are mainly clustered in the areas exposed to the greatest risk of injuries and traumas. The non-mastocyte histamine is found mainly in the brain, where it functions as an important neurotransmitter, and in enterochromaffi n like (ECL) cells in the stomach. The main mechanism for release of histamine from mastocytes and basophils is immunological . The IgE- antibodies are located on the cell membranes which are triggered by specifi c antigens. As a result, begins destruction of the histamine granules, and histamine secretes. [1,2,3,4] .Certain amines and alkaloids, including medicaments, surfactants and alchohol, curare alkaloids substitute the histamine in the granules causing it to discharge or block the DAO and provoke various symptoms: diarrhea, headache, [14], nasal congestion, lung wheezing, asthma attacks [6,8,15], hypotension, arrhythmia,urticaria [16,17], pruritus, fl ushing, and other complications. Serum DAO concentrations showed no signifi cant changes and no signifi cant gender differences. [27]

Histamine is metabolized in two main ways: by oxidative deamination by DAO or by cyclic methylation of HNMT [28]. Whether histamine is catabolised from DAO or HNMT is determined by the local location of the histamine. The DAO protein is stored in the plasma membrane associated vesicular structures in epithelial cells and is secreted in stimulation [29,30] . It is therefore presumed that the DAO is responsible for the extracellular histamine (e.g., following consumption of rich histamine food). Conversely, HNMT is a cytosolic protein [31], that can be converted to histamine in the cells only, i.e. intracellulary. [32, 33] Thus, the enzymes seemingly, do not compete for the substrate, although they have a similar affi nity for the histamine and are expressed in some tissues. HNMT has a slightly higher affi nity than does DAO. In mammals, DAO- release is restricted to certain tissues: highest in the small intestine and ascending colon [18,19,34], in the placenta and in the kidneys . [29,32] HNMT is widely expressed in the kidneys and the liver, then in the spleen, the colon, the prostate, the ovary , the cells of the spinal cord, trachea and bronchi . [35] In the Central nervous system (CNS), the released in the synapses histamine gets metabolized by HNMT. Several other enzymes, including MAO-B and ALDH2, may also participate in the “ processing “ of the immediate metabolites of histamine .

The histamine exerts its effect by binding to specifi c histamine receptors on cells. There are 4 types of such receptors - H1, H2, H3 and H4, which belong to the group of

Page 142: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

140

G protein-coupled receptor (GPCR). Histamine H1 receptor is localized predominantly in the smooth muscle, and the endothelium of the CNS. It plays a role in the regulation of the sleep and the appetite suppression. The H2 receptor is located in the parietal cells, and vascular smooth muscle cells. The histamines main role is the vasodilation. It also stimulates gastric acid secretion.

The H3 receptor is localized in the CNS, and to a lesser extent in peripheral NS. The correlation between histamine neurotransmission and that of acetylcholine, noradrenaline, serotonin is proven. H4 receptor is located predominantly in basophils in the bone marrow. Also, there are receptors in the thymus, small intestine, the spleen and the colon. It plays a role in chemotaxis.

The release of histamine is a sensitive indicator of stress. It is a major neurotransmitter in the CNS and a key regulator of behaviour, and thus plays a role in the pathogenesis of the sleep disorders, in the pathophysiology of many encephalopathies and possibly such of toxic (alcoholic) genesis due to metabolic failure. The histaminergic neurons that release histamine, are located in the posterior hypothalamus, the tuberomamilaric nuclei and are involved in the modulation of sleep.

The histaminergic neurons in the hypothalamus are connected with some major brain functions and the vegetative and endocrine control. Histamine levels in the brain are determined by the presence of histidine, which is increased manyfold in patients with liver changes, and liver cirrhosis. This leads to about 13 times higher brain histamine levels, particularly in the hypothalamus, as well as to changes in telemethyl- histamine and histamine-N-methyl transferase activity.

Gastrointestinal diseases with altered enterocytes may result at low production of DAO [20, 34 ,36]. Another reason may be the competitive inhibition of histamine degradation of DAO from other biogenic amines, alcohol [24,25] , or medicines. [22,23,37] DAO -inhibits the penetration of exogenous histamine [38,39] and impaired DAO-activity results in increased histamine enteral absorption with consequent increase in the plasma concentrations of the histamine - [22 , 39] and the corresponding symptoms.

Elevated levels of histamine metabolites may inhibit HNMT, metabolizing enzymes responsible for the change volume of histamine in the CNS. [21, 40] In patients with alcohol dependence, the main metabolite of the ethanol – the acetaldehyde, is presumed to be a histamine-releasing substance. [26]

Conclusions: From the study group of 52 patients with chronic alcohol abuse we can make the

following conclusions:1. The change of histamine levels is not connected with the type of alcohol abuse. 2. 16 patients (30.77% of the entire group, or 47.06% of the subgroup with elevated histamine

thymine) with withdrawal syndrom showed high levels of histamine, which supports its role as an indicator of stress; two of them were with alcoholic delirium (5.88% or 3.85% of the total).

3. 13 patients or 25% of the group, were with levels 3-4 times higher than the reference norm (an average of 3.615 ng / ml). 10 of them had steatotic changes.

4. To the remaining 13.46% (or 7 patients) with clear withdrawal, but no changes in histamine levels, other neurotransmitter mechanisms and factors were included;

Page 143: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

141

5. The relationship between the degree of increase in histamine level and the severity of withdrawal expression was confi rmed. Six of the patients had pre-delirium predisposition , which is 17.65% of the whole group (34 patients) with elevated histamine levels or 46.15% of the group with 3-4 times higher values .

6. The correlation between liver function and levels of histamine thymine was confi rmed. Ten patients (6 women: 4 men), or 19.23% were with no changes in the liver enzymes or steatotic changes.

7. The obtained by us results support of the thesis of the competitive inhibition of histamine DAO-degradation of alcohol. 34 patients (65.38%) had abnormal levels of histamine, which might be explained by the elevated amounts of histamine metabolites, that can inhibit HNMT, metabolizing enzyme responsible for the metabolism of histamine in the CNS.

8. The above fi ndings show the need for further surveys of histamine levels.

References: [1] http://www.sciencelab.com/msds.php?msdsId=9924264[2] Marieb, Е. (2001). Human anatomy and physiology. San Francisco: Benjamin Cum-mings. p. 414. ISBN

0-8053-4989-8.[3] Di Giuseppe, M.,et al. (2003). Nelson Biology 12. Toronto: Thomson Canada. p. 473. ISBN 0-17-625987-2. [4] Paiva, TB; Tominaga, M.; Paiva, ACM (1970). “Ionisation histamine, N-acetylhistamine, and iodinated

derivatives” Journal of Medicinal Chemistry 13 (4):. 689-692. DOI: 10.1021/ jm00298a025. PMID 5452432. [5] http://astrobiology.berkeley.edu/PDFs_articles/WineAnalysisAnalChem.pdf[6] Jump up to: a b c Noszal, B.; Kraszni, M.; Racz, A. (2004). “Histamine: Fun-damentals of Biochemistry.”

In Falus, A.; Grosman, N.; Darvas, Z. Histamine: Biology and Medical Aspects. Budapest: SpringMed. p 15-28. ISBN 380557715X.

[7] Hardie RC (June 1989). “A histamine-activated chloride channel involved in neurotrans-mission photoreceptor synapse” Nature 339 (6227): 704-6 DOI: 10.1038/339704a0. PMID 2472552.

[8] Pantazis A, Segaran A, Liu CH, and others. (July 2008). “Different roles for two histamine receptors (hclA and hclB) in Drosophila photoreceptor synapse.” J. Neurosci 28 (29):. 7250-9. DOI: 10.1523/JNEUROSCI.1654-08.2008. PMID 18632929.

[9] Monroe EW, Daly AF,Shalhoub RF (February 1997). “Evaluation of the validity of histamine-induced wheal to predict the clinical effi cacy of antihistamines.” J. Allergy Clin. Immunol 99 (2): .. S798-806 PMID 9042073.

[10] Yanai, K; Tashiro, M (2007). “The physiological and pathological roles of neuro-nal histamine.: To inform studies of human positron emission tomography,” Pharmacology and Therapeutics 113 (1):1-15 DOI: 10.1016/j.pharmthera.2006.06.008. PMID 16890992.

[11] Alvarez, EO (2009). “The role of histamine on cognition.” Behavioural Brain Research 199 (2):. 183-9. DOI: 10.1016/j.bbr.2008.12.010. PMID 19126417.

[12] White JM; Rumbold, GR (1988). “Behavioral effects of histamine and its antago-nists: a review.” Psychopharmacology 95 (1):1-14. PMID 3133686.

[13] Cara, AM; Lopes-Martins, RA; Antunes, E; Nahoum, CR; De Nucci, G (1995). “Role of histamine in the human penise erection.” British Journal of Urology 75 (2): 220-4. DOI: 10.1111/j.1464-410X.1995.tb07315.x. PMID 7850330.

[14] Ito, C (2004). “The role of the central histaminergic system on schizophrenia” Drug News & Perspectives 17(6):383-7DOI:10.1358/dnp.2004.17.6.829029.PMID 15334189.

[15] Jadidi-Niaragh F, Mirshafi ey A (September 2010), “The histamine and histamine recep-tors in the pathogenesis and treatment of multiple sclerosis” Neuropharmacology 59 (3):180-9 DOI: 10.1016/j.neuropharm.2010.05.005. PMID 20493888.

[16] Valent P, Horny HP, Escribano L, etc. (July 2001) “Diagnostic criteria and classifi -cation of mastocytosis: a consensus proposal” Leuk .. Res 25 (7): 603-25. PMID 11377686.

Page 144: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

142

[17] Dale HH, Laidlaw PP (December 1910). “The physiological action of β-imina-zolylethylamine” (PDF). J. Physiol. (Lond.), 41 (5): 318-44. PMC 1512903. PMID 16993030.

[18] Bieganski T, Kusche J, Lorenz W, Hesterberg R. Stahlknecht CD, Feussner KD. Dist-ribution and properties of human intestinal diamine oxidase and its relevance for the hista-mine catabolism. Biochim Biophys Acta 1983;756:196-203.

[19] Bieganski T. Biochemical, physiological and pathophysiological aspects of intes-tinal diamine oxidase. Acta Physiol Pol 1983;34:139-54.

[20] Schmidt WU, Sattler J, Hesterberg R, et al. Human intestinal diamine oxidase (DAO) activity in Crohn’s disease: a new marker for disease assessment? Agents Actions 1990;30:267-70.

[21] Sattler J, Hafner D, Klotter HJ, Lorenz W, Wagner PK. Food-induced histaminosis as an epidemiological problem: plasma histamine elevation and haemodynamic alterations after oral histamine administration and blockade of diamine oxidase (DAO). Agents Actions I988;23: 361-5.

[22] Sattler J, Lorenz W. Intestinal diamine oxidases and enteral-induced histaminosis: studies on three prognostic variables in an epidemiological model. J Neural Transm Suppl 1990; 32:291-314.

[23] Sattler J, Hesterberg R, Schmidt U,CrombachM, Lorenz W. Inhibition of intestinal diami-ne oxidase by detergents: a problem for drug formulations with water insoluble agents applied by the intravenous route? Agents Actions 1987;20:270-3.

[24] Wantke F, Gotz M, Jarisch R. The red wine provocation test: intolerance to his-tamine as a model for food intolerance. Allergy Proc 1994; 15:27-32.

[25] Zimatkin SM, Anichtchik OV. Alcohol- histamine interactions. Alcohol Alcohol 1999; 34:141-7.[26] Kanny G, Gerbaux V, Olszewski A, et al. Nocorelation between wine intolerance and histamine content of

wine. J. Allergy Clin. Immunol.2001; 107:375-8.[27] Wantke F, Proud D, Siekierski E, Sobotka A. Daily variations of serum diamine oxidase and the infl uence

of H1 and H2 blockers: a critical approach to routine diamine oxi-dase assessment.Infl amm Res 1998; 47:396-400.

[28] Schwelberger HG. Diamine oxidase (DAO) enzyme and gene.In: Falus A, ed. Histamine: biology and medical aspects. Budapest, Hungary: SpringMed Publishing, 2004:43-52.

[29] Schwelberger HG, Hittmair A, Kohlwein SD. Analysis of tissue and subcellular localization of mammalian diamine oxidase by confocal laser scanning fl uorescence mic-roscopy. Infl amm Res 1998;47(suppl): S60-1.

[30] Schwelberger HG, Bodner E. Purifi cation and characterization of diamine oxidase from porcine kidney and intestine. Biochim Biophys Acta 1997; 1340:152-64.

[31] Brown DD, Tomchick R, Axelrod J.The distribution and properties of a histami-ne-methylating enzyme. J Biol Chem1959; 234:2948-50.

[32] Klocker J, Matzler SA, Huetz GN, Drasche A, Kolbitsch C, Schwelberger HG. Expression of histamine degrading enzymes in porcine tissues.Infl amm Res 2005; 54 (suppl): S54-7.

[33] Kufner MA, Ulrich P, Raithel M, Schwelberger HG. Determination of histamine degadation capacity in extremely small human colon samples.Infl amm Res 2001; 50(suppl): S96-7.

[34] Raithel M, Kufner M. Ulrich P, Hahn EG. The involvement of the histamine degradation pathway by diamine oxidase in manifest gastrointestinal allergies. Infl amm Res 1999;48(suppl):S75-6.

[35] Schwelberger HG. Histamine W-methyltransferase (HNMT) enzyme and gene. In: Falus A, ed. Histamine: biology and medical aspects. Budapest, Hungary: SpringMed Publishing, 2004:53-9.

[36] Yarnauchi K, Sekizawa K, Suzuki H, et al. Structure and function of human histamine /V-methyltransferase: critical enzyme in histamine metabolism in airway. Am J Physiol 1994;267:L342-9.

[37] Sattler J. Hesterberg R, Lorenz W, Schmidt U, Crombach M, Stahlknecht CD. Inhibition of human and canine diamine oxidase by drugs used in an intensive care unit: relevance for clinical side effects? Agents Actions 1985;16:91-4.

[38] Ahrens F. Gabel G, Garz B, Aschenbach JR. Release and permeation of histamine are affected by diamine oxidase in the pig large intestine. Infl amm Res 2002;51(suppl):S83-4.

[39] Aschenbach JR, Oswald R, Gabel G. Gastrointestinal epithelia as bar¬riers to luminal histamine of microbial origin. Z Gastroenterol 1998; 36:12-7.

[40] Sattler J. Lorenz W, Kubo K, Schmal A, Sauer S, Luben L. Food- induced histaminosis under diamine oxidase (DAO) blockade in pigs: further evidence of the key role of elevated plasma histamine levels as demonstrated by successful prophylaxis with antihistamines. Agents Actions 1989;27:212-4.

Page 145: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

143

Chapter 17

Traditional and New Treatments of Addiction – Ethical and Legal Aspects

Julia RADENKOVA-SAEVAa1, Eva SAEVAb,aToxicology Clinic, “Pirogov” University Hospital, Sofi a, Bulgaria

bUniversity “La Sapienza”, Rome, Italy

Abstract: Drug addiction is a psychiatric disorder, usually classifi ed as a “chronic andrelapsing brain disease” and is characterized by a loss of control over drug consumption. Animal studies and other evidence suggest great variability in vulnerability to addiction. Recent advances in neurobiological studieshave enabledscientists to identify neuropsychological and genetic differences in individuals that may infl uence their chances of developing addiction if they use drugs.By providing a better understanding of how addiction develops, these investigations may bring the basis for new psychological and pharmacological treatments and prevention strategies.Unfortunately the data can be inappropriately used to justify involuntary, high invasive or even harmful treatmentof persons, addicted to psychoactive substances, without a suffi cient concern for human rights and ethical implications.The risks of over-simplistic interpretation of the new neurobiological data have been discussed in this review.In our opinion a balanced approach is required in addiction policies with a concern for human rights and ethical principles.Key words: drug addiction, neurobiological studies, human rights

Introduction

Drug addiction is a behavior characterized by the individual exhibiting a loss of control over their consumption. Modern developments in neurobiology now provide strong scientifi c grounds for viewing drug addiction as a psychiatric disorder, usually classifi ed as a ‘chronic and relapsing brain disease’.

The effective treatment of drug dependence aims to achieve the following:reduction or elimination of illicit drug use; reduction of the health damages associated with drug use (e.g. transmission of infectious diseases such as HIV and viral hepatitis, drug-related deaths by overdose and other causes); reduction of the harms to society, principally in the form of crimes that drug users may commit while under the infl uence of drugs, in order to buy drugs, or in resolving confl icts in illicit drug markets[10, 22, 23].

1 Corresponding author: Assoc. Prof. Julia Radenkova - Saeva, MD, PhD, Clinic of Toxicology, UMHATEM „ N. I. Pirogov”,Totleben 21, 1606 Sofi a, Bulgaria, e-mail: [email protected]

Page 146: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

144

The promise of neuroscience and genetic research on drug addiction raises major ethical and social issues [13, 14]. These can be considered under two broad headings: ethical issues that arise from neuroscience and genetic research on addiction; and the broader social and ethical implications of the potential technological applications of neuroscience (e.g. for therapeutic and preventive purposes) [12, 9].Ethical treatment will be considered to be treatment which complies with both international human rights law and leading codes of medical ethics [3, 4, 5, 11].

Neurobiological research in addictionAlmost all drugs known to induce abuse or addiction in humans increase the release

of a neurotransmitter called dopamine in a sub-cortical structure, the nucleus accumbens. The cell bodies of neurons which release dopamine are located in the ventral tegmental area and the substantia nigra. These dopaminergic neurons form the meso-corticolimbic pathway. They stimulate different cerebral structures, such as the prefrontal cortex, the amygdala and the hippocampus, which are part of a circuitry called the “reward system”. Most neurobiological models of addiction argue that, because drugs of abuse release dopamine and activate the reward system, addiction is due to a modifi cation of kinetic reactions and increased dopamine release. This dysregulation would correspond either to an increased reactivity of dopaminergic neurons to specifi c stimuli linked to the pleasurable and addictive product or to a down-regulation of dopamine signaling and a dampening of activity in the reward pathway [20].

Some recent studies suggest that, despite the critical and unquestionable role dopamine plays in reward, drugs of abuse may not necessarily induce addiction via a direct effect on dopaminergic neurons. There is some evidence that dopamine acts downstream to two other neuromodulators, noradrenaline and serotonin, responsible for vigilance and for the control of impulsivity, respectively.

New technologies in addiction researchAnimal studies and other evidence suggest great variability in vulnerability to

addiction. New technologies mean that neurobiological research can start to identify

neuropsychological and genetic differences in individuals that may infl uence their chances of developing addiction if they use drugs.

Advances in genomic and molecular biology, such as the ability to clone and sequence receptor subtypes, transporters and endogenous agonists, have enabled scientists to identify and specifi cally target relevant receptor or transporter sites with drugs that either block (antagonists) or facilitate (agonists or partial agonists) activity. In humans, genetic studies have tried to identify specifi c addiction susceptibility genes [7, 12, 16]. Neuroimaging, using technologies such asfunctional magnetic resonance imaging (fMRI), positron tomography (PET), singlephoton emission computed tomography(SPECT), magnetoencephalograph (MEG),and electroencephalograph (EEG), hasprovided insight into the way in whichdrug-induced changes in the brain canproduce the type of cognitive defi cits seen inpeople addicted to drugs. These are non-invasive techniques which may help to identify neuropsychological defi cits that maybe the primary source of an individual’sinability to stop using drugs [9].

Page 147: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

145

Traditional and new treatments of addictionAddiction has traditionally been treated bya combination of pharmacological and

psychosocial treatments. Usualpharmacological treatments include:(a) drugs that either block the addictive

drugfrom working (e.g. naltrexone as a relapseprevention of heroin dependence) or make it unpleasant (e.g. disulfi ram for alcoholdependence) or (b) drugs that replace the addictive drug with a less harmful version ofthe drug (e.g. opioid substitution treatment using methadone). Nicotine replacement therapy is a common form of substitution treatment for smoked tobacco but is not particularly effective. Some treatments mayalso be used for a short period to help wean individuals off all drugs [6, 8].

Psychosocial interventions include cognitive behavioural therapy; motivational interviewing; drug counseling; 12-step support groups.

These therapies provide an important adjunct to pharmacological and medical treatments in achieving a long-term successful outcome.

Progress in neurobiological research on addiction has led to the use of drugs which target is the dopaminergic system. However, this strategy has not yet proven effective in treating addiction, possibly because the wrong dopamine receptor has been targeted (i.e. D2) or because other modulatory neurotransmitter systems also require consideration.

A number of other novel treatment approaches are in development or are being researched which may provide new approaches to treating some forms of drug dependence as immunotherapies and approaches of neurosurgery.

Immunotherapies in the form of ‘vaccines’against the effects of nicotine, cocaine andheroin that act by binding to the target drugin the bloodstream, thus preventing it fromreaching the brain [1].

Neurosurgery is the mostinvasive and permanent form ofexperimental treatment, but there exist strongethical objections to this approach.

Less extreme but still raising ethical concerns is deep brain stimulation which involves the insertion of electrical stimulating electrodes into the regions of the brain involved in addiction, such as the insula.

A less invasive approach is transcranial magnetic stimulation that involves placing a small magnetic coil against an individual’s skull in order to block or enhance neural activity. None of these approaches are currently proven and all bring with them potential costs as well as possible benefi ts.

Balanced approach in addiction policies. Ethical principles and human rights.Drug use encompasses a complex set of behaviors and even the autonomy of dependent

individuals is variable. One risk of an overly simplifi ed interpretation of the emerging neurobiological evidence is that it could be inappropriately used to justify coerced, highly invasive or even damaging treatments, by proponents overly optimistic about their ability to cure addiction and without showing suffi cient concern for broader human rights and ethical implications [10, 11, 17].

Developments in neuroscience suggest that a balanced approach is required in addiction policies. The justifi cation for such an approach is grounded in the medical

Page 148: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

146

model of addiction and the requirement for punitive measures. Currently, policies tend to be often weighted towards the deployment of criminal responses to addiction. Advances in addiction neuroscience could be selectively used to justify more coercive policies towards illicit drug use in the name of preventing adolescents from acquiring a ‘chronic brain disease’ [14, 15]. Advances in treatment for addiction or mental health disorders are to be welcomed, whether through vaccinations or a greater understanding of genetics. However, the use of such advances must be balanced against certain key ethical questions [13]. If neurobiological research leads to the development of new treatment approaches, they will join and hopefully complement existing treatments. Patients will need to be given information on different treatment options, and the costs and benefi ts of any new therapy should be carefully considered along with its potential effi cacy. Treatments that are invasive or dangerous are diffi cult to justify if safer options already exist. Important ethical considerations will surely arise if patients are denied free choices over which treatment they can pursue: these issues are likely to be particularly important for treatments offered within the criminal justice system where some degree of coercion may be present. A generally accepted ethical principle is that care available in prison settings should be equivalent to that available to the wider community [2]. Ethical concerns would arise if new therapies were disproportionately targeted on those in custody and other treatments of proven effi cacy denied.

From a human rights perspective, the following principles could be taken to be requirements for treatment to be regarded as ethical:

1) There should be rigorous evidence of the safety and effectiveness of the treatment that is provided;

2) Effective treatment should be provided safely in well-structured, well-resourced and well-managed treatment programmes;

3) Human rights law should be clearly understood and prioritised over the competing claims of the public interest. A balance must be found between these competing claims and this should be expressed in the ethical values of autonomy, liberty, privacy and consent;

4) Restricting individual rights in the public interest must only be done for compelling reasons based on empirical, clinical and scientifi c evidence;

5) Policies should observe the ethical values of respecting patients’ autonomy by defi ning the constraints of their liberty and by ensuring that they give free and informed consent to participate in treatment, protecting their privacy of information;

6) Treatment programmes should ensure that dependent persons have equitable access to treatment which maximises its effectiveness for each individual (by matching patients to the treatment that meets their individual needs and situation), and ensures that they do not bear a disproportionate social burden in accepting treatment;

7) It is important that pharmacological treatment should not be used to compensate for poor social policies that lead some to drug abuse and addiction, contribute to a general erosion of human rights, or inappropriate ‘public interest’ drug policies that may be over-focused on the negative and criminal impact of drug addiction. [15, 17, 21].

ConclusionModern developments in neurobiology help us to better understand how addiction

Page 149: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

147

develops. These new studies may bring a basis for new psychological and pharmacological treatments and prevention strategies. Unfortunatelythe data can be inappropriately used to justify involuntary, high invasive or even harmful treatmentof persons, addicted to psychoactive substances, without a suffi cient concern for human rights and ethical implications.In our opinion a balanced approach is required in addiction policies with a concern for human rights and ethical principles.

References

[1]. Ashcroft, R. and Franey, C. 2004. Further Ethical and Social Issues in Using a Cocaine Vaccine: Response to Hall and Carter, Journal of Medical Ethics 30: 341–343.

[2]. Ashworth, A. 2003. Principles of Criminal Law, 4th edition. Oxford: Oxford University Press.[3]. British Medical Association (BMA) 2001. The Medical Profession and Human Rights: Handbook for a

Changing Agenda. London: Zed Books and BMA.[4]. Caplan, A. 2002. No-brainer: Can We Cope with the Ethical Ramifi cations of New Knowledge of the

Human Brain? In: Marcus, S. (ed.). Neuroethics: Mapping the Field. New York: Dana Press. pp. 95–106.[5]. Capron, A. 1999. Ethical and Human Rights Issues in Research on Mental Disorders that may Affect

Decision-Making Capacity. New England Journal of Medicine 340: 1430–1434.[6]. Centre for Cognitive Liberty and Ethics (CCLE). 2004. Threats to Cogitative Liberty: Pharmacotherapy and

the Future of the Drug War. Davis, California: CCLE.[7]. Crabbe, J. and Phillips, T. 1998. Genetics of Alcohol and Other Abused Drugs. Drug and Alcohol

Dependence 51: 61–71. Crombag, H. and Robinson, T. 2004. Drugs, Environment, Brain and Behaviour. Current Directions in Psychological Science 13: 107–111.

[8]. Darke, S. and Hall, W. 2003. Heroin Overdose: Research and Evidence-Based Intervention. Journal of Urban Health 80: 189– 200.

[9]. Farah, M. and Wolpe, P. 2004. Monitoring and Manipulating Brain Function: New Neuroscience Technologies and their Ethical Implications. Hasting Centre Report 43: 35–45.

[10]. Hall, W., Carter, L. and Morley, K. 2003, Addiction, Neuroscience and Ethics. Addiction 98: 867–970.[11]. Hall, W., Carter, L. and Morley, K. 2004, Neuroscience Research on the Addictions: A Prospectus for Future

Ethical and Policy Analysis. Addictive Behaviours 29: 1481–1495.[12]. Hall, W., Morley, K. and Lucke, J. 2004, The Prediction of Disease Risk in Genomic Medicine. EMBO

Reports 5: (special issue) S22–S26.[13]. Husak, D. 2004. The Moral Relevance of Addiction. Substance Use and Misuse 39: 399–436.[14]. Kleiman, M. 2003. The ‘ Brain Disease’ Idea, Drug Policy and Research Ethics. Addiction 98: 871–872.[15]. Mason, J., McCall Smith, R. and Laurie, G. 2003. Law and Medical Ethics, 6th edition. London: LexisNexis

Butterworths.[16]. Mohn, A.R., Yao, W.D. and Caron, M.G. 2004. Genetic and Genomic Approaches to Reward and Addiction.

Neuropharmacology 47 (Supplement 1): 101–110.[17]. Morse, S. 2000. Hooked on Hype: Addiction and Responsibility. Law and Philosophy 19: 3–49.[18]. Morse, S. 2004. Medicine and Morals, Craving and Compulsion. Substance Use and Misuse 39: 437–460.[19]. Nuffi eld Council on Bioethics (NCB). 2003. Pharmacogenetics: Ethical Issues. London: Nuffi eld Council

on Bioethics. [20]. ]Nutt, D. 1997. The Neurochemistry of Addiction. Human Psychopharmacology 12: S53–S58.[21]. Watson, G. 1999. Excusing Addiction. Law and Philosophy 19: 589– 619.[22]. World Health Organisation (WHO). 2004. Neuroscience of Psychoactive Substance Use and Dependence.

Geneva: WHO. [23]. Worthington, C. and Myers, T. 2003. Factors Underlying Anxiety in HIV Testing: Risk Perceptions, Stigma,

and the Patient– Provider Power Dynamic. Qualitative Health Research 13: 636– 655.

Page 150: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

148

Chapter 18

Quercetin Use and Human Health: Risk and Benefi ts

Virginia TZANKOVAa, Stoyan DIRIMANOVb1

aMedical University of Sofi a, Faculty of Pharmacy, Dunav 2 Str. 1000 bUniversity of Würzburg, Institute of Pharmacology and Toxicology, Versbacher Str. 9

Abstract: The use of quercetin (a fl avonoid aglicone notably presented in some dietary plants) as a dietary supplement is growing in the last few decades. Considerable epidemiological evidences indicate that quercetin exhibits plenty of benefi cial effects on human health ( antioxidant, anti-proliferative, anti-infl ammatory, vitamin P-like, antithrombotic, vasorelaxant, etc.) Nevertheless, some potential toxicity might be expected, like a phenomenon better known as quercetin paradox. The present rewiew aims to evaluate the toxicity and the safety profi le of quercetin, as well as to critically assess the value of quercetin as a therapeutic and nutraceutical agent. The integrated information from a huge number of in vitro and in vivo studies of quercetin and its pro-drugs, as well as from some clinical trials is assessed. We mainly focused on evaluation of the intrinsic toxicity of quercetin, and the pro-oxidant effects of oxidized quercetin, as well as its interactions with drug-metabolizing enzymes and systems. We especially emphasized on inhibition/induction of cytochrome P450, xanthine oxidases (XO), N-acethyltransferase (NAT). Conclusion: The analysis assuming the data from preclinical and clinical studies indicates that quercetin exhibits signifi cant benefi ts for the human health, while it is unlikely to cause severe adverse effects in case of proper preparation and use.

Key words: Flavonoids, Quercetin, Antioxidant, Enzyme interactions, Adverse effects

Introduction Flavonoids are plant-derived polyphenolic compounds. They were found in fruits,

vegetables, nuts, seeds, herbs, spices, stems, fl owers, as well as tea and red wine. They are prominent components of many food sources [1] and are consumed regularly within the human diet. Over 4000 structurally unique fl avonoids have been identifi ed in plant sources [2, 3, 4, 5]. These low molecular weight substances are phenylbenzo-pyrones (phenylchromones) with an assortment of structures based on a common three ring nucleus. This basic structure is comprised of two benzene rings (A and B) linked through a heterocyclic pyran or pyrone (with a double bond) ring (C) in the middle (Figure 1).

1 Corresponding Author: Stoyan Dirimanov, University of Wьrzburg, Institute of Pharmacology and Toxicology, Versbacher Str. 9, Wьrzburg, Germany; e-mail: [email protected]

Page 151: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

149

Figure 1. Common Structure of Flavonoids

Due to their structure, in fact presence in the molecule of active phenol hydroxyl groups and a carbonyl group, fl avonoids display high biological activity and diverse effects. One of the most important fl avonoids in terms of biological activity and presence in the human diet is quercetin (3,3’,4’,5,7-pentahydroxyfl avone). It is found in abundance in grapes, apples, onions, some beverages (red wine, tee). A lot of efforts were concentrated on studying quercetin because of the plethora of benefi cial effects on human health. Quercetin has antioxidant, anti-proliferative, anti-infl ammatory activities (the last is especially used in treatment of gout, pancreatitis and prostatitis [6]). Furthermore, it manifests profi table effects on the cardiovascular system, such as prevention of platelets aggregation (antithrombotic), prevention of atherosclerotic plaque formation, modifi cation of eicosanoid biosynthesis, [7] promotion of relaxation of vascular smooth muscles (antihypertensive effect), reduction of left ventricular hypertrophy, improvement of endothelial function etc [8].

These facts clearly demonstrate the reason for growing use of quercetin in the last few decades as a therapeutic agent and as a dietary supplement, as well. The present study aims to evaluate the toxicity and the safety profi le of quercetin, as well as to critically assess the value of quercetin as a therapeutic and nutraceutical agent. The safety profi le of quercetin is subjected to various discussions. For that purpose, full range of in vitro and in vivo studies, concerning the quercetin’s toxicity were conducted (see references). We integrated this data with the information available from clinical trials, in order to receive a more comprehensive image of the fl avonol’s safety.

In 20th century, quercetin was approved for nutritional use. Although it was wide-spread as a food additive, data about its safety was insuffi cient. The problem of quercetin toxicity was elucidated for the fi rst time in more details by the International Agency for Research on Cancer (IARC). The article integrates information from different studies about quercetin’s carcinogenic potential and toxicity. Overall, IARC concludes that quercetin is “not classifi able as to its carcinogenicity to humans” [9].

Toxicity of Quercetin2.1. Acute, Subchronic, Chronic ToxicityThe data from long-term toxicity studies show that quercetin is well-tolerated after oral

admission (see below). An in vivo rabbit toxicity experiment was performed to evaluate the acute toxicity of the fl avonoid. No symptoms of toxicity were observed following intravenous dose of quercetin of 100-150 mg/kg b.w. [26].

Page 152: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

150

Ruiz et al. [44] carried out a 28-day study that addressed subchronic toxicity of quercetin. The doses of 30, 300, 3000 mg/kg b.w./day quercetin were given to male and female Swiss mice. No signifi cant differences in some standard toxicological parameters, clinical chemistry parameters, and organ weights were detected in comparison to the control animals. Again, another study on Swiss mice did not describe any signifi cant changes following quercetin administration of 950 mg/kg b.w./day [45].

Considering that the fl avonol is mainly excreted in urine, the National Toxicology program performed a study to analyze potential toxicity in the renal system. Chronic nephropathy, hyperplasia, and neoplasia of the renal tubular epithelium were observed in male, but not in female F344/N rats exposed to approximately 2000 mg/kg b.w./day. At lower dosages (i.e. 50 and 500 mg/kg b.w./day) no signifi cantly adverse effects were reported [17].

Jazvinscak Jembrek M et al. (2012) [46] claimed that quercetin could have potentially harmful effects on the neurodegenerative prevention, based on 24 h in vitro study with healthy P19 neurons. Morphological changes upon quercetin treatment (up to 150 μM) revealed decrease in lactate dehydrogenase (LDH) activity (“quercetin paradox”). It was reported that intracellular glutathione was depleted. The study particularly emphasized on changes in gene expression of cell survival regulating genes Bcl-2, p53, c-fos. The authors warned about a possible risk of complex intracellular interactions between the affected genes.

2.2. Long-Term ToxicityOne of the fi rst toxicological investigations on quercetin was conducted in 1977 [10].

Ames test reported its mutagenic activity in Salmonella typhimurium strains. This statement was confi rmed also for eukaryotic organisms – in yeasts [11] and in rodent cell-lines [12], but at relativеly high concentrаtions. Quercetin induced chromosomal aberrations, single strand of DNA breaks, and micronuclei formation [12, 13, 14, 15, 16, 17, 18, 19].

Surprisingly, the animal experiments have not validated the results from the in vitro studies. Quercetin was administrated orally to mice and rats for considerably long period of time and no signifi cant changes in mutagenicity/genotoxicity endpoints in somatic cells, in comparison with untreated controls were induced [20, 21, 22, 23, 24, 25].

Numerous of studies investigate the carcinogenic potential of quercetin after continuous oral administration. The studies performed with variety of laboratory animals (ACI and F344 rats, ddY and A/JJms mice, and golden hamsters) at concentrations of up to 10% dietary quercetin (i.e. up to 12 g/kg b.w./day) produced negative results for enhanced induction of any organ tumors in a comparison to the controlls [26, 27, 28, 29, 30, 31, 32, 33, 17]. Only two animal studies have shown positive outcome for occurrences of tumors after quercetin administration. Pamukcu et al. [34] reported that urinary bladder and intestinal tumors in Norwegian rats were observed after quercetin administration. A 2-year rat feeding study of National Toxicology Program (NTP) indicates that quercetin admission (2000 mg/kg b.w./day) caused renal tubule adenomas or adenocarcinomas in male rats, but only following step-section analysis. Female rats were not affected. It must be considered that such dose corresponds to the 140 g intake of quercetin by a 70 kg individual [35, 17]. Quercetin’s carcinogenic potential was evaluated also by topical application and the outcome was negative [9]. The studies for co-administration with known carcinogens are controversial [9].

Page 153: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

151

The fl avonoid has been the subject to various experiments for evaluation of its reproductive/teratogenic potential. The in vitro experiments on mammalian germ cells are with variable results [36, 37, 38, 39]. Quercetin did not induce any sperm abnormalities after oral administration in male Swiss albino mice (dose: up to 150 mg/kg b.w./day) [40], but after intraperitoneal application some induction was observed [41]. The investigations about teratological abnormalities has also negative outcome [42, 32]. It was reported that quercetin can cross placental barrier [43].

2.3. Safety Profi le of Quercetin – Evidences from Clinical and Epidemiological Studies

The data obtained from the clinical and epidemiological studies of products containing a standardized content of quercetin, pure quercetin or its pro-drugs, indicate that the fl avonol is well-tolerated in humans [47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59]. No signifi cant severe adverse events were reported during the pointed studies. In studies investigating pure quercetin or plant extracts containing its glycosides (doses: 3 – 1000 mg) for periods of time up to 12 weeks no negative changes in hematology, clinical chemistry parameters, and urine parameters were detected [60, 61, 62]. A clinical trial phase I, assessing anti-proliferative activity of quercetin in patients not susceptible to the standard cancer therapy, revealed some adverse effects of quercetin administered intravenously in high dosages [63]. They were increasing from 60 mg/m2 to 2000 mg/m2, which may be given in either 3-week or weekly interval. The maximal dose admitted was 2000 mg/m2 (~51.3 mg/kg b.w.). At the fi rst dose levels (up to 420 mg/m2 or 10.8 mg/kg b.w.) the only adverse affects were mild local pain, brief period of fl ushing and sweating. At dose 1400 mg/m2 dyspnea, nausea, and vomiting were reported. Some cardiovascular events occurred, during the trial: hypertension crisis, drop in creatinine clearance, central chest pain, and severe nephrotoxicity, followed by fatal heart failure. At the highest dose of quercetin (1700 and 2000 mg/m2) myelosuppression was not observed, but dose-limiting toxicities were reported: 1 acute renal failure, 2 cases of clinically signifi cant nephrotoxicity, 3 cases of clinically signifi cant renal impairment.

A water-soluble pro-drug of quercetin was also subjected to investigation in a clinical trial [64]. No severe adverse effects followed oral or intravenous administration of quercetin.

Egert et al., (2008) [65], Reshef N. et al., (2005) [66], Taubert D. et al., (2007) [67], Conquer et al., (1998) [47], Edwards RL et al., (2007) [68] performed clinical trials to assess the benefi cial effects of quercetin and quercetin-rich foods on the cardiovascular system. Similarly, these studies did not show any signifi cant adverse events.

Reid et al., (2004) [69] described isolated clinical case report related to quercetin/bromelanin supplement use. 69-years-old male patient diagnosed with lung carcinoma decided to start self- treatment with quercetin (400 mg/day) and bromelanin (100 mg/day). After fi ve weeks he had elevated liver enzyme activity, i.e. AST (Aspartate transaminase) (265%), Alk Phos (140%), and LDH (Lactate dehydrogenase) (112%) compared to the normal levels. 14 days after cessation of quercetin/bromelanin treatment, the enzyme levels were normalized. As this is a single case report, it must be discussed with caution in regard with its clinical interpretation.

In addition, a considerable number of epidemiological studies support the absence of

Page 154: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

152

any relationship between quercetin consumption from the human diet (up to 68 mg/day) and an increased risk of cancer [70, 71, 72, 73, 74, 75 ] Goldbohm et al., 1995; Knekt et al., 1997, 2002; Lam TK et al., 2010; Lin et al., 2006). Knekt et al., (1997, 2002) [73, 74] demonstrated an inverse association between total fl avonoid intake (a majority of which quercetin ~95%), and overall cancer incidence, as well as the incidence of lung cancer. In 2010, Lam TK et al. [75] investigate the relationship between dietary quercetin intake, quercetin-gene interaction, metabolic gene expression in lung tissue, and lung cancer risk. Thus, inverse association of quercetin-rich food intake and lung cancer risk exists. Furthermore, a possible mechanism of quercetin-related changes in expression of genes involved in the tobacco carcinogens metabolism in humans is elucidated.

3. Quercetin paradox/Pro-oxidative effects of quercetin Quercetin is well-known to possess excellent anti-oxidant activity. It is reported that

quercetin can protect DNA against damage induced by strong oxidants. However, in certain circumstances quercetin may act as a pro-oxidant (a phenomenon, better-known as “quercetin paradox” explained by Boots et al., 2007 [76]). When quercetin scavenges free radicals, it is chemically converted into oxidation products (such as ortho-quinone/quinone methide intermediates). These products display a high reactivity towards thiols and this can be a reason for the loss of some protein functions. Boots et al. found that quercetin pre- treatment can protect DNA from oxidative damage. However, furthermore, it was found that quercetin, in presence of hydrogen peroxide, leaded to signifi cant lactate dehydrogenase (LDH) leakage and intracellular glutathione depletion, respectively affecting cell viability (Figure 2). Thus, the authors concluded that the apparent anti-oxidative protection offered by quercetin is a trade-off with other type toxicity.

Figure 2. “ Quercetin paradox” (Boots et al., 2007). In absence of quercetin, DNA is damaged by the oxidative factor, respectively by the formed free oxygen radicals; In presence of quercetin it scavenges free radicals and protects the DNA. However, during this process quercetin is converted into its oxidized form. It is thiol reactive, which leads to protein binding, GSH consumption, an increase in intracellular free calcium concentration and LDH leakage [76].

Page 155: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

153

4. Quercetin drug interactionsThe fi rst report that more seriously raised the possibility of fl avonoid-drug interactions

is of Galati and O’Brien [77]. Various in vitro, in vivo and human experiments were conducted. In 1985 Sousa RL, et al [78] revealed that quercetin inhibits cytochrome P-450 activity in rat liver microsomes. Roots et al [79] reported that quercetin inhibits CYP1A1 in a genotype-selective mode. In some studies it is reported that quercetin has a potent CYP3A inhibitor activity [80]. Choi EJ et al. (2012) [81] reported that the fl avonoid downregulates CYP1A1 and CYP1B1 in mice. A human study investigated quercetin’s ability to modulate the activity of several drug-metabolising enzymes. 12 unrelated healthy volunteers were involved and as probe drug was used caffeine. Urinary caffeine metabolite ratios were used as indicators of the activity of CYP1A2, CYP2A6, NAT and XO. Quercetin affects activity of the enzymes in vivo. The results of this study indicate that quercetin inhibits CYP1A2 function, but enhances CYP2A6, NAT-2 and XO activity [82]. Quercetin is reported to inhibit a drug-effl ux transporter P-glicoprotein (Pgp) in vivo [83]. In 2004, Wang YH [84] performed an experiment on pigs, revealed lethal interaction between quercetin and digoxin due to P-glicoprotein inhibition. Concomitant administration of digoxin and quercetin/quercetin-containing plants/dietary supplements to be avoided.

Interactions between quercetin and a different drugs have been studied, particularly because of its interactions with CYP3A4 and Pgp. Observations in animals and humans prove some signifi cant changes in quercetin bioavailability: decrease of the cyclosporine and simvastatin bioavailability, increased bioavailability of etoposide, doxorubicin, paclitaxel, digoxin, tamoxifen and verapamil [85, 86, 87, 88, 83, 84, 89, 90, 91, 92, 93]. The studies suggest that quercetin infl uences the bioavailability, when admitted for a long period of time or 30 min before drug intake (Table 1).

In addition, in BAYER clinical review form 2013 about Riociguat (Adempas- a drug undergoing a clinical trial phase III), quercetin is considered as inhibitor of human CYP1A1 [94].

Table 1. The Effect of Quercetin on Drug Bioavailability (by in vivo and human studies) [98]

Drug Name Quercetin Dosing Drug Bioavailability Notes

Cyclosporin Co-administration Decreased Interaction with CYP3A4 and Pgp (Rat and Pig Study)

Digoxin Co-administration Increased Potentially lethal interaction, Pgp (Pig Study)

Etoposide Co-administration Increased CYP3A4- and Pgp- Interactions (Rat Study)

Nifedipine Co-administration with 400 mg of quercetin No signifi cant change ( Human Study)

Paclitaxel Pretreatment prior to Paclitaxel Increased (Rat Study)

Simvastatin Pretreatment for 1 week Decreased CYP3A4-interaction (Pig Study)

Tamoxifen Co-administration Increased (Rat study)

Verapamil Pretreatment prior to Verapamil Increased CYP3A4-interaction (Rabbit Study)

Page 156: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

154

5. DiscussionDue to the variety of benefi cial effects of quercetin on human health, its popularity in

the clinical practice and use as a dietary supplement is growing in the last few decades. Considerable epidemiological evidences indicate that quercetin exhibits plenty of benefi cial effects on human health ( antioxidant, anti-proliferative, anti-infl ammatory, vitamin P-like, antithrombotic, vasorelaxant, etc). A big number of protocols for clinical trials are available for the past several years [95]. Widespread use of the fl avonoid raises the issue for further evaluation of its safety profi le. Quercetin was reported to possess mutagenic activity in vitro [10], but this result has not been confi rmed by in vivo studies [23]. Possible reasons for that is lower intrinsic bioavailability of the fl avonoid after oral administration and also protective mechanisms against oxidized products of quercetin, acting within the organism in vivo. Only two animal studies [34, 17] succeed to prove quercetin carcinogenic potential. Considering that, IARC claimed that quercetin is “not classifi able as to its carcinogenicity to humans” [9]. In addition, a review by Okamoto on “Safety of Quercetin for Clinical Application” emphasized on safety of Quercetin and concluded that quercetin is genotoxic to Salmonella typhimurium but its safety upon human application is approved [96]. In agreement with this statement, a number of epidemiological studies demonstrated an inverse association between total quercetin intake, and overall cancer incidence [73, 74]. The data from several clinical trials also displays that quercetin is well-tolerated in humans. In very high dosages, a myelosuppression was not detected, but other dose-limiting toxicities appeared: acute renal failure, nephrotoxicity, renal impairment [63]. The mechanism of renal toxicity has not yet been elucidated. Possibilities include alternation of renal blood fl ow or direct renal tubular damage.

Dirimanov, Fusi, and Tzankova (2010) carried out some preliminary studies on the vasorelaxant properties of several quercetin pro-drugs, in order to investigate their ability to cause adverse hemodynamic effects (data not published).

The possible reason for the observed toxicity of the fl avonoid is the pro-oxidative effect of quercetin, known also as “quercetin paradox”. The oxidized forms of quercetin can interact with DNA, some proteins and glutathione. Upon glutathione depletion, LDH leakage can occur and to infl uence cell viability [76].

Further investigations are needed to contribute for more complete outlook on the quercetin safety profi le. Quercetin-related changes in the gene expression must be studied in more details with an emphasis on possible risks of complex intracellular interactions between the affected genes. In addition, quercetin is reported to interact with a variety of important signal pathways. Thus, the fl avonoid can affect some vital processes such as division, differentiation, gene expression, apoptosis, secretion, smooth muscle relaxation, etc. For instance, quercetin was shown to inhibit protein kinase C (PKC), which is involved in large range of cellular activities like tumor promotion, mitogenesis, secretory processes, infl ammatory cell function, and T-lymphocyte function [97]. The fl avonol is known to inhibit also phosphatidyl inositol-3 kinase and tyrosine kinase. This should be taken into account in case of increased bioavailability of quercetin within the body, i.e. pro-drugs, or quercetin-loaded nanoparticles.

Page 157: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

155

6. ConclusionThe analysis assuming the data from preclinical and clinical studies indicates that

quercetin exhibits signifi cant benefi ts for the human health, while it is unlikely to cause severe adverse effects in case of proper preparation and use.

References

[1] K Herrmann(1976) Flavonols and fl avones in food plants: A review. J Food Technol 11:433–448.[2] JB Harborne (1986) Nature, distribution, and function of plant fl avonoids, inPlant Flavonoids in Biology

and Medicine: Biochemical, Pharmacological, andStructure-Activity Relationships (Cody V, Middleton E, Jr and Harborne JBeds) pp 15-24, Alan R. Liss, Inc., New York

[3] JB Harborne (1988a) The Flavonoids: Advances in Research Since 1986.Chapman & Hall, New York[4] JB Harborne(1988b) Flavonoids in the environment: Structure-activityrelationships, in Plant Flavonoids in

Biology and Medicine II: BiochemicalCellular and Medicinal Properties (Cody V, Middleton E, Harborne JB andBeretz A eds) pp 17-28, Alan R. Liss, Inc, New York

[5] JB Harborne,TJ Mabry, H Mabry (1975) The Flavonoids. Academic Press, NewYork [6] KV Hirpara, P Aggarwal, AJ Mukherjee, N Joshi, AC Burman (2009) Quercetin and its derivatives:

synthesis, pharmacological uses with special emphasis on anti-tumor properties and prodrug with enhanced bio-availability Anticancer Agents Med Chem. 2009 Feb;9(2):138-61.

[7] JV Formica, W Regelson (1995) Review of the biology of Quercetin and relatedbiofl avonoids. Food Chem Toxicol 33:1061-1080

[8] Perez-Vizcaino,M Ibarra, AL Cogolludo, Duarte J, Zaragoza-Arnaez F, Moreno L, Lopez-Lopez G, Tamargo J (2002) Endothelium-independent vasodilator effects of the fl avonoid quercetin and itsmethylated metabolites in rat conductance and resistance arteries. J Pharmacol Exp Ther 302:66-72

[9] International Agency for Research on Cancer (IARC)Monographs on the Evaluation of Carcinogenic risks to Humans”, volume 73

[10] L.F.Bjeldanes, G.W. Chang, 1977. Mutagenic activity of quercetin andrelated compounds. Science 197, 577–578.[11] A.A.Hardigree, J.L.Epler, 1978. Comparative mutagenesis of plantfl avonoids in microbial systems. Mutat.

Res. 58, 231–239.[12] M.A.Yoshida, M.Sasaki, Sugimura,Kawachi, T., 1980. Cytogeneticeffects of quercetin on cultured

mammalian cells. Proc. Jpn. Acad. Ser.B 56, 443–447.[13] J.H.Carver, A.V.Carrano, J.T.MacGregor, 1983. Genetic effects of thefl avonols quercetin, kaempferol, and

galangin on Chinese hamsterovary cells in vitro. Mutat. Res. 113, 45–60.[14] J.Rueff, A.Laires, H.Borba, T.Chaveca, M.Gomes, M.Halpern, 1986. Genetic toxicology of fl avonoids:

The role of metabolicconditions in the induction of reverse mutation, SOS function andsister-chromatid exchanges. Mutagenesis 1, 179–183.

[15] R.Kubiak, Z.Rudek, 1990. SCEs and chromosome aberrations inmammalian cells in vitro treated with quercetin. Acta Biol. Hung. 41,121–124.

[16] R.Popp, O.Schimmer, 1991. Induction of sister-chromatid exchanges(SCE), polyploidy, and micronuclei by plant fl avonoids in humanlymphocyte cultures. A comparative study of 19 fl avonoids. Mutat.Res. 246, 205–213.

[17] NTP, 1992. Toxicology and Carcinogenesis Studies of Quercetin (CASNo., 117-39-5) in F344/N Rats (Feed Study). NTP Technical ReportSeries, No. 409. National Toxicology Program (NTP), ResearchTriangle Park, North Carolina. Available from: <http://ntp.niehs.nih.gov/ntp/htdocs/LT_rpts/tr409. pdf>.

[18] J.Gasper, A.Rodrigues, A. Laires, F.Silva, S.Costa, Monteiro, M.J.,Monterio, C., Rueff, J., 1994. On the metabolisms of genotoxicity andmetabolism of quercetin. Mutagenesis 9, 445–449.

[19] H.Caria, T.Chaveca, A.Laires, J.Rueff, 1995. Genotoxicity ofquercetin in the micronucleus assay in mouse bone marrow erythrocytes, human lymphocytes, V79 cell line and identifi cation of kinetochore-containing (crest staining) micronuclei in human lymphocytes.Mutat. Res. 343, 85–94.

[20] J.T.MacGregor, 1979. Mutagenicity studies of fl avonoids in vivo andin vitro. Toxicol. Appl. Pharmacol. 48, A47

[21] H.-U.Aeschbacher, H.Meier, E.Ruch, 1982. Nonmutagenicity in vivoof the food fl avonol quercetin. Nutr. Cancer 4, 90–98.

Page 158: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

156

[22] M. Ishikawa, T. Oikawa, M. Hosokawa, J. Hamada, K. Morikawa, Kobayashih, H., 1985. Enhancing effect of quercetin on 3-methylcholanthrenecarcinogenesis in C57Bl/6 mice. Neoplasma 32, 435–442.

[23] A.J.Ngomuo, R.S.Jones, 1996. Genotoxicity studies of quercetin andshikimate in vivo in the bone marrow of mice and gastric mucosal cellsof rats. Vet. Hum. Toxicol. 38, 176–180.

[24] S.Taj, B.Nagarajan, 1996. Inhibition by quercetin and luteolin ofchromosomal alterations induced by salted, deep-fried fi sh and muttonin rats. Mutat. Res. 369, 97–106.

[25] A.Cierniak, M.Papiez, M.Kapiszewska, 2004. Modulatory effect ofquercetin on DNA damage, induced by etoposide in bone marrow cellsand on changes in the activity of antioxidant enzymes in rats. Rocz. Akad. Med. Bialymst. 49, 167–169.

[26] A.M.Ambrose, D.J.Robbins, F.DeEds, 1952. Comparative toxicitiesof quercetin and quercitrin. J. Am. Pharm. Assoc. Sci. Ed. 41, 119–122.

[27] D.Saito, Shirai, A., Matsushima, T., Sugimura, T., Hirono, I., 1980. Testof carcinogenicity of quercetin, a widely distributed mutagen in food.Teratog. Carcinog. Mutagen. 1, 213–221.

[28] I.Hirono, Ueno, I., Hosaka, S., Takanashi, H., Matsushima, T.,Sugimura, T., Natori, S., 1981. Carcinogenicity examination ofquercetin and rutin in ACI rats. Cancer Lett. 13, 15–21.

[29] S.Hosaka, Hirono, I., 1981. Carcinogenicity test of quercetin bypulmonary–adenoma bioassay in strain A mice. GANN 72, 327–328.

[30] K.Morino, Matsukara, N., Kawachi, T., Ohgaki, H., Sugimura, T.,Hirono, I., 1982. Carcinogenicity test of quercetin and rutin in goldenhamsters by oral administration. Carcinogenesis 3, 93–97.

[31] H.Takanashi, Aiso, S., Hirono, I., Matsushima, T., Sugimura, T., 1983.Carcinogenicity test of quercetin and kaempferol in rats by oraladministration. J. Food Saf. 5, 55–60.

[32] G.S.Stoewsand, Anderson, J.L., Boyd, J.N., Hrazdina, G., Babish, J.G.,Walsh, K.M., Losco, P., 1984. Quercetin: A mutagen, not a carcinogen,in Fischer rats. J. Toxicol. Environ. Health 14, 105–114.

[33] N. Ito, Hagiwara, A., Tamano, S., Kagawa, M., Shibata, M., Kurata, Y., Fukushima, S., 1989. Lack of carcinogenicity of quercetin in F344/DuCrj rats. Jpn. J. Cancer Res. 80, 317–325.

[34] A.M.Pamukcu, Yalcёiner, S_., Hatcher, J.F., Bryan, G.T., 1980. Quercetin,a rat intestinal and bladder carcinogen present in Bracken Fern(Pteridium aquilinum). Cancer Res. 40, 3468–3472.

[35] J.K.Dunnick, Hailey, J.R., 1992. Toxicity and carcinogenicity studies ofquercetin, a natural component of foods. Fundam. Appl. Toxicol. 19,423–431.

[36] L.Nass-Arden, Breitbart, H., 1990. Modulation of mammalian spermmotility by quercetin. Mol. Reprod. Dev. 25, 369–373.

[37] D.K.Swisher, Harris, R.D., Tague, R.M., Seyler, D.E., 1994. Effects ofquercetin on rabbit sperm. Biol. Reprod. 50, 107 (Abstract no. 211).

[38] R.Trejo, Valadeґz-Salazar, A., Delhumeau, G., 1995. Effects of quercetinon rat testis aerobic glycolysis. Can. J. Physiol. Pharmacol. 73, 1605–1615.

[39] K.L.Khanduja, Verma, A., Bhardwaj, A., 2001. Impairment of humansperm motility and viability by quercetin is independent of lipidperoxidation. Andrologia 33, 277–281.

[40] S.D.Nandan, Rao, M.S., 1983. Lack of mutagenic effects of quercetin inthe germ cells of mice. IRCS Med. Sci. 11, 210.

[41] P.B.Rastogi, Levin, R.E., 1987. Induction of sperm abnormalities in miceby quercetin. Environ. Mutagen. 9, 79–86.

[42] C.C.Willhite, 1982. Teratogenic potential of quercetin in the rat. FoodChem. Toxicol. 20, 75–77.[43] K. Vanhees , Godschalk RW, Sanders A, van Waalwijk van Doorn-Khosrovani SB, van Schooten

FJ. Maternal quercetin intake during pregnancy results in an adapted iron homeostasis at adulthood. Toxicology. 2011 Dec 18;290(2-3):350-8. doi: 10.1016/j.tox.2011.10.017. Epub 2011 Oct 28.

[44] M.J. Ruiz, Fernaґndez, M., Estela, J.M., Asensi, M., Man˜ es, J., Picoґ, Y., 2006. Short-term oral toxicity of quercetin and pterostibene in Swiss mice. Toxicol. Lett. 164S, S1–S324 (Abstract P22-15).

[45] J. Barrenetxe, Aranguren, P., Grijalba, A., Martinez-Penuela, J.M., Marzo, F., Urdaneta, E., 2006. Effect of dietary quercetin and sphingomyelin on intestinal nutrient absorption and animal growth. Br. J. Nutr. 95, 455–461.

[46] Jazvinšćak Jembrek M, Vuković L, Puhović J, Erhardt J, Oršolić N. Neuroprotective effect of quercetin against hydrogen peroxide-induced oxidative injury in P19 neurons. J Mol Neurosci. 2012 Jun;47(2):286-99. doi: 10.1007/s12031-012-9737-1. Epub 2012 Mar 14.

[47] J.A. Conquer, Maiani, G.; Azzini, E.; Raguzzini, A.; Holub, B.J. 1998. Supplementation with quercetin markedly increases plasma quercetin concentrations without effect on selected risk factors for heart disease in healthy subjects. J Nutr 128:593-597.

[48] K.J. Cureton, Tomporowski, P.D., Singhal, A., Pasley, J.D., Bigelman, K.A., Lambourne, K., Trilk, J.L.,

Page 159: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

157

McCully, K.K., Arnaud, M.J., and Zhao, Q. (2009) Dietary quercetin supplementation is not ergogenic in untrained men. J Appl. Physiol. 107(4):1095-104.

[49] J.M. Davis, Carlstedt, C.J., Chen, S., Carmichael, M.D., and Murphy, E.A. 2010. The dietary fl avonoid quercetin increases V02 max and endurance capacity. Int. J Sport Nutr. Exerc. Metab. 20(1):56-62.

[50] C.L. Dumke, Nieman, D.C.; Utter, A.C., Rigby, M.D.; Quindry, J.C.; Triplett, N.T.; McAnulty, S.R.; McAnulty, L.S. 2009. Quercetin’s effect on cycling effi ciency and substrate utilization. Appl Physiol Nutr Metab 34:993-1000.

[51] D.A. Henson, Nieman, D.C.; Davis, J.M.; Murphy, E. A.; Carmichael, M.D.; Dumke, C.L.; Gross, S.J.; Jenkins, D.P.; Quindry, J.C.; Mcanulty, S.R.; Mcanulty, L.S.; Utter, A.C.; Mayer, E.P. 2008. Post-I 60-km race illness rates and decreases in granulocyte respiratory burst and salivary IgA output are not countered by quercetin ingestion, Int J Sports Med. 29 ( 10):856-863.

[52] H.S. MacRae, and Mefferd, K.M. (2006) Dietary antioxidant supplementation combined with quercetin improves cycling time trial performance. Int. J Sport Nutr. Exerc. Metab. 6(4):405-419.

[53] S.R. McAnulty, McAnulty, L.S.; Nieman, D.C.; Quindry, J.C.; Hosick, P.A.; Hudson, M.H.; Still, L.; Henson, D.A.; Milne, G.L.; Morrow, J.D.; Dumke, C.L.; Utter, A.C.; Triplett, N.T.; Dibarnardi, A. 2008. Chronic quercetin ingestion and exercise-induced oxidative damage and infl ammation. Appl Physiol Nutr Metab 33(2):254-262.

[54] D.C. Nieman, Henson, D.A.; Davis, J.M.; Murphy, E.A.; Jenkins, D.P.; Gross, S.J.; Carmichael, M.D.; Quindry, J.C.; Dumke, C.L.; Utter, A.C.; McAnulty, S.R.; McAnulty, L.S.; Triplett, N.T.; Mayer, E.P. 2007a. Quercetin’s infl uence on exercise-induced changes in plasma cytokines and muscle and leukocyte cytokine mRNA. J Appl Physiol 103(5):1728-1735.

[55] D.C. Nieman, Henson, D.A.; Davis, J.M.; Dumke, C.L.; Gross, S.J.; Jenkins, D.P.; Murphy, E.A.; Carmichael, M.D.; Quindry, J.C.; McAnulty, S.R.; McAnulty, L.S.; Utter, A.C.; and Mayer, E.P. 2007b. Quercetin ingestion does not alter cytokine changes in athletes competing in the Western States Endurance Run. J Interferon Cytokine Res 27(12):1003-1011.

[56] D.C. Nieman, Henson, D.A.; Gross, S.J.; Jenkins, D.P.; Davis, J.M.; Murphy, E.A.; Carmichael, M.D.; Dumke, C.L.; Utter, A.C.; Mcanulty, S.R.; Mcanulty, L.S.; Mayer, E.P. 2007c. Quercetin reduces illness but not immune perturbations after intensive exercise. Med Sci Sports Exerc 39(9):1561-1569.

[57] D.C. Nieman, Henson, D.A.; Maxwell, K.R.; Williams, A.S.; McAnulty, S.R.; Jin, F.; Shanely, R.A.; Lines, T.C. 2009. Effects of quercetin and EGCG on mitochondrial biogenesis and immunity. Med Sci Sports Exerc 41(7):1467-1475.

[58] D.C. Nieman, Williams, A.S., Shanely, R.A., Jin, F., McAnulty, S.R., Triplett, N.T., Austin, M.D., and Henson, D.A. 2010. Quercetin’s infl uence on exercise performance and muscle mitochondrial biogenesis. Med. Sci. Sports Exerc. 42(2):338-45.

[59] J.C. Quindry, McAnulty, S.R.; Hudson, M.B.; Hosick, P.; Dumke, C.; McAnulty, L.S.; Henson, D.; Morrow, J.D.; Nieman D. 2008. Oral quercetin supplementation and blood oxidative capacity in response to ultramarathon competition. Int. J Sport Nutr. Exerc. Metab. 18(6):601-616.

[60] D.A. Shoskes, Zeitlin, S.I.; Shahed, A.; Rajfer, J. 1999. Quercetin in men with category Ill chronic prostatitis: A preliminary prospective, double-blind, placebo-controlled trial. Urology 54(6): 960-963.

[61] H. Kiesewtter, Koscielny, J., Kalus, U., Vix, J.-M., Peil, H., Petrini, O., van Toor, B.S.J., de Mey, C., 2000. Effi cacy of orally administered extract of red vine leaf AS 195 (folia Vitis viniferae) in chronic venous insuffi ciency (Stages I-II). Arzneimittelforschung 50, 109–117.

[62] X. Lozoya, Reyes-Morales, H., Chavez-Soto, M.A., Martinez-Garcia Mdel, C., Soto-Gonzalez, Y., Doubova, S.V., 2002. Intestinal antispasmodic effect of a phytodrug of Psidium guajava folia in the treatment of acute diarrheic disease. J. Ethnopharmacol. 83, 19–24.

[63] H.J. Cross, Tilby, M., Chipman, K., Ferry, D.R., Gescher, A., 1996.Effect of quercetin on the genotoxic potential of cisplatin. Int. J.Cancer 66, 404–408.

[64] P.J. Mulholland, Ferry DR, Anderson D, et al. Pre-clinical and clinical study of QC12, a water-soluble, pro-drug of quercetin. Ann Oncol 2001;12(2):245-248.

[65] S. Egert, Bosy-Westphal, A.; Seiberl, J.; Kurbitz, C.; Settler, U.; Plachta-Danielzik, S.; Wagner, A.E.; Frank, J.; Schrezenmeir, J.; Rimbach, G.; Wolffram, S.; Muller, M.J. 2009. Quercetin reduces systolic blood pressure and plasma oxidised low-density lipoprotein concentration in overweight subjects with a high-cardiovascular disease risk phenotype: a double-blinded, placebo-controlled cross-over study. Br J Nutr 102(7): 1065-1 074.

[66] N. Reshef, Hayari Y, Goren C, Boaz M, Madar Z, Knobler H (2005) Antihypertensive effect of sweetie fruit in patients with stage I hypertension. AmJHypertens, 18:1360–1363

Page 160: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

158

[67] D. Taubert, Roesen R, Lehmann C, Jung N, Schцmig E(2007)Effects of low habitual cocoa intake onblood pressure and bioactive nitric oxide: a randomized controlled trial. JAMA, 298:49–60.

[68] RL Edwards, Lyon T, Litwin S E, Rabovsky A, Symons J D, Jalili T (2007) Quercetin reduces blood pressure in hypertensive subjects. JNutr, 137:2405–2411.

[69] C Reid, Smith K, Kocha W: Case Report: Quercetin/bromelain useassociated with increased liver enzymes. J Oncol Pharm Practice 2004,10, 225-227.

[70] M.G. Hertog, Feskens, E.J.M.; Hollman, P.C.H.; Katan, M.B.; Kromhout, D. 1994. Dietary fl avonoids and cancer risk in the Zutphen elderly study. Nutr Cancer 22(2):175-184.

[71] M.G.L. Hertog, Kromhout, D.; Aravanis, C.; Blackburn, H.; Buzina, R.; Fidanza, F.; Giampaoli, S.; Jansen, A.; Menotti, A.; Nedeljkovic, S.; Pekkarinen, M.; Simic, B.S.; Toshima, H.; Feskens, E.J.M.; Hollman, P.C.H.; Katan, M.B. 1995. Flavonoid intake and long-term risk of coronary heart disease and cancer in the seven countries study. Arch Intern Med 155(4):381-386

[72] R.A. Goldbohm, Brandt, P.A.; Hertog, M.G.L.; Brants, H.A.M.; van Poppel, G. 1995. Flavonoid intake and risk of cancer: A prospective cohort study. Am J Epidemiol 141(11, Suppl.):S61 [Abstract No. 2431.

[73] P. Knekt, Jarvinen, R.; Seppanen, R.; Heliovaara, M.; Teppo, L.; Pukkala, E.; Aromaa, A. 1997. Dietary intake of fl avonoids and the risk of lung cancer and other malignant neoplasms. Am J Epidemiol 146:223-230.

[74] P. Knekt, Kumpulainen, J.; Jarvinen, R.; Rissanen, H.; Heliovaara, M.; Reunanen, A.; Hakulinen, T.; Aromaa, A. 2002. Flavonoid intake and risk of chronic diseases. Am J Clin Nutr 76(3):560-568.

[75] TK Lam , Rotunno M, Lubin JH, Wacholder S, Consonni D, Pesatori AC, Bertazzi PA, Chanock SJ, Burdette L, Goldstein AM, Tucker MA, Caporaso NE, Subar AF,Landi MT. Dietary quercetin, quercetin-gene interaction, metabolic gene expression in lung tissue and lung cancer risk. Carcinogenesis. 2010 Apr;31(4):634-42

[76] A.W. Boots, Li, H.; Schins, R.P.F.; Duffi n, R.; Heemskerk, J.W.M.; Bast, A.; Haenen, G.R.M.M. 2007. The quercetin paradox. Toxicol Appl Pharmacol 222(1):89-96.

[77] G. Galati, O’Brien, P.J., 2004. Potential Toxicity of fl avonoids and other dietary phenolics: signifi cance for chemopreventive and anticancer properties. Free Radic. Biol. Med. 37 (3), 287–303.

[78] R.L. Sousa, & Marletta, M.A. (1985) Inhibition of cytochrome P-450 activity in rat liver microsomes by the naturally occurring fl avonoid, quercetin. Arch. Biochem. Biophys., 240, 345–357.

[79] D. Schwarz, Kisselev, P. and Roots, I. 2003. St. John’s wort extracts and some of their constituents potently inhibit ultimate carcinogen formation from benzo[a]pyrene-7,8- dihydrodiol by human CYPl AI. Cancer Res. 63(22):8062-8068.

[80] A. Miniscalco , Lundahl J, Regеrdh CG, 1992. Inhibition of dihydropyridine metabolism in rat and human liver microsomes by fl avonoids found in grapefruit juice.J Pharmacol Exp Ther June 1992261:1195-1199

[81] EJ Choi , Kim T, Kim GH. Quercetin acts as an antioxidant and downregulates CYP1A1 and CYP1B1 against DMBA-induced oxidative stress in mice. Oncol Rep. 2012 Jul;28(1):291-6

[82] Y Chen, Xiao P, Ou-Yang DS, Fan L, Guo D (2009) Simultaneous action of the fl avonoid quercetin on cytochrome P450 (CYP) 1A2, CYP2A6, N-acetyltransferase and xanthine oxidase activity in healthy volunteers. Clin Exp Pharmacol Physiol 36(8):828-33

[83] P Limtrakul, Khantamat O, Pintha K. Inhibition of P-glycoprotein function and expression by kaempferol and quercetin. J Chemother 2005;17:86-95.

[84] YH Wang, Chao PD, Hsiu SL, et al. Lethal quercetin-digoxin interaction in pigs. Life Sci 2004;74:1191-1197.[85] SL Hsiu, Hou YC, Wang YH, et al. Quercetin signifi cantly decreased cyclosporin oral bioavailability in

pigs and rats. Life Sci 2002;72:227-235.[86] D Pal, Mitra AK. MDR- and CYP3A4-mediated drug-herbal interactions. LifeSci 2006;78:2131-2145.[87] T Sergent, Dupont I, Van der HeidenE, et al. CYP1A1 and CYP3A4 modulation by dietary fl avonoids in

human intestinal Caco-2 cells. ToxicolLett 2009;191:216-222.[88] JS Choi, Piao YJ, Kang KW. Effects of quercetin on the bioavailability ofdoxorubicin in rats: Role of

CYP3A4 and P-gp inhibition by quercetin. Arch PharmRes 2011;34:607-613.[89] R Cermak, Wein S, Wolffram S, Langguth P. Effects of the fl avonol quercetin on the bioavailability

ofsimvastatin in pigs. Eur J Pharm Sci 2009;38:519-524.[90] X Li, Choi JS. Effects of quercetin onthe pharmacokinetics of etoposide afteroral or intravenous

administration ofetoposide in rats. AnticancerRes 2009;29:1411-1415.[91] JS Choi, Jo BW, Kim YC. Enhancedpaclitaxel bioavailability after oraladministration of paclitaxel or

prodrugto rats pretreated with quercetin. Eur JPharm Biopharm 2004;57:313-318.

Page 161: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

159

[92] SC Shin, Choi JS, Li X. Enhanced bioavailability of tamoxifen after oral administration of tamoxifen with quercetin in rats. Int J Pharm 2006;313:144-149.

[93] JS Choi, Han HK. The effect of quercetin on the pharmacokinetics of verapamil and its major metabolite, norverapamil, in rabbits. J PharmPharmacol 2004;56:1537-1542.

[94] http://www.fda.gov/downloads/advisorycommittees/committeesmeetingmaterials/drugs/cardiovascularandrenaldrugsadvisorycommittee/ucm363541.pdf

[95] http://clinicaltrials.gov/ct2/results?term=quercetin&Search=Search[96] T., Okamoto, 2005. Safety of quercetin for clinical application (Review).Int. J. Mol. Med. 16, 275–278.[97] Alyssa X. WU-ZHANG and Alexandra C. NEWTON. Protein kinase C pharmacology: refi ning the

toolbox. Biochem. J. (2013) 452, 195–209[98] S. Gregory Kelly, ND, Quercetin, 2011. Alternative Medicine Review

Page 162: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

160

Chapter 19

Protective Effect of Aronia melanocarpa Fruit Juice in a Model of Paracetamol-induced Hepatotoxicity

in Rats

S. VALCHEVA-KUZMANOVAa, K. KUZMANOVb

aDepartment of Preclinical and Clinical Pharmacology, Medical University – Varna, Bulgaria

bVivarium, Medical University – Varna, Bulgaria

Abstract: Aronia melanocarpa fruits are extremely rich in polyphenolic substances with pronounced antioxidant and anti-infl ammatory activities. We studied the effect of Aronia melanocarpa fruit juice (AMFJ) in a model of paracetamol-induced hepatotoxicity in rats. AMFJ was applied daily orally at doses of 2.5, 5.0 and 10.0 ml/kg from day 1 to day 7 of the experiment. Paracetamol was applied intraperitoneally (1.0 g/kg) on day 5. Blood and liver ware taken for biochemical investigations on day 7 (48 hours after paracetamol administration). Liver toxicity was estimated by the activities of serum enzymes – aspartate aminotransferase (AST), alanine aminotransferase (ALT) and alkaline phosphatase (ALP). Oxidative stress was estimated by the levels of thiobarbituric acid reactive substances (TBARS) in serum and liver homogenate. The administration of paracetamol caused a signifi cant elevation of serum AST (p < 0.01 vs. control) and ALT (p < 0.05 vs. control), and induced lipid peroxidation as measured by the signifi cant increase of TBARS in serum (p < 0.01 vs. control) and liver (p < 0.05 vs. control). AMFJ at the three tested doses prevented the increase of plasma AST and ALT activities. AMFJ prevented also the elevation of TBARS in the liver at the three applied doses and in the serum at the doses of 5.0 and 10.0 ml/kg. The present results showed that AMFJ had a protective effect in a model of paracetamol-induced liver toxicity as evidenced by the biochemical indices of hepatotoxicity and oxidative stress. This effect is probably due to the polyphenolic ingredients of the juice.

Key words: Aronia melanocarpa fruit juice, paracetamol, hepatotoxicity, rats

IntroductionThe liver is subject to acute and potentially lethal injury by several substances including

some drugs such as paracetamol. Paracetamol is one of the most widely used analgesic and antipyretic drugs worldwide. It is associated with both intentional and accidental poisoning. It is a major cause of liver failure and causes death when taken in excess. Paracetamol is metabolized by conjugation in the liver to non-toxic agents. In overdose the normal conjugative pathways of metabolism become saturated. Excess paracetamol is

Page 163: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

161

then oxidatively metabolized in the liver via the mixed function oxidase P450 system to a toxic metabolite N-acetyl-P-benzoquinoneimine (NAPQI). NAPQI has an extremely short half-life and is rapidly conjugated with glutathione, a sulphydryl donor. Under conditions of excessive NAPQI formation or reduced glutathione store, NAPQI covalently binds to vital proteins and the lipid bilayer of hepatocyte membranes. The result is hepatocellular death and centrilobular liver necrosis [1].

A wide variety of polyphenolic substances present in dietary and medicinal plants possess striking antioxidant and anti-infl ammatory properties. Aronia melanocarpa [Michx.] Elliot (black chokeberry) fruits are extremely rich in phenolic compounds: procyanidins, fl avonoids (mainly from the subclass of anthocyanins) and phenolic acids (chlorogenic and neochlorogenic)[2]. Our previous studies [3,4], as well as the studies of other authors [5-8] have investigated the antioxidant propertiesof Aronia juice, Aronia extract or its phenolic constituentsusing different well established assays.FreshAronia berries possess the highest antioxidant capacity amongberries and other fruits investigated so far as measured withoxygen radical absorbance capacity [5,7].

The aim of the present study was to investigate the effect of Aronia melanocarpa fruit juice (AMFJ) on biochemical parameters of liver toxicity and oxidative stress in a model of paracetamol-induced hepatotoxicity in rats.

Materials and methodsAnimals:Male Wistar rats (200 – 250 g) were used in the experiment. The animals

were kept under the standard conditions of the animal house with 12-h light-dark cycle (light 7:00-19:00) at a temperature 23-25 °C. They had free access to food and drinking water with the exception of the period of 42 hours before paracetamol administration when all experimental groups were deprived of food. All procedures concerning animal treatment and experimentation were in accordance with the European Communities Council Directives 86/609/EEC and the National regulations.

Experimental substances: Paracetamol (acetaminophen) was from Sigma-Aldrich Chemie GmbH (Germany). It was prepared as a suspension using 2% Tween 80 (Merck, Germany) in distilled water. Standart test kids of BioSystems S.A., Barcelona, Spain were used for the measurement of liver enzyme activities. All chemicals used for the determination of thiobarbituric acid reactive substances (TBARS) were of analytical grade and were obtained from Merck (Germany).

AMFJ was prepared from Aronia melanocarpa fruits which were handpicked, crushed and squeezed. The juice was fi ltered, pasteurized at 80 °C for 10 min and stored at room temperature till the experiment. The contents of phenolic substances in 100 ml AMFJ were: total phenolics, 709.3 ±28.1 mg as gallic acid equivalents, determined spectrophotometrically according to the Folin-Ciocalteu procedure [9];total fl avonoids, 189.4 ± 8.6 mg as catechin equivalents, measured by a colorimetric assay developed by Zhishen et al. [10]; total anthocyanins, 106.8 ± 6.2 mg as cyanidin-3-glucoside equivalents, determined by a pH-differential spectrophotometry at pH 1.0 and pH 4.5 [11]; quercetin, 11.8 mg, measured by a high-performance liquid chromatography method [12]. The values were the mean of duplicate determinations of three samples.

Experimental procedure:The animals were randomly divided in fi ve experimental

Page 164: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

162

groups each of 10rats (Table 1). AMFJ was applied daily orally by direct stomach intubation at doses of 2.5, 5.0 and 10.0 ml/kg from day 1 to day 7 of the experiment. The doses of 2.5 and 5.0 ml/kg were diluted with distilled water to a total volume of 10 ml/kg. Paracetamol (1.0 g/kg) was prepared as a suspension with 2% Tween 80. The suspension was applied intraperitoneally at a volume of 4.0 ml/kg on day 5 of the experiment 2 hours after the oral treatment.

Table 1. Experimental procedure

Group Oral treatment From day1 to day 7

Itraperitoneal treatmentOn day 5

Control (C) Distilled water (10.0 ml/kg) 2% Tween 80 (4.0ml/kg) Paracetamol (P) Distilled water (10.0 ml/kg) Paracetamol 1.0 g/kg (susp.)AMFJ2.5 + P AMFJ 2.5 ml/kg Paracetamol 1.0 g/kg (susp.)AMFJ 5 + P AMFJ 5.0 ml/kg Paracetamol 1.0 g/kg (susp.)AMFJ 10 + P AMFJ 10.0ml/kg Paracetamol 1.0 g/kg (susp.)

Serum and liver homogenate preparation: On day 7, the animals were anaesthetized with diethyl ether 48 hours after paracetamol administration and 2 hours after the last treatment with distilled water or AMFJ, respectively. Blood was collected from the sublingual veins. It was centrifuged at 2000 rpm for 10 min and serum was obtained for the biochemical analyses.

After the dacapitation of the animals parts of rat livers were minced in a beaker with a pair of scissors. Samples of liver tissue were homogenized with ice cold Tris/HCl, 50 mM, pH 7.4 (1:10). The homogenate was centrifuged (2000 rpm, 10 min, 4 °С) and the supernatant was used for the biochemical investigations.

Biochemical analyses: Serum activities of liver enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT) and alkaline phosphatase (ALP) were determined spectrophotometrically (Aurius 2021 UV-VIS, Cecil Instruments Ltd, UK) using the standard test kids.

Lipid peroxidation levels were estimated by thethiobarbituric acid (TBA) reaction using the method of Ohkawa et al. [13].The method measures spectrophotometrically the color produced by the reactionof TBA with lipid peroxides (thiobarbituric acid reactive substances, TBARS) at 532 nm. TBARS were determined in nmol/ml serum and nmol/g tissue.Malondialdehyde, the major reactive aldehyde resultingfrom the peroxidation of biological membrane polyunsaturatedfatty acids, was used as a standard.

Statistical analysis: Data were analysed statistically by one-way analysis of variance (ANOVA) followed by Dunnett,s multiple comparison post test. A value of p<0.05 was considered as statistically signifi cant. Data are expressed as mean ± SEM. GraphPad Prism statistical software was used.

ResultsThe liver enzyme activities are presented on Figure 1. The administration of

paracetamol caused a signifi cant elevation of serum AST (p < 0.01 vs. control) and ALT (p < 0.05 vs. control) and had no signifi cant effect on ALP activities in rat serum. AMFJ at the three tested doses prevented the increase of plasma AST and ALT activities and had no

Page 165: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

163

signifi cant effect on ALP activity. Thus, the measured liver enzyme activities in rats treated both with AMFJ and paracetamol did not differ signifi cantly from the control ones.

Figure 1. Effect of AMFJ applied at doses of 2.5, 5.0 and 10.0 ml/kg on the activities of AST (panel A), ALT (panel B) and ALP (panel C) in a model of paracetamol (P)-induced hepatotoxicity in rats.

Values are mean ± SEM; n = 10; *p < 0.05, **p < 0.01 vs. control (C)

C P + P

2.5

AMFJ

+ P5

AMFJ + P

10

AMFJ

0

100

200

300

400 A **

AST

(U/l)

C P + P2.5

AMFJ

+ P5

AMFJ

+ P10

AMFJ

0

50

100

150 *B

ALT

(U/l)

C P + P

2.5

AMFJ

+ P5

AMFJ + P

10

AMFJ

0

20

40

60

80 C

AlP

(U/l)

Page 166: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

164

Paracetamol induced lipid peroxidation as measured by the signifi cant increase of TBARS in liver (p < 0.05 vs. control) and serum (p < 0.01 vs. control) (Figure 2). As is obvious from Figure 2, the TBARS in the livers of rats treated with paracetamol and the three AMFJ doses did not differ signifi cantly from the control ones. In rat serum, there was a dose-dependent effect of AMFJ on the concentration of TBARS. In rats treated with AMFJ at the dose of 2.5 ml/kg the concentration of TBARS in serum was signifi cantly higher (p < 0.05) than the control one. In rats treated with AMFJ at the doses of 5.0 and 10.0 ml/kg the concentrations of TBARS in serum were not signifi cantly different from the control one. Thus, AMFJ prevented the paracetamol-induced elevation of TBARS in the liver at the three applied doses and in the serum at the doses of 5.0 and 10.0 ml/kg.

Figure 2. Effect of AMFJ applied at doses of 2.5, 5.0 and 10.0 ml/kg on the concentration of TBARS in liver (panel A) and serum (panel B) in a model of paracetamol (P)-induced hepatotoxicity

in rats. Values are mean ± SEM; n = 10; *p < 0.05, **p < 0.01 vs. control (C)

DiscussionThere are no literature data for investigation of the effect of Aronia melanocarpa

in paracetamol hepatotoxicity models. In a previous study, AMFJ showed pronounced hepatoprotective effect in a model of carbon tetrachloride-induced liver damage in which the leading mechanism of injury is the oxidative stress [14].

C P + P2.5

AMFJ

+ P5

AMFJ

+ P10

AMFJ

0

200

400

600

800

*A

TB

AR

S (n

mol

/g)

C P + P2.5

AMFJ

+ P5

AMFJ + P

10

AMFJ

0

20

40

60

80

***

B

TB

AR

S (n

mol

/ml)

Page 167: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

165

Literature data suggest that the metabolite of paracetamol NAPQI binds to the mitochondrial proteins and triggers a mitochondrial oxidant stress [15] which fi nally causes activation of apoptosis [16] and cellular necrosis [17]. In humans and rodents, CYP2E1 and CYP1A2 are the major enzymes of paracetamol bioactivation [18]. In particular, CYP2E1 is an important isoform that has been implicated in the generation of reactive oxygen species such as superoxide and hydrogen peroxide and may mediate the toxic effects of a variety of xenobiotic compounds [19,20]. Furthermore, when CYP2E1-knockout mice were challenged with paracetamol, they were found to be considerably less sensitive to its hepatotoxic effects than wild-type animals [21].

The results from the present study showed that AMFJ at the three applied doses decreased the biochemical indices of liver toxicity and oxidative stress in rats. At least two mechanisms might account for that effect of AMFJ. One of them is the extremely high antioxidant activity of the juice and the capacity to scavenge reactive oxygen species which has been very intensively studied [2-8]. The other mechanism might be the effect of the juice on the activity of liver enzymes engaged in the metabolism of paracetamol. The study of Krajka-Kuźniak et al. [22] showed that the forced feeding with chokeberry juice alone decreased the activities of cytochrome CYP1A1 and CYP 1A2, and thepretreatment with the juice further reduced the activity of CYP2E1 which was decreased by N-nitrosodiethylamine.These data suggest that AMFJ might inhibit the generation of the toxic metabolite of paracetamol NAPQI.

In conclusion, this study showed that AMFJ might have a protective effect in paracetamol-induced hepatotoxicity in rats which is probably due to the biological activity of its polyphenolic ingredients. The possible mechanisms of this effect are the antioxidant activity of the juice and its inhibitory effect on enzymes participating in the generation of the toxic paracetamol metabolite.

References:

[1]. L.A. McConnachie, I. Mohar, F.N. Hudson, C.B. Ware, W.C.Ladiges, C. Fernandez, S. Chatterton-Kirchmeier, C.C. White, R.H. Pierce, T.J. Kavanagh, Glutamate cysteine ligase modifi er subunit defi ciency and gender as determinants of acetaminophen-induced hepatotoxicity in mice, Toxicological Sciences99 (2007), 628–636.

[2]. J. Oszmianski, A. Wojdylo, Aronia melanocarpa phenolics and their antioxidant activity,Eurpean Food Research and Technology221 (2005), 809–813.

[3]. S. Valcheva-Kuzmanova, V. Gadjeva, D. Ivanova,A. Belcheva,Antioxidant activity of Aronia melanocarpa fruit juice in vitro,Acta Alimentaria36(2007), 425–428.

[4]. S. Valcheva-Kuzmanova, B. Blagović,S. Valić,Electron spin resonance measurement of radical scavenging activity of Aronia melanocarpa fruit juice. Pharmacognosy Magazine8(2012), 171–174.

[5]. W. Zheng, S. Y. Wang,Oxygen radical absorbing capacity of phenolics in blueberries, cranberries, chokeberries, and lingonberries, Journal of Agricultural and Food Chemistry51 (2003), 502–509.

[6]. M. J. Bermudez-Soto, F. A. Tomas-Barberan, Evaluation of commercial red fruit juice concentrates as ingredients for antioxidant functional juices, Eurpean Food Research and Technology219 (2004), 133–141.

[7]. X. L. Wu, L. W. Gu, R. L. Prior,S. McKay,Characterization of anthocyanins and proanthocyanidins in some cultivars of Ribes, Aronia, and Sambucus and their antioxidant capacity,Journal of Agricultural and Food Chemistry52 (2004), 7846–7856.

Page 168: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

166

[8]. L. Jakobek, M. Seruga, P. Krivak, The infl uence of interactions among phenolic compounds on the antiradical activity of chokeberries ( Aronia melanocarpa),International Journal of Food Science and Nutrition62 (2011), 345–352.

[9]. V. L. Singleton, J. A. Rossi,Colorimetry of total phenolics with phosphomolybdic phosphotungstic acid reagents,American Journal of Enology and Viticulture16(1965), 144–158.

[10]. J. Zhishen, T. Mengcheng,W. Jianming,The determination of fl avonoid contents in mulberry and their scavenging effects on superoxide radicals,Food Chemistry64(1999), 555–559.

[11]. M. M. Guisti, L. E. Rodrigues-Saona,R. E. Wrolstad,Spectral characteristics, molar absorptivity and color of pelargonidin derivatives,Journal of Agricultural and Food Chemistry47 (1999), 4631–4637.

[12]. M. G. L. Hertog, P. C. H. Hollman,D. P. Vanema,Optimization of a quantitative HPLC determination of potentially anticarcinogenic fl avonoids in vegetables and fruits,Journal of Agricultural and Food Chemistry40 (1992), 1591–1598.

[13]. H. Ohkawa, Ν. Ohishi, Κ. Yagi, Assay for lipid peroxides in animal tissues by thiobarbituric acidreaction,Analytica Biochemistry,95 (1979), 351–358.

[14]. S. Valcheva-Kuzmanova, P. Borisova, B. Galunska, I. Krasnaliev, A. Belcheva, Hepatoprotective effect of the natural fruit juice from Aronia melanocarpa on carbon tetrachloride-induced acute liver damage in rats, Experimental and Toxicologic Pathology56 (2004), 195–201.

[15]. H. Jaeschke, C. D. Williams, M. R. McGill, Y. Xie, A. Ramachandran, Models of drug-induced liver injury for evaluation of phytotherapeutics and other natural products,Food and Chemical Toxicology55 (2013), 279–289.

[16]. H. Nakagawa, S. Maeda, Y. Hikiba, T. Ohmae, W. Shibata, A. Yanai, K.Sakamoto, K. Ogura, T. Noguchi, M. Karin, H. Ichijo, M. Omata, Deletion ofapoptosis signal-regulating kinase 1 attenuates acetaminophen-induced liverinjury by inhibiting c-Jun N-terminal kinase activation, Gastroenterology135 (2008),1311–1321.

[17]. M.L. Bajt, A. Farhood, J.J. Lemasters, H. Jaeschke, Mitochondrial baxtranslocation accelerates DNA fragmentation and cell necrosis in a murinemodel of acetaminophen hepatotoxicity,Journal of Pharmacololgy and Experimental Therapeutics324 (2008), 8–14.

[18]. S. S. Kalsi, D. M. Wood, W. S. Waring, P. I.Dargan, Does cytochrome P450 liver isoenzyme induction increase the risk of liver toxicity after paracetamol overdose?,Open Access Emergency Medicine3(2011), 69–76.

[19]. T. Castillo, D. R. Koop, S. Kamimura, G. Triadafi lopoulos, H. Tsukamoto, Role of cytochrome P-450 2E1 in ethanol-, carbon tetrachloride- and iron-dependent microsomal lipid peroxidation,Hepatology16 (1992), 992–996.

[20]. S. S. Lee, J. T. Buters, T. Pineau, P. Fernandez-Salguero, F. J. Gonzalez, Role ofCYP2E1 in the hepatotoxicity of acetaminophen,Journal of Biological Chemistry271 (1996), 12063–12067.

[21]. R. M. Shayiq, D. W. Roberts, K. Rothstein, J. E. Snawder, W. Benson, X. Ma, M. Black, Repeat exposure to incremental doses of acetaminophen provides protection against acetaminophen-induced lethality in mice: An explanation for high acetaminophen dosage in humans without hepatic injury, Hepatology 29 (1999), 451–463.

[22]. V. Krajka-Kuźniak, H. Szaefer, E. Ignatowicz, T. Adamska, J. Oszmiański, W. Baer-Dubowska, Effect of Chokeberry ( Aronia melanocarpa) juice on the metabolic activation and detoxication of carcinogenic N-nitrosodiethylamine in rat liver,Journal of Agricultural and Food Chemistry57 (2009), 5071–5077.

Page 169: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

167

Chapter 20

Exogenic Intoxication with Neuroleptix and Antipsychotic Medications –

Clinical Case

Severina DAKOVA a, Nikola RAMSHEV a, Zorka RAMSHEVAb,Konstantin RAMSHEV a, Kamen KANEV c

a Clinic of Intensive TherapyMilitary; b Department of Clinical Laboratory,c Department Disaster Medicine and Toxicology

Military Medical Academy, Sofi a, Bulgaria

Abstract –We present a case of suicidal attempt of a woman who had taken toxic doses of medications: Rivotril 0,5 mg-200pills, Gerodorm 40mg-20 tablets, Hedonin 100mg-120 pills, Seroxat 10 tablets ,Atenolol 50mg-15 pills. She was found 6-8 hours after the incidence and taken to the hospital in comma- with pulmonary, heart and renal insuffi ciency. After gastric lavement,was undertaken artifi cial pulmonary ventilation, antidotic therapy and hemodialisis procedures. The patient was given antioedemic, gastro- and hepatoprotective and symptomatic therapy.Stimulation treatment with cathecholamines was stopped, because of the paradoxical effect upon the blood pressure. The woman was discharged from the hospital stabilized haemodinamically, in good condition and given advice to visit her psychiatrist.

Approximately 11% of the drugs associated with surveyed emergency room drug abuse problems were benzodiazepines. Seven of the top 50 drugs of abuse seen in emergency rooms were benzodiazepines. In nearly 70% of surveyed emergency room (ER) visits involving benzodiazepines, use was in conjunction with at least one other drug. In more than half of the ER drug episodes involving benzodiazepines, the patient had attempted suicide. Because of the pharmacology of benzodiazepines, only a small percentage of those suicide attempts caused death unless the benzodiazepines were used in combination with other drugs having more lethal pharmacological properties.[1,2]

Oral benzodiazepine (BZD) overdoses, without co-ingestions, rarely result in signifi cant morbidity (eg, aspiration pneumonia, rhabdomyolysis) or mortality. In mixed overdoses, they can potentiate the effect of alcohol or other sedative-hypnotics. Acute intravenous administration of BZDs is associated with greater degrees of respiratory depression.[3]

We present a case of suicidal attempt of a woman who had taken toxic doses of medications: Rivotril 0.5 mg-200pills, Gerodorm 40mg-20 tablets, Hedonin 100mg-120 pills, Seroxat 10 tablets ,Atenolol 50mg-15 pills.

Page 170: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

168

She was found 6-8 hours after the incidence and taken to the hospital in comma- with pulmonary, heart and renal insuffi ciency.

Toxochemical analysis was done with the following results-Urine / +/ for BZD and TCA Blood tests -Clonazepam /Rivotril/ -0.2μg/mlQuetiapine /Seroquel , Hedonim/-1. 2μg/mlCinolazepam /Geridorm/ -2.5μg/mlParoxetine /Seroxat/-0.06μg/mlAtenolol -0.7μg/mlAetanoli, metanoli and Amitriptilini were not found.According to literature and data from the Toxicology laboratory in MMA Sofi a Bulgaria

[4, 5, 6] the therapeutic and toxical dosis of these medications are

Medication

Therapeutic range μg/ml

Toxic rangeμg/ml

Clonazepam /Rivotril/ 0.01-0.08 >0.1

Quetiapine / Hedonim/ 0.075-0.5 /0.9/ >1.0

Cinolazepam /Geridorm/ >0.2

Paroxetine /Seroxat/ 0.07-0.15 >0.3

Atenolol 0.1-1.0 >2.0

From the rest haematology and biochemistry results- Hb-153/101 g/l, Leuc-9.7/19.1ASAT- 163/75 U/l; ALAT-102/195 U/l;GGTP-226 U/l; LDH-587/644 U/l; Amyl/S/-

1617/253 U/l; KK-8143/609 U/l; KK-MB-340/16 U/l. CRP-6.0/182. Myogl.-800/1000 U/l.

Fibrg-8.2/4.6g/l; D-Dimer-19.44/2.06. Ro-gr. Pulmo-oedema pulmonum; pleuritis exudativa dextra; CT –head-small ischemic zones intacerebrally in the right hemisphereAfter gastric lavement, was undertaken artifi cial pulmonary ventilation, antidotic

therapy and hemodialisis procedures. The patient was given antioedemic, gastro- and hepatoprotective and symptomatic therapy. Stimulation treatment with cathecholamines was stopped, because of the paradoxical effect upon the blood pressure. The woman was discharged from the hospital stabilized haemodynamically, in good condition and given advice to visit her psychiatrist.

Page 171: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

169

Reference:

[1]. Bronstein AC, Spyker DA, Cantilena LR Jr, Rumack BH, Dart RC. 2011 Annual report of the American Association of Poison Control Centers’ National Poison Data System (NPDS): 29th Annual Report. Clin Toxicol (Phila). Dec 2012;50(10):911-1164. [Medline].

[2]. The Flumazenil in Benzodiazepine Intoxication Multicenter Study Group. Treatment of benzodiazepine overdose with fl umazenil. Clin Ther. Nov-Dec 1992;14(6):978-95. [Medline].

[3]. Bronstein AC, Spyker DA, Cantilena LR Jr, Green JL, Rumack BH, Giffi n SL. 2009 Annual Report of the American Association of Poison Control Centers’ National Poison Data System (NPDS): 27th Annual Report. Clin Toxicol (Phila). Dec 2010;48(10):979-1178. [Medline].

[4]. Schulz, M., Schmuldt. Therapeutic and toxic blood concentrations of more than 800 drugs and xenobiotics, Pharmazie, 58(7), 2003, 447-474.

[5]. TJAFT Database.[6]. Authors database.

Page 172: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

170

Chapter 21

Toxic Pneumofi brosis – Late Occurrence of Chronic

Cadmium ExpositionDiana APOSTOLOVA1, Tanya KUNEVA, Vera PETKOVA,

Daniela MEDZHIDIEVAClinic of Occupational Diseases, Medical University-Sofi a,

Faculty of Medicine, Sofi a, BULGARIA

Abstract. The subject of this examination are 15 people who have been exposed to above-norm concentrations of cadmium in a factory producing accumulators for cars. The duration of the expositions is anywhere between 7 and 25 years. The professionals have been dynamically examined for 10 years in the Department of Occupation Diseases in Sofi a. Hematologic, biochemical, toxicochemical examinations, x-rays of lungs, functional examination of breathing and blood gases. The levels of cadmium in blood and urine before and after antidote therapy with CaNa2 EDTA have also been monitored. The most commonly observed clinical symptoms in relation to chronic cadmium exposition are partial tooth loss, atrophic rhinopharyngitis, hypo/anosmia, chronic bronchitis and high-level antidote-invoked cadmium in urine. A sight of kidney infection – a low-degree proteinuria has been observed in two of the patients. After a prolonged latent period and expressed, persistent cadmium absorption, a diffused interstitial fi brosis is diagnosed as a late expression of the cumulative effects of cadmium.

Key words. cadmium, pneumofi brosis, bronchitis, proteinuria

IntroductionToxic effects of cadmium on the respiratory system as a critical organ and manifestation

of intoxication have been analysed in many scientifi c reports [1, 2, 3, 4, 5]. Chemical pneumonitis development is a common cause of lethal outcome in case of acute poisoning with the metal [6,7].

After acute intoxication, as well as after years of exposure to cadmium, symptoms of damages to the respiratory system, including pneumofi brosis, pulmonary emphysema, chronic rhinopharingitis, hypo/anosmia, are observed [1, 8].

Data from large epidemiological studies have demonstrated the relation between lung cancer and exposure to cadmium. Cadmium exposure is an important and independent risk

1 Corresponding author: Diana Apostolova, Acad. Ivan Geshov blvd. 15, 1431 Sofi a, Bulgaria; E-mail: [email protected]

Page 173: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

171

factor for mortality from cancer in adults [9, 10, 11, 12, 13]. Recent studies have been focusing attention on other tumor sites, such as kidneys, mammary gland and prostate [14, 15].

This work presents the results of many years of clinical studies of a group of retired workers who during their labour activity have been in contact with cadmium, aiming at fi nding late effects on health as a result of past chronic exposure to cadmium.

1. Materials and methodsThe objective of the study are 15 persons (14 women and one man) who have been

exposed to excessive cadmium concentration levels in a plant for nickel- cadmium batteries and have been traced several times within ten years under stationary conditions in the Clinic of Occupational Diseases – Sofi a. The average age of the tested people is 47.5 years (aged 39 to 63).

The risky length of service duration at the power batteries plant has been averaged to 8.7 years, ranging between 3 and 25 years. The time from the risky occupation resignation to the clinical observation averages to 15.3 years (from 7 to 26 years). Four participants reported smoking 3-4 cigarettes daily, that had been quitted for six or seven years, and one of the patients smokes 10-15 cigarettes a day and continues doing so.

A full range of haematological tests (ESR - erythrocyte sedimentation rate and full blood tests with differential count), biochemical tests (glucose, urea, creatinine, uric acid, liver enzymes) and blood gas analysis have been made - performed on the SYSMEX KX21N automatic analyzer.

Indicators/Parameters of total urine (relative weight, chemical indicators and sediment) and the amount of protein in a 24-hour urine have been analysed. Lung front x-ray and functional investigation of respiratory capacities (FIR) have been conducted.

The concentration of cadmium in blood and urine have been tested by an extraction method on the atomic absorption spectrometer (AAS) Perkin-Elmer 3030 (23). Were traced cadmium levels in blood and urine (before and after the СаNa2EDTA antidotal therapy) have been traced.

The tested people’s ENT (otorhinolaryngological) status has been determined and olfactometry implemented.

2. Results and DiscussionThe concentrations of cadmium in blood and in urine (before and after decorporation

by СаNa2EDTA antidote) for each tested person are presented in Table 1.

Table 1. Values of cadmium concentrations in blood and urine (before and after СаNa2EDTA antidote) for each tested person.

Person No.

Risky length of service (years)

Time after resigning from

work(years)

Number of tests (No.)

Blood Cadmium (μmol/L)

Urinary Cadmium–

basal (μmol/L)

Urinary Cadmium– after EDTA

(μmol/L)1. 25

(smoker)7 4 0.102

(0.054÷0.13)0.204

(0.109÷0.3)0.364

(0.259÷0.57)2. 8 14 2 0.055

(0.038÷0.072)0.0675

-0.14

-

Page 174: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

172

3. 16 11 2 0.016(0.015÷0.018)

0.019-

0.067 -

4. 6 18 3 0.023(0.012÷0.03)

0.079-

0.111(0.082÷0.14)

5. 14 14 2 0.058(0.048÷0.069)

0.075-

0.126(0.082÷0.17)

6. 15 7 2 0. 067-

0.11-

0.275(0.19÷0.36)

7. 8 13 3 0.055(0.03÷0.084)

0.021-

0.078(0.045÷0.11)

8. 4 26 2 0.016(0.009÷0.022)

0.0225-

0.0075-

9. 3 17 2 0.0245(0.019÷0.03)

0.05-

0.0325 (0.015÷0.05)

10. 3,5 18 2 0.045-

0.037-

0.0695(0.049÷0.09)

11. 3 20 2 0.064(0.044÷0.084)

0.0625(0.026÷0.099)

0.24 (0.11÷0.37)

12. 3 11 2 0.067(0.048÷0.086)

0.0895(0.052÷0.127)

0.083(0.08÷0.111)

13. 5,6 15 1 0.023 0.11 0.01914. 13 18 2 0.059

-0.072

(0.049÷0.094)0.32

(0.101÷0.54)15. 3 20 1 0.059 0.049 0.54

Reference values of urinary cadmium in unexposed people are 0.01 μmol/L, and of blood cadmium for non-smokers they are 0.004–0.009 μmol/L. Data from the biomonitoring studies on cadmium concentrations/levels in blood demonstrate higher values in smokers than in non-smokers [16] . The table includes only one smoker, for whom reference values for smokers are valid, which for blood are 0.013–0.04 μmol/L. With the observed three persons with a history of smoking, we have used the reference values for non-smokers because of the negligible and long discontinued use. As it follows from Table 1, in some of the investigated people (persons No. 1, 2, 4, 5, 6 and 13), signifi cantly higher cadmium levels than the reference ones are also observed after decorporation with EDTA, which is mainly determined by the risk length of service duration. Cadmium elimination after having spent exposure to cadmium is a very slow process due to its accumulation in the body while its biological half-life ranges from 7 to 30 years. In case of short 3-year length of service (participants No. 9, 11 and 15) elevated levels of cadmium in the samples tested have been also observed, which we associate with high-level exposure and long-term retention of the metal in the body. Studies show high correlation between the concentration of cadmium in the working environment, duration of exposure, tissue concentration and exhibited clinical syndromes [17]. The results of the diagnosed pathology in the studied contingency are presented in Table 2.

Table 2. Diagnosed pathology in each tested/studied person.

No. Respiratory system ENT status Other pathology

1. Diffuse interstitial pneumofi brosis. COPD. RD І deg. Obstructive VD.

Chronic rhinopharyngitis. Hyposmia of combined, predominantly peripheral, type.

Spondylarthrosis. Bilateral coxarthrosis and gonarthrosis.

Page 175: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

173

2. Diffuse interstitial pneumofi brosis. COPD. RD І deg. Obstructive VD.

Chronic rhinopharyngitis. Hyposmia of peripheral type.

Kidney stones disease. Chr. gastroduodenitis. Cervicoarthrosis

3. Diffuse interstitial pneumofi brosis. COPD. Obstructive VD.

Chronic rhinitis.Hyposmia of peripheral type.

Left-sided gonarthrosis. Bilateral cervical radiculopathy.

4. Bilateral, mainly basal, pneumofi brosis. COPD. RD І-ІІ deg. Obstructive VD.

Chronic rhino-pharyngo-laryngitis.

Arterial hypertension. Duodenal ulcer. Chr. gastritis. GERD.

5. Diffuse interstitial pneumofi brosis. Pulmonary emphysema. RD І deg. Obstructive VD.

Chronic rhino-pharyngo-laryngitis. Hyposmia of peripheral type.

Arterial hypertension.Vertigo of central type.

6. Diffuse interstitial pneumofi brosis. Pulmonary emphysema. RD І-ІІ deg. Obstructive VD.

Chronic rhino-pharyngo-laryngitis. Hyposmia of peripheral type.

Arterial hypertension.Neurotic disorder.

7. Diffuse interstitial pneumofi brosis. COPD. RD І-ІІ deg. Obstructive VD.

Chronic rhinosinusitis. Arterial hypertension.Diabetes mellitus. Kidney stones disease. Coxarthrosis.

8. Bilateral, mainly basal, pneumofi brosis. COPD - bronchitic form. Compoun VD

Chronic rhinopharyngitis. Chronic gastroduodenitis. Spondylarthrosis.

9. Diffuse interstitial pneumofi brosis. COPD. Obstructive VD.

Chronic rhino-pharyngo-laryngitis.

Arterial hypertension.

10. Diffuse interstitial pneumofi brosis. COPD. RD І-ІІ deg. Compound VD.

Chronic rhino-pharyngo-laryngitis.

Arterial hypertension.CAD. Kidney stones disease. Duodenal ulcer.

11. Diffuse interstitial pneumofi brosis. COPD. Obstructive VD.

Chronic rhino-pharyngo-laryngitis.Hyposmia of peripheral type.

Chr. gastroduodenitis. Vestibulopathy. Hypodepressive syndrome. Generalized osteoarthrosis.

12. Diffuse interstitial pneumofi brosis. Pulmonary emphysema. CRD І deg. Obstructiv VD.

Chronic rhinopharyngitis. Hyposmia of peripheral type.

Neurosis with hypodepressive elements. Lumbo-sacral radiculopathy.

13. Bilateral basal pneumofi brosis. COPD. CRD І deg. CompoundVD.

Chronic rhinopharyngitis. Hyposmia of peripheral type.

Arterial hypertension.CAD. Diabetes mellitus. Chronic pyelonephritis.

14. Bilateral pneumofi brosis. Chronic bronchitis. CRD І deg. Obstructive VD.

Chronic rhinopharyngitis. Hyposmia of peripheral type.

Arterial hypertension.CAD. Chr. gastroduodenitis. Chr. colitis. Neurasthenic neurosis.

15. Bilateral, mainly basal, pneumofi brosis. COPD. CRD І deg. Obstructive VD.

Deviatio septi nazi. Chr. gastroduodenitis. Duodenal ulcer.

Key: COPD = chronic obstructive pulmonary disease; VD = ventilatory disturbance; RD = respiratory disorders; CRD = chronic respiratory disorders; CAD = coronary artery disease; GERD = gastroesophageal refl ux disease.

The data in Table 2 show in all observed individuals expressed pathology of the respiratory and otorhinolaryngological systems, nosologically exhibited with bilateral pneumofi brosis, chronic bronchitis and pulmonary emphysema, chronic rhinopharyngitis and hyposmia of peripheral type.

We associate these results with a history of high-level cadmium exposure leading to increased accumulation of the metal in the body of workers and the cumulative effect of

Page 176: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

174

cadmium in the development of these late toxicities, despite the already terminated risky activity. Such statements can be found in the studies of Swaddiwudhipong W. et al [18] who after 5-year follow-up of people with long and excessive cadmium exposure fi nd progressive toxic health effects even after its termination.

Inhalation of cadmium is associated in the literature with lung function decline, pneumofi brosis development, chronic obstructive pulmonary disease , and lung cancer [19, 20 ]. In our study, all participants have developed bilateral interstitial pneumofi brosis with underlying chronic bronchitis and pulmonary emphysema , while 73 % of these people have exhibited chronic respiratory failure/insuffi ciency mainly in the early fi rst stage of display. In the radiographic changes of lungs are observed bilaterally, mainly streak-grid pattern with small grainy inclusions, predominantly in the lung bases and parahilar, with the presence of peribronchial seals and enlarged hilar shadows. The results of the parameters of the conducted functional respiratory capacity tests evidence a predominance of obstructive ventilatory syndrome with obstruction of the minor respiratory tracts in 80% of the followed-up people.

Most common changes, we have observed in the oral cavity and upper respiratory tract of the workers after chronic cadmium exposure, are partial edentulism (in 80 % of cases), hyposmia of peripheral type (in 53 % of cases) and atrophic rhinitis, laryngitis and rhinopharingitis in 93 % of cases.

Toxicologic studies show that metallothionein is a protein carrying cadmium to the kidneys, and after prolonged cadmium exposure, even at low exposure levels, higher impairment of renal function can be observed [21].

In our study, in only two of the followe-up workers was found low-grade proteinuria as a manifestation of renal impairment, which we associate with the recovery of renal function after cessation of cadmium exposure. A similar result was also observed in studies of Liang Y. et al [22].

Epidemiological studies of Li Q. et al in Japan show correlation between the amount of cadmium in the body, the mortality from cardiovascular and cerebrovascular diseases, nephritis and nephrosis [23]. Similar studies of Tellez-Plaza M. et al in the United States found a relation between cadmium values in blood and in urine, and mortality from cardiovascular diseases in adult population [24]. Unconvincing relation between cadmium exposure and hypertension, diabetes and kidney stone disease development is found by Swaddiwudhipong W. et al [25].

In our study, we found hypertension in almost half of the observed persons (53.3 %), nephrolithiasis at a very low rate (20 %) and diabetes mellitus (13 %). In one third of the followed-up people we fi nd pathology of the digestive system , mainly manifested by chronic gastritis gastroduodenites and duodenal ulcers and in 40 % of participants – degenerative injuries of the musculoskeletal system. Due to the small number of participants, we cannot draw any fi rm conclusions about these diseases and cadmium exposure experienced.

3. ConclusionAfter a long latency period against the background of expressed persistent cadmium

absorption and excretion are diagnosed as a late manifestation of the cumulative effect of cadmium, diffuse interstitial fi brosis with concomitant chronic bronchitis and emphysema,

Page 177: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

175

which requires dynamic monitoring of people who have worked under the conditions of cadmium exposure in the years after the latter have left work .

References

[1]. S. Moitra,PD. Blanc,S. Sahu, Adverse respiratory effects associated with cadmium exposure in small-scale jewellery workshops in India,Thorax68 (6) (2013), 565-570.

[2]. KE. Driscoll, JK.Maurer, J. Poynter, J. Higgins, T. Asquith, NS. Miller, Stimulation of rat alveolar macrophage fi bronectin release in a cadmium chloride model of lung injury and fi brosis, Toxicology & Applied Pharmacology116 (1) (1992), 30-37.

[3]. FR. Frankel, JR. Steeger, VV. Damiano, M. Sohn, D. Oppenheim, G. Weinbaum, Induction of unilateral pulmonary fi brosis in the rat by cadmium chloride, American Journal of Respiratory Cell & Molecular Biology 5 (4) (1991), 385-394.

[4]. VV. Damiano, PV. Cherian, FR. Frankel, JR. Steeger, M. Sohn, D. Oppenheim, G. Weinbaum, Intraluminal fi brosis induced unilaterally by lobar instillation of CdCl2 into the rat lung, American Journal of Pathology, 137 (4) (1990), 883-894.

[5]. K. Zajusz, K. Marek, A. Kujawska, M. Zylka-Wloszczyk, B. Romaniec,F. Sonecka,A. Stachura,Effect of metal dust on the respiratory system. I. Experimental studies, Medycyna Pracy, 30 (1) (1979), 15-20.

[6]. K. Seidal, N. Jorgensen, CG. Elinder, B. Sjogren, M. Vahter, Fatal cadmium-induced pneumonitis, Scandinavian Journal of Work, Environment & Health, 19 (6) (1993), 429-431.

[7]. K. Yamamoto, M. Ueda, H. Kikuchi, H. Hattori, Y. Hiraoka, An acute fatal occupational cadmium poisoning by inhalation, Zeitschrift fur Rechtsmedizin - Journal of Legal Medicine91 (2) (1983),139-143.

[8]. GL. Snider, EC. Lucey, B. Faris, Y. Jung-Legg, PJ. Stone, C. Franzblau, Cadmium-chloride-induced air-space enlargement with interstitial pulmonary fi brosis is not associated with destruction of lung elastin. Implications for the pathogenesis of human emphysema, American Review of Respiratory Disease, 137 (4) (1988), 918-923.

[9]. YO. Son Wang, L. P. Poyil, A. Budhraja, JA. Hitron, Z. Zhang, JC. Lee, X. Shi, Cadmium induces carcinogenesis in BEAS-2B cells through ROS-dependent activation of PI3K/AKT/GSK-3/-catenin signaling, Toxicology & Applied Pharmacology, 264 (2) (2012),153-160.

[10]. RM. Park, LT. Stayner, MR. Petersen, M. Finley-Couch, R. Hornung, C. Rice, Cadmium and lung cancer mortality accounting for simultaneous arsenic exposure, Occupational & Environmental Medicine, 69 (5) (2012), 303-309.

[11]. A. Hartwig, Cadmium and cancer, Metal Ions in Life Sciences, 11 (2013), 491-507. [12]. B. Wang, Y. Li, C. Shao, Y. Tan, L. Cai, Cadmium and its epigenetic effects, Current Medicinal

Chemistry,19 (16) (2012), 2611-2620. [13]. RH. Verhoeven, MW. Louwman, F. Buntinx, AM. Botterweck, D. Lousbergh, C. Faes, JW. Coebergh,

Variation in cancer incidence in northeastern Belgium and southeastern Netherlands seems unrelated to cadmium emission of zinc smelters, European Journal of Cancer Prevention, 20 (6) (2011), 549-555.

[14]. NB. Aquino, MB. Sevigny, J. Sabangan, MC. Louie, The role of cadmium and nickel in estrogen receptor signaling and breast cancer: metalloestrogens or not?, Journal of Environmental Science & Health Part C Environmental Carcinogenesis & Ecotoxicology Reviews,30 (3) (2012),189-224.

[15]. YS. Lin, JL. Caffrey, JW. Lin, D. Bayliss, MF. Faramawi, TF. Bateson, B. Sonawane, Increased risk of cancer mortality associated with cadmium exposures in older Americans with low zinc intake, Journal of Toxicology & Environmental Health, Part A. 76 (1) (2013), 1-15.

[16]. KM. Marano, ZS. Naufal, SJ. Kathman, JA. Bodnar, MF. Borgerding, CD. Garner, CL. Wilson, Cadmium exposure and tobacco consumption: Biomarkers and risk assessment, Regulatory Toxicology & Pharmacology, 64 (2) (2012), 243-252.

[17]. I. Krzywy, E. Krzywy, J. Peregud-Pogorzelski, K. Luksza, A. Brodkiewicz, Cadmium--is there something to fear?, Annales Academiae Medicae Stetinensis, 57 (3) (2011), 49-63.

[18]. W. Swaddiwudhipong, P. Limpatanachote, P. Mahasakpan, S. Krintratun, B. Punta, T. Funkhiew, Progress in cadmium-related health effects in persons with high environmental exposure in northwestern Thailand: a fi ve-year follow-up, Environmental Research, 112 (2012), 194-198.

[19]. J. Rennolds, S. Butler, K. Maloney, PN. Boyaka, IC. Davis, DL. Knoell, NL. Parinandi, E. Cormet-Boyaka, Cadmium regulates the expression of the CFTR chloride channel in human airway epithelial cells, Toxicological Sciences, 116 (1) (2010), 349-358l.

Page 178: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

176

[20]. TJ. Smith, TL. Petty, JC. Reading, S. Lakshminarayan, Pulmonary effects of chronic exposure to airborne cadmium, American Review of Respiratory Disease, 114 (1) (1976), 161-169.

[21]. G. Nordberg, T. Jin, X. Wu, J. Lu, L. Chen, Y. Liang, L. Lei, F. Hong, IA. Bergdahl, M. Nordberg, Kidney dysfunction and cadmium exposure-- factors infl uencing dose-response relationships, Journal of Trace Elements in Medicine & Biology, 26 (2-3) (2012), 197-200.

[22]. Q. Li, M. Nishijo, H. Nakagawa, Y. Morikawa, M. Sakurai, K. Nakamura, T. Kido, K. Nogawa, M. Dai, Relationship between urinary cadmium and mortality in habitants of a cadmium-polluted area: a 22-year follow-up study in Japan, Chinese Medical Journal,124(21)(2011), 3504-3509.

[23]. M. Tellez-Plaza, A. Navas-Acien, A. Menke, CM. Crainiceanu, R. Pastor-Barriuso, E. Guallar, Cadmium exposure and all-cause and cardiovascular mortality in the U.S. general population, Environmental Health Perspectives, 120 (7) (2012),1017-1022.

[24]. W. Swaddiwudhipong, P. Limpatanachote, M. Nishijo, R. Honda, P. Mahasakpan, S. Krintratun, Cadmium-exposed population in Mae Sot district, Tak province: 3. Associations between urinary cadmium and renal dysfunction, hypertension, diabetes, and urinary stones, Journal of the Medical Association of Thailand, 93 (2) (2010), 231-238.

Page 179: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

177

Chapter 22

Sick Building Syndrome – Proper Management for Protect

Human Health

Julia RADENKOVA - SAEVA, Eva SAEVA*,Toxicology Clinic, “Pirogov” University Hospital, Sofi a, Bulgaria

University “La Sapienza”, Rome, Italy*

Abstract: The term “ sick building syndrome” (SBS) is used to describe situations in which building occupants experience acute adversehealth and discomfort effects, that appear to be linked to time spent in a building, but no specifi c illness or cause can be identifi ed.SBS complaints seem to be the result of the interaction of various environmental, occupational, and psychological factors. Symptoms caused by these factors can cause either short-term or long-term adverse health effects that do not always resolve when the occupant leaves the building. The feeling of poor health increases sickness absenteeism and causes a decrease in productivity of the work. The cause, signs and symptoms, management and prevention of this illness have been discussed in this review.

Key words: sick building syndrome, health effects

Introduction“Sick building syndrome” (SBS) is term, used to describe situations in which building

occupants experience acute adverse health and discomfort effects, that appear to be linked to time spent in a building, but no specifi c illness or cause can be identifi ed [4, 10, 30].

This term or another one “building-related illness” (BRI) is used to defi ne illnesses related to non-industrial buildings, mainly modern offi ces, in which people spend many working hours [23].

The term “Sick Building Syndrome” was coined by WHO in 1986, when they also estimated that 10-30% of newly built offi ce buildings in the West had indoor air problems. Early Danish and British studies reported symptoms[14, 18, 27].

In July 1968, an explosive epidemic of illness characterized principally by fever, headaches and muscular pains affected at least 144 people, including about 100 employees of a health department building in Pontiac Michigan, USA. A defective air conditioning system was implicated as the source and mechanism that spread the sickness, but extensive laboratory and environmental investigations failed to identify the origin of the disease, so

Page 180: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

178

it was simply labeled “Pontiac Fever”. Many years later it was learned that this illness was caused by a bacterium identifi ed as Legionella pneumophila. This name was fi rst attributed to the causative organisms of the 1976 outbreak of a disease that struck 182 people attending an American Legion Convention in an air conditioned hotel in Philadelphia, Pennsylvania, USA. Thirty-four people died from the illness. In Australia’s largest epidemic in Wollongong in 1987, forty-four cases required hospitalization and fourteen victims died[13, 33, 35].

SBS symptoms include headache, dizziness, nausea, eye, nose or throat irritation, dry cough, dry or itching skin, diffi culty in concentration, fatigue, sensitivity to odours, hoarseness of voice, allergies, cold, fl u-like symptoms, increased incidence of asthma attacks and personality changes [10, 6, 7, 12, 22, 24, 25].

If these symptoms subside once the individual leaves the building, then it is a strong indicator that he or she is suffering from SBS. However, if a specifi c chemical or biological contaminant is found to be the cause of discomfort, then the diagnosis may be augmented to building-related illness [22].

The aim of this article is to summarize current understanding of “ sick building syndrome” - the cause, signs and symptoms, management and prevention of this illness and to stress the importance of thiscontemporary phenomenon.

EtiologyThe following factors might be primarily responsible for SBS: chemical contaminants,

biological contaminants, inadequate ventilation, electromagnetic radiation, psychological factors, poor and inappropriate lighting with absence of sunlight, bad acoustics, poor ergonomics and humidity.

1. Chemical contaminants.1.1. From outdoor sources: Contaminants from outside like pollutants from motor

vehicle exhaust, plumbing vents and building exhausts (bathrooms and kitchens) can enter the building through poorly located air intake vents, windows and other openings. Combustion byproducts can enter a building from a nearby garage. Radon, formaldehyde, asbestos, dust and lead paint can enter through poorly located air intake vents and other openings [16, 20, 25, 27, 34].

1.2. From indoor sources: The most common contaminant of indoor air includes the volatile organic compounds (VOC). The main sources of VOC are adhesives, upholstery, carpeting, copy machines, manufactured wood products, pesticides, cleaning agents, etc. Environmental tobacco smoke, respirable particulate matter, combustion byproducts from stove, fi replace and unvented space heater also increase the chemical contamination. Synthetic fragrances in personal care products or in cleaning and maintenance products also contribute to the contamination [1, 10, 21, 26].

2. Biological contaminants. The biological contaminants include pollen, bacteria, viruses, fungus, molds, etc. These contaminants can breed in stagnant water that has accumulated in humidifi ers, drainpipes and ducts or where water has collected on ceiling tiles, insulation, carpets and upholstery[1, 3, 5, 6, 8, 15, 19, 32].

Insect and bird droppings can also be a source of biological contamination. Biological

Page 181: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

179

contamination causes fever, chills, cough, chest tightness, muscle aches and allergic reactions. In offi ces with a high density of occupancy, airborne diseases can spread rapidly from one worker to another. Air-conditioning systems can recirculate pathogens and spread them throughout the building e.g., Legionnaire’s disease is due to contamination of cooling towers by legionella organisms. Legionella is also responsible for Pontiac fever. This is caused by inhaling the antigen of the gram negative bacilli Legionellae. Symptoms of Pontiac Fever are a rapidly rising fever alternating with chills. The infected person usually has anorexia, abdominal pain, malaise, myalgia, headaches, a non productive cough and diarrhoea is common. Pontiac Fever usually affects healthy young people and they recover spontaneously in 2–3 days with no treatment Legionnaire’s Disease is a more severe form of Pontiac Fever and is caused by the same micro organism. The gram negative bacilli Legionellae (of which there are 35 species and at least 45 serogroups) lives in hot water systems, air conditioning cooling towers, evaporative air conditioners, hot and cold water taps, showers and anything in the building that has running water as the transmission of this micro organism from the water to humans is via the air that the person breaths. Legionnaire’s Disease occurs more commonly in people who are over 50 years old. As well as causing a severe form of pneumonia with Legionnaire’s Disease there may also be brain, bowel and liver damage and kidney failure [13, 28, 33, 35].

Humidifi er fever is caused by breathing in water droplets from humidifi ers heavily contaminated with microorganisms causing respiratory infections, asthma and extrinsic allergic alveolitis. The disease is noninfective in nature. The patient may have fl u-like symptoms. It is sometimes called Monday Fever. Permanent lung damage does not occur.

3. Inadequate ventilation.In 1970, oil embargo led building designers to make buildings more airtight, with less outdoor air ventilation, in order to improve energy effi ciency. The ventilation was reduced to 5 cfm/person. This reduced ventilation rate was found to be inadequate to maintain the health and comfort of building occupants. Malfunctioning heating, ventilation and air-conditioning systems (HVAC systems) also increase the indoor air pollution. Poor design and construction of buildings with more number of offi ces cramped in a building to increase the salable area also contribute to inadequate ventilation [4, 11, 29, 31].

4. Electromagnetic radiation. Gadgets like microwaves, televisions and computers emit electromagnetic radiation, which ionizes the air. Extensive wiring without proper grounding also creates high magnetic fi elds, which have been linked to cancer [9, 20].

5. Psychological factors. Excessive work stress or dissatisfaction, poor interpersonal relationships and poor communication are often seen to be associated with SBS [6, 21, 23].

6. Poor and inappropriate lighting with absence of sunlight, bad acoustics, poor ergonomics and humidity may also contribute to SBS.The symptoms of SBS are commonly seen in people with clerical jobs than in people with managerial jobs because professionals or managers have better working conditions. The symptoms are more common in females than in males probably because more females are in secretarial jobs, they are more aware of their health or a lesser dose of pollutants is required to manifest the effects. The symptoms are more common in air-conditioned buildings than in naturally ventilated buildings and are more common in a public sector building than in a private sector building

Page 182: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

180

[2, 12, 14, 20].Environmental Protection Agency (2010), Burge (2004), Edmondson DA, et al. 2005, Evans (2008), Gomzi and Bobic (2009), Greer (2007), Joshi (2008), Kilburn KH. 2003 Kipen (2002), Mendelson et al. (2000), Milica (2009), Shoemaker and House (2005), point out, that Sick Building Syndrome can cause the following ill health effects:

• Respiratory:Runny nose; Sneezing; Dry sore throat; Blocked nose;Nose bleeds;Allergic Rhinitis (repetitive sneezing and a runny nose);Sinus congestion;Colds;Infl uenza like symptoms;Dry Cough;Throat irritation;Wheezing when breathing;Shortness of breath;Sensation of having dry mucus membranes;Hoarseness of the voice due to infl ammation of the throat and larynx;Sensitivity to odours;Increased incidences of building related asthma attacks;

• Eye irritation: Eye dryness;Itching of the eyes;Watering of the eyes;Gritty eyes;Burning of the eyes;Visual disturbances;Light sensitivity;

• Dermal irritation: Skin rashes;Itchy skin;Dry skin;Erythema (Redness or infl ammation due to congestion in, and dilation of, thesuperfi cial capillaries of the skin.);Irritation and dryness of the lips;Seborrheic dermatitis;Periorbital eczema;Rosacca;Uritcaria;Itching folliculitis;

• Cognitive complaints: Functional headache that affect a person’s performance, but which fail toreveal evidence of physiological or structural abnormalities;Migraine headache;Tension headache;Sinus headache due to swelling of the mucus membranes;Mental confusion;

• Lethargy: Lethargic (The word “lethargy” comes from the Greek word lethargos which

• means forgetful.); Diffi culty in concentrating; Mental fatigue; General fatigue that starts within a few hours of coming to work and which ceases after the person leaves the building; Unable to think clearly; Drowsy;

• Gastrointestinal symptoms:nausea;• Other: Dizziness;Unspecifi ed hypersensitivity reactions;Personality changes (that

may be due to stress or ill health);Exacerbation of pre-existing illnesses such as asthma, sinusitis or eczema.

Preventing and curing SBSWHO has set guidelines for proper management of building ventilation systems

to minimize introduction of contaminants and prevent SBS in buildingoccupants. Nine statements and comments were established at a WHO Working Group Meeting in Bilthoven, the Netherlands, May15–17, 2000 [25].

1. Under the principle of the human rightto health, everyone has the right to breathe healthy indoor air.

2. Under the principle of respect for autonomy (self-determination), everyone has the right to adequate information about potentially harmful exposures, and to be provided with effective means for controlling at least part of their indoor exposures

3. Under the principle of non-malefi cence(doing no harm), no agent at a concentration that exposes any occupant to an unnecessary health risk should be introduced into indoor air.

Page 183: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

181

4. Under the principle of benefi cence (doing good), all individuals, groups and organizations associated with a building, whether private, public or governmental, bear responsibility to advocate or work for acceptable air quality for the occupants.

5. Under the principle of social justice, the socio-economic status of occupants should have no bearing on their access to healthy indoor air, but health status may determine special needs for some groups.

6. Under the principle of accountability, all relevant organizations should establish explicit criteria for evaluating and assessing building air quality and its impacts on the health of the population and on the environment.

7. Under the precautionary principle, where there is a risk of harmful indoor air exposure, the presence of uncertainty shall not be used as a reason for postponing cost-effective measures to prevent such exposure.

8. Under the “polluter pays principle”, the polluter is accountable for any harm to health and for welfare resulting from unhealthy indoor air exposures. In addition, the polluter is responsible for mitigation and remediation.

9. Under the principle of sustainability, health and environmental concerns cannot be separated, and the provision of healthy indoor air should not compromise global or local ecologic integrity, or the rights of future generations.

There are a number of potential solutions to sick building syndrome. For the best results, a combination of several solutions may be necessary [2, 11, 12, 17, 27, 29]. Common ways to eliminate sick building syndrome include:

• Upgrading ventilation rates so that HVAC systems meet suggested ventilation standards. Installing and maintaining high-performance indoor air fi lters

• Increase air fl ow – where possible, natural air fl ow should be encouraged through opening windows, for example.

• Eliminate harmful substances as asbestos and lead paint• If indoor or biological contaminants are to blame, then taking steps to eliminate or

minimize their prevalence is the preferred solution to SBS. Potential methods to achieve this include removing water-soaked carpet, drywall or ceiling tiles, improving ventilation in areas of high contaminant concentration (storage closets, bathrooms, etc.), implementing indoor smoking bans.

• Regulate humidity levels – an ideal relative humidity in building is 40 to 70%. • Create the right temperature – ideally a building temperature of around 20 to 22

degrees is healthy. • Use correct lighting – make the most of natural light in a building and add in artifi cial

light according to what is need.• Educating building occupants about the hazards, causes and solutions of sick

building syndrome so that they may take individual steps to reduce symptoms.

ConclusionSBS is considered a multifactorial health problem, being at the same time a medical

and psychosocial phenomenon.SBS complaints seem to be the result of the interaction

Page 184: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

182

of environmental, occupational, and psychological factors. Symptoms caused by these factors can cause either short-term or long-term health effects that do not always resolve when the occupant leaves the building.

The major impact of sick building syndrome on employees are often hidden in increased incidences of sick leave and medical claims, lower productivity of employees and in increased employee turnover. Most people in the work force do not complain about their ill health. They just leave the company to fi nd another organization to work for where they can have better health.

The best way to prevent sick building syndrome is proper design of buildings and ventilation systems so that people have plenty of natural light and individual control over heating and ventilation.

Using proper building materials and technologies could effectively reduce health problems caused by SBS.

References

[1]. Bholah R, Subratty A (2002) Indoor biological contaminants and symptoms of sick building syndrome in offi ce buildings in Mauritius. Int J Environ Health Res 12:93–98.

[2]. Bornehag CG, Blomquist G, Gyntelberg F et al2001 Dampness in buildings and health: Nordic interdisciplinaryreview of the scientifi c evidence on associations between exposure to”dampness” in buildings and health effects (NORDDAMP). Indoor Air 11: 72–86.

[3]. Bunger J, Westphal G, Monnich A, Hinnendahl B, Hallier E, Muller M. Cytotoxicity of occupationally and environmentally relevant mycotoxins. Toxicology. 2004 Oct 1; 202(3):199-211. (s)

[4]. Burge, P. S. “Sick Building Syndrome.” Occupational and Environmental Medicine (February 2004): 185–191.

[5]. Chapman JA, Terr AI, Jacobs RL et al 2003 Toxic mold: phantom risk vs. science. AnnAllergy Asthma Immunol 91: 222-232

[6]. Crago BR, Gray MR, Nelson LA, Davis M, Arnold L, Thrasher JD. Psychological, neuropsychological, and electrocortical effects of mixed mold exposure. Arch Environ Health. 2003 Aug; 58(8):452-63. (s)

[7]. Edmondson DA, Nordness ME, Zacharisen MC, Kurup VP, Fink JN. Allergy and “toxic mold syndrome”. Ann Allergy Asthma Immunol. 2005 Feb; 94(2):234-9. (s)

[8]. Effects of toxic exposure to molds and mycotoxins in building-related illnesses. Arch Environ Health. [9]. "Electromagnetic fi elds and public health: Electromagnetic hypersensitivity". Fact sheet No.296. World

Health Organization. December 2005. Retrieved November 17, 2007.[10]. Environmental Protection Agency (2010) Indoor Air Facts No. 4 (revised) Sick building syndrome.[11]. Evans P (2008) Innovative building materials and sick building syndrome: Liabilities of manufacturers

and importers of defective materials. Innovat Technol 10:37–46.[12]. Fang L, Wyon D P, Clausen G, Fanger P O, 2002 Sick building syndrome symptoms and performance in

a fi eld laboratory study at different levels of temperature and humidity. Indoor Air ‘02:Proceedings of the 9th International Conference on Indoor Air Qualityand Climate 3: 466–471.

[13]. Fields BS, Benson RF, Besser RE. Legionella and Legionnaires‘ Disease: 25 Years of Investigation. Clinical Microbiology Reviews 2002; 15 (3): 506–526.

[14]. Gomzi M, Bobic J (2009) Sick building syndrome. Do we live and work in unhealthy environment? Period Biol 111(1):79–84.

[15]. Gray MR, Thrasher JD, Crago R, Madison RA, Arnold L, Campbell AW, Vojdani A. Mixed mold mycotoxicosis: immunological changes in humans following exposure in water-damaged buildings. Arch Environ Health. 2003 Jul; 58(7):410-20. (s)

[16]. Greer C (2007) Something in the air: A critical review of literature on the topic of sick building syndrome. World Saf J 16(1):23–26.

Page 185: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

183

[17]. Henckel, Leslie. “IAQ FYI: Proactive Management Can Help to Clear Buildings of Indoor Air Quality Problems.” Journal of Property Management (July-August 2003): 48–52.

[18]. Joshi S (2008) The sick building syndrome. Indian J Occup Environ Med 12(2):61–64.[19]. Kilburn KH. Indoor mold exposure associated with neurobehavioral and pulmonary impairment: a

preliminary report. Arch Environ Health. 2003 Jul; 58(7):390-8. (s)[20]. Kipen HM, Fiedler N, 2002, Environmental factors in medically unexplained symptoms and related

syndromes: the evidence and the challenge. Environ Health Persp 110 (suppl 4): 597-599.[21]. Mendell MJ, Fisk WJ, Petersen MR et al. 2002, Indoor particles and symptoms among [Mendelson M,

Catano V, Kelloway K (2000) The role of stress and social support in sick building syndrome. Work Stress 14(2):137–155.

[22]. Menzides D, Bourbeau J, 1997, Building-related illnesses. N Engl J Med 337: 1524-31.[23]. Michelle Murphy, Sick Building Syndrome and the Problem of Uncertainty, 2006.[24]. [Milica G (2009) Sick building syndrome. Do we live and work in unhealthy environment? Period Biol

111(1):79–84.[25]. Molhave L, Krzyzanovski M, 2002, The right to healthy indoor air: the status by 2002. Indoor Air 13

(suppl. 6): 50-53).[26]. Passarelli G (2009) Sick building syndrome: An overview to raise awareness. J Build Appraisal 5(1):55–

66.[27]. Pancer K, Stypulkowska-Misiurewicz H: Pontiac fever – non-pneumonic legionellosis. Przegl Epidemiol

2003, 57:607-12 [28]. ReinkainenLM, Jaakkola JJ, 2001, Effects of temperature and humidifi cation in the offi ce environment.

Arch Environ Health56: 365–368.[29]. Sadovsky, Richard. “Assessing Patients with Medically Unknown Symptoms.” American Family

Physician (June 1, 2000): 3455.[30]. Seppaneno A, Fisk WJ, 2004, Summary of human responses to ventilation. Indoor Air 14(s7): 102-118[31]. Shoemaker R, House D (2005) A time-series study of sick building syndrome: chronic, bio-toxin associated

illness from exposure to water damaged buildings. Neurotoxicol Teratol 27:29–46.[32]. Spalekova M. Epidemiology of Legionellosis in Europe. Bratisl Lek Listy 2006; 107-221.[33]. Wargocki P, Wyon DP, Sundell Jet al. 2000 The effects of outdoor air supply rate in an offi ce on perceived

air quality, sick building syndrome (SBS) symptoms and productivity. Indoor Air 10: 222–236.[34]. World Health Organization 2007. Legionella and the prevention of legionellosis. Geneva, Switzerland:

WHO.

Page 186: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

184

Chapter 23

Case of Severe Intoxication and Anaphylactic Reaction from

Multiple Bee Stings

Katerina STEFANOVA, Evgenia BARZASHKADepartment of Clinical Toxicology

UMHAT “Dr Georgi Stranski” – Pleven, Medical University – Pleven, Bulgaria

Abstract: Multiple stings of stinging Hymenoptera kind insects – honey-bees, wasps and hornets, although comparatively rare, may cause severe life-threatening bee venom poisoning with or without allergic reaction.We represent a casuistic case of multiple simultaneous stings by a bee swarm caused a severe anaphylaxis and intoxication with local and total toxic clinical symptomatic. The case is interesting because of the great number of stings led to a severe allergic reaction, as well as the fact that despite the advanced in years patient and concomitant diseases, active reanimation and complex detoxical therapy led to the favourable outcome..

Key words: multiple stings, allergic reaction, anaphylaxis, intoxication

Introduction: Toxic reactions caused by stinging insects of the Hymenoptera order are well known.

Clinically important insects belong to Apoidea superfamily (bees – pic.1), Vespoidea superfamily (wasps – pic. 2 and hornets – pic. 3) and Solenopsis genus (fi re ants – pic. 4).

pic. 1 pic. 2 pic. 3

pic. 4

Page 187: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

185

The composition of the insect venom has been well studied. Some of its components react as allergens (phospholipase A2, hyaluronidase, melitine), but they have pharmacological effects too. Others have only pharmacological action ( histamine, kinin), due to which mainly local toxic reactions occur.

Multiple simultaneous honeybee stings (from Apis mellifera) occur with generalized toxic reaction. It is the result of the introduction into the body of a high dose of bee venom.

Different authors reported a severe intoxication after multiple bee stings a versatile and multi-organ pathology. Except with local and systemic allergic symptoms, the disease can occur with rhabdomyolysis, acute renal failure and toxic hepatitis [6. R S Vetter]. Casuistic cases of delayed systemic reactions such as serum disease, vaskulitis, Arthus reaction and such unusual reactions as myocardial infarction or cardiac arrhythmias, thrombocytopenic purpura, nephritis, encephalitis, optic neuritis, generalized polyneuropathy have been described.

Aim To present a casuistic case of multiple simultaneous stings from a swarm of bees with

the development of severe intoxication and anaphylactic reaction.

Case report V. I. B., 84 years old man, was hospitalized with the following diagnose: Toxic effect,

caused by stings of poisonous insects (bees). The patient was in condition after anaphylactic shock with aggravated premorbid anamnesis: chronic ischemic heart disease. He had heart rhythm disorder - absolute arrhythmia, caused by permanent atrial fi brillation of the heart. Ischemic cardio-myopathy. Chronic Congestive Heart Failure ІІ NYHA. Arterial hypertension gr ІІІ. Brain atherosclerosis, discirculatory and dysmetabolic encephalopathy were observed. The patient had prostate adenoma and presbyacusis.

1. The patient has lived in a house with a yard, where his relatives have raised honey bees. On July 31, 2011 at nearly 8 p.m. the patient stumbled, fell down and was immediately attacked by a bee swarm. Simultaneously he was stung by hundreds of bees in the face, limbs and all over the body. The patient got local hyperaemia, pain, edema and infi ltrates at the sites of the stings. He complained of general indisposition, faintness, vertigo, pain in the chest and epigastric, shortness of breath, tachycardia, limbs formication sensation, fever, sweating, nausea and vomiting. Тhe patient was unconscious for a while. The fi rst emergency medical treatment was given by a medical team in ChervenBriagHospital at 9 p.m. The manifestations of anaphylactic shock were overcome and sanitary transport was provided.

2. The patient was hospitalized at 11 p.m. in severe general condition, with dizziness and dysarthria, hardly cooperative and disoriented. He has no fever, but and he had acrocyanosis. In the patient’s skin of the face, body, limbs and hairy part of the head there were more than 540 bee stings. The area around the sting puncture marks had local erythema, infl ammatory infi ltrate, and pus-like pustules with a necrotic center here and there. The patient had diffuse erythema of the face and edema of the soft tissues. The visible mucosae had conjunctival inection. The breathing was spontaneous with 22 ins/min frequency. The auscultation showed bronchospasm and lung stagnation. The heart activity

Page 188: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

186

was arrhythmic because of permanent atrial fi brillation of the heart, muffl ed heart tones, without murmurs, blood pressure of 130/80 mmHg (after medication). The belly was soft and non painful. Arthritic changes of the limbs and mild passive movement defi ciency were observed.

3. The laboratory tests detected high blood sugar, urea and creatinine, coagulation status disorders, metabolic acidosis and hypoxemia – correctedinthecourseoftreatment.

Laboratory Tests: ESR15 mm, Hb 133 g/l; Еr 5,29 х1012/l; Hct 0,382; Leuсо 9,3 х109/l; Thr 187 х109/l; CCA: Gra 85,4%; Ly 10,2%; Mo 4,4%; bloodsugar 9,0 mmol/l; urea 13,3 -15,1 mmol/l; creatinine 179 – 139 μmol/l; generalalbumen – 65 g/l. Ionogram - Nа 137 mmol/l; К 4,8 mmol/l; CL 104 mmol/l. Coagulation - Prothrombinindex 22% - 64%; INR 3,20 - 1,34; fi brinogen 4,06 g/l; Kaolin cephalin clotting time 19,9 sec – 42,2 sec.

There were no records of anemia, hemolysis, icterus and bleeding from gastro intestinorum system.

ECG data showed absolute tachyarrhythmia because of permanent atrial fi brillation of the heart. Abnormal QS-teeth on the lower wall of the left ventricle and V1 – V4 (probably spent acute myocardial infarction, an old one).

Treatment: Extraction of more than 540 bee stings; local anti-infl ammatory mixtures; infusion of glucose and electrolyt solutions in capacity up to 1000 ml/d; systemic corticosteoroids - Urbason 3 mg/kg/d i.v.; Н1 blockers - Allergosan i.m., Aerius p. os; Н2 blocker - Quamatel i.v.; antibiotic - Medaxone 2 g/d i.v.; vasodilatators – Cavinton 2x1 amp. i.v. diuretics - Sol. Mannitoli 10% 500ml, Furantril x1 amp. i.v.; vitamins (Vit. C, Vit. B complex); Concor 5mg/per day.

Course of illness The general patient condition improved quickly. The patient’s consciousness started

to clear up on 1st day, but till his discharge from hospital manifestations of discirculatory encephalopathy prevailed. The transient oliguria, which had been found out, was overcome by 5th hour from the initial time of hospitalization after the applying of furanthril. The subsequent manifestations of acute kidney insuffi ciency, hemolysis, rhabdomyolysis and liver changes were not observed. Infl ammatory skin changes were observed to be faded by 5th day (pic. 5, 6, 7, 8). After the health stabilization the patient was discharged from hospital in the improving condition on 6th day.

pic. 5 pic. 6 pic. 7

pic. 8

Page 189: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

187

DiscussionThe manifestation of aggressive behaviour of the bee swarm was due to the provocative

behaviour of the patient.4. The major component of the bee venom is protein, called melittin, which constitutes

50% of dry venom. It contains 12 amino acids and possesses heavy allergenic and cytotoxic effect.

5. Multiple simultaneous bee stings bring unusually large amount of bee venom in the body. Its direct toxic effect causes multiple organ dysfunction. The concentration of apitoxin from 500 simultaneous bee stings is a lethal dose for adult humans and 200 to 300 stings cause severe poisoning.

6. In the described case the manifestations of anaphylactic shock were managed overcome during the prehospital period. Angioneurotic edema was infl uenced successfully by the systemic anti allergic therapy. The toxic reaction ran with the local symptomatology –pain, edema and hemorrhagic necrotic pustulous changes on the spots of the stings. Systemic toxic effects were presented mainly by the clinical picture of toxic, discirculatory and dysmetabolic encephalopathy as well as complicated myocardial ischemia and arrhythmia.

7. The determined changes in the coagulation status happened as a result of uncontrolled ambulatory anticoagulation therapy with Sintrom. The pause in the applying of the medicine led to the fast restoration of the indexes - INR.

8. The transient oliguria was a clinical manifestation of the shock condition and the extrarenal nitrogen retention was due to accompanying pathology - prostate adenoma.

9. Our observations coincide with those described by other authors, that vasoconstriction as a result of vasoactive and infl ammatory mediators released after stinging, contributes to cerebral and myocardial ischemia. Vasodilatative, anti-ischemic and antiplatelet effect of intravenous Cavinton in combination with osmotic diuretic mannitol therapy successfully overcome the symptomatology.

10. Despite the great number of stings, advanced age of the patient and the accompanying premorbid pathology the intoxication outcome is favorable.

Conclusion 11. During the period 2000 - 2012 was established a lasting trend of increasing the

number of severe toxic allergic reactions caused by insects of the Hymenoptera order that require hospitalization.

12. For the same period among morbidity in the clinic are not established cases of multiple bee stings as described.

References:

[1] Betten D., W. Richardson, T. Tong et R. Clerk. Massive honey Bee Envenomation-Induced Rhabdomyolyin an Adolescent. Pediatrics vol 117, №1, 2006 1 231-235.

[2] Iliev Y., St. Tufkova, M. Prancheva, A rare case of severe intoxication from multiple bee stings with a favorable outcome. Folia Medica 2010; 52(3): 74-77 doi: 10.2478/v10153-010-0010-5

[3] Insect bites and stings -

Page 190: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

188

[4] Kini PG, Baliga M et Bhaskaravad N. Severe derangement of the coagulation profi le following multiple bee sting in a 2 year-old boy Ann Trop. Paediatr. 1994; 14, 153-5

[5] Koszalka MF. Multiple bee sting with haemoglobinuria et recovery. Bull USArmy Med Dept. 1949; 9, 21-217.

[6] Mьller UR., Hymenoptera venom anaphylaxis and cardiovascular disease. Hautarzt. 2008 Mar;59(3):206, 208-11. http://www.ncbi.nlm.nih.gov/pubmed/18259720

[7] Nittner-Marszalska M, Małolepszy J, Młynarczewski A, Niedziółka A.Toxic reaction induced by Hymenoptera stings. Pol Arch Med Wewn. 1998 Sep;100(3):252-6.

[8] Rajendiran C, Puvanalingam A, Thangam D, Ragunanthanan S, Ramesh D, Venkatesan S, Sundar C. Stroke after multiple bee sting. J Assoc Physicians India. 2012 Feb;60:122-4.http://www.ncbi.nlm.nih.gov/pubmed/22715562

[9] Shilkin KB, BT Chen, OT Khoo, Rhabdomyolysis caused by hornet venom. Br Med J. 1972 January 15; 1(5793): 156–157. PMCID: PMC1787091

[10] Vetter RS, P K Visscher and S Camazine. Mass envenomations by honey bees and wasps. West J. Med. 1999, april; 170 (4): 223-227

http://emedicine.medscape.com/article/833315-overview Updated: Aug 29, 2011[11] Vetter RS, Visscher PK. Bites and stings of medically important venomous arthropods. Int J Dermatol.

1998 Jul;37(7):481–496. [PubMed][12] Young Kim et al. Severe rhabdomilysis and acute renal failure due to multiple wasp stings. Nephrol Dial

Transplant, 2003, vol 18, n.6, p.1235

Page 191: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

189

Chapter 24

Special Features in the Treatment after Pepper Spray KO jet Expoture in Childhood ( clinical case)

Katerina STEFANOVADepartment of Clinical Toxicology

UMHAT “Dr Georgi Stranski” – Pleven, Medical University – Pleven, Bulgaria

Abstract: The Pepper spray (OC spray) is a refl ex irritant containing Oleoresin Capsicum (oil extract of hot red pepper) and is used as non-lethal agent deterrent in a riot control and personal self-defense. A clinical case of toxic infl uence on an 11-year-old boy, after Pepper spray KO Jet exposure is presented. The concentration of the active ingredient capsaicin is 11%. The clinical picture is manifested by typical signs of severe irritation of the eyes, burning pain, refl ex blepharospasm, photophobia, erythema and edema of the eyelids, abundantly lacrimation and rhinorrhea, agitation, disorientation, uncoordinated movements, temporary blindness and incapacity. Rapidly and permanently taking hold of the symptoms is reported only after decontamination with water and application of local anesthetic Alcain eye drops (Proparacaine 0,5%). Our results supports the view that local ophthalmic anesthesia optimizes treatment, leading to early recovery of the patient and his capability, reduces the risk of complications and permanent damage to the visual analyzer. Rapid and effective management of pain is a prerequisite for preventing the psycho-emotional stress in children exposed to Pepper spray.

Key words: Pepperspray, exposure, localanesthetic, Alcain, OleoresinCapsicum, OCspray, capsaicin.

Introduction:Sprays with irritate poisonous substances are used more and more often in our daily

lives as a means of riot control, guard and self-defense. The most popular are considered CS, CR and OC, which are offered in different packaging variations like pens, deodorants, key chains etc. The free access to their purchase represents a signifi cant risk of their uncontrolled and not always of lawful use, including minors.

Pepper spray (OC spray) is a refl ex irritant containing Oleoresin Capsicum (oil extract from a special variety of hot red pepper). The concentration of the active ingredient capsaicin in various sizes may be from 3 to 11% solution, depending on the manufacturer. Pepper spray KO Jet is the strongest pepper spray available on the European market, containing 11% Oleoresin Capsicum (OC). It is equally effective against aggressive, drunk or drugged people, as well as against animals [7].

Page 192: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

190

Capsaicin acts directly on peripheral sensory nerves (n. trigeminus, n. vagus) and activates the pain receptors (nociceptors), when is in contact with face skin, eye and nasal mucous membranes. This is causing the release of tachykinins or neuropeptides (substance P and neurokinin A). That causes pain, irritation and neurogenic infl ammation [3, 4, 7]. The irritative effect is potent and begins with burning pain, a feeling of “sand” in the eyes, swelling of the eyelids blepharospasm accompanied by erythema, excessive tearing, photophobia and rhinorrhoea. This causes temporary blindness and disorientation. The inhalation causes symptoms of acute burning pain along the upper respiratory tract, irritating cough, tachypnea, feelings of breathlessness. The ingestion of capsaicin induces severe irritation with burning pain and mucous swellingsin oral and nose cavities, excessive salivation and rhinorrhoea, nausea, sometimes-vomiting, headache, diffi culties in breathing [5, 6, 7].

In case of a single exposure to OC outdoors, symptomatology is moderate, continues up to 60 minutes and usually managed without complications. The serious disabilities are associated with additional components of the sprays and high concentrations of OC when using pepper spray in small enclosed spaces. These are for example destructive changes of the mucous membranes, punctate epithelial erosions, corneal abrasions, clouding of cornea, purulent conjunctivitis, acute hypertension, toxic pulmonary edema and damage to the brain, liver and kidneys [3, 5, 6, 9, 10].

Pathophysiology of OS is irritative refl ex mechanism of action. The ensuing psychic symptoms are result of pain, temporary blindness, lost sense of direction and inability to coordinate movements. Havoc shows typical stress reactions provoked by fear, panic and lack of self-control.

The children’s psyche is especially sensitive and vulnerable and pain threshold is signifi cantly lower compared to that of the adults. The irritant causes temporary incapacity of the individual, leading to severe psycho-emotional stress [1]and creates conditions of bodily and psychological control over children [7,8]. Despite the detailed description of the effects of pepper sprays on the websites, this information does not create real enough idea about the strength of the victim’s dramatic experience. Their observation shows that out of 1531 people exposed to OC spray, which are registered in poison control centers -64% are children and adolescents. In the literature there is no data available for our country, especially for childhood and therefore we consider the presentation of this case reportas substantial.

Aim The aim of this report is to present an effective therapeutic approach with administration

of topical ophthalmic anesthetic suitable for rapid relief of symptoms when exposed to Pepper spray in childhood.

Case report An 11-year-old boy (medical history № 17705/2012) was hospitalized with diagnosis:

Toxic effects of Pepper Spray (moderate degree). Conjunctiuitis acuta oc. utr. Dermatitis contacta irritativa. The patient is without comorbidities and allergies.

The patient has been sprayed on the face with Pepper spray KO Jet during a “game” from a friend after school hours. The symptoms manifested immediately after exposure with severe pain, blepharospasm, lacrimation, photophobia, burning in the eyes, on the skin of the face, ear and neck clams. He becomes agitated, restless, with uncoordinated

Page 193: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

191

movements, but he has no shortness of breath and throat and nose irritation. First pre-hospital care is provided by the school nurse, so eyes and face are washed with cold water. There is a report to 112 to police and emergency medical services.

On the twentieth minute from the incident the patient enters the Emergency department of University Hospital Pleven in clear consciousness, contact, but agitated, restless, disoriented at times, with increased motor activity - uncoordinated movements of limbs and continuously rubbing his face and eyes. The ophthalmic examination establishes refl ectory blepharospasm and photophobia, moderate mixed conjunctive redness with tears, intact corneas. Anterior segment is without aberration. Fundoscopy - papillae, maculae, vessels are without aberration. Other visible mucous membranes are disengaged. The voice is clear. Respiratory system – tachypnea, respirations at 24 breaths / min, with no evidence of dyspnea, auscultatory - clean vesicular breathing. Cardiac activity is rhythmic with frequency of 90 beats / min, blood pressure of 90/60 mmHg. The remaining somatic status is without bias.

Laboratory tests reveal leukocytosis 14.6 x 109 / l. Acid-base balance is in reference values. ECG - sinus rhythm without changes in repolarization.

Our treatment includes: Decontamination of the face and eyes with a tangential stream of cold water; application of topical ophthalmic anesthetic Alcain eye drops (Proparacaine, Proxymetacaine 0,5%) - two drops in each eye, ten minutes after entry into ER; infusion of glucose-electrolyte solutions; Systemic corticosteroid - Urbason 2 mg/kg/24 h a scheme; H2 blocker therapy - Quamatel; Ca gluconici, vitamin “C” iv; topical ophthalmic therapy -Ciloxan coll. 5x1 drop in both eyes and Tobrex ung. ophthal. evening.

The development of the clinical picture of toxic effects of Pepper spray KO Jet - moderate degree is presented in Table 1. The symptoms were studied in the following time intervals - at admission to the emergency room, immediately after the 10-minute decontamination with water, the fi fth and the tenth minute after administration of local anesthetic Alcain eye drops, and later on the 1st and 24th hour.

Table 1. Development of clinical symptoms after exposure Pepper spray KO Jet (moderate degree).

Time and applied treatment

Entry into the ER and start of

decontamination with water

After 10 minutes decontamination

with water

5 minutesafter Alcain

10 minutesafter Alcain

1st

hour24th

hour

Intensity of the pain - 10-speed VAS 9 th level 8 th level 4 th level 2 nd level 0 level 0 level

Burning + + + + + + - - -

Blepharospasm + + + + + + - - -

Lacrimation + + + + + + + + + - -

Swelling of the eyelids + + + + + + + + + + -

Psycho-emotional condition

agitated, restlessanddisoriented

restless, butoriented

visibly relaxed relaxed relaxed relaxed

Capacity incapacitated incapacitated recovered capacity YES YES YES

Page 194: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

192

The intensity of the pain was evaluated using a ten-speed visual analogue scale (VAS) and then every 10 minutes for a total of 60 minutes. Duration of blepharospasm, burning, lacrimation and swelling of the eyelids and also the change of the psycho-emotional condition and capacity of the patient has been observed.

When a patient enters to the Emergency Department (ED) the survey showed pain intensity from the 9 th level, presence of burning, powerful blepharospasm, profuse tearing and swelling of the eyelids, agitation and disorientation. After 10 minutes decontamination with water the patient is still agitated, restless and incapacitated. He felt severe pain from the 8th level, persist tearing and swelling of the eyelids but blepharospasm and burning are insignifi cantly reduced. On the fi fth minute after using a local anesthetic Alcain are reported meaningful improvements in signs - twice lessening of pain from eighth to fourth level, slightly stinging, blepharospasm and swelling of the eyelids, still with tearing. Patient is visibly relaxed with recovered capacity. Within tenth minute (photo 1) of the local anesthesia there has been observed controlling of the symptoms: pain is the second level, there is no refl ex blepharospasm, no subjective burning sensation, slightly residual tearing, swelling of eyelids has absorbed. On the fi rst hour of the investigated signs is at hand only mild edema of the eyelids. Erythema of the skin, ear and neck clams disappeared up to the fi fth hour, as a result of timeliness application of topical ophthalmic anesthetic and systemic corticosteroid and antihistamine therapy. On the 24th hour the patient is asymptomatic.

Discusion: The presented case after exposure to irritant Pepper spray KO Jet is rare in childhood.

It is complicated with contact conjunctivitis and dermatitis, with no evidence of inhalation or ingestion of aerosols. Used OC Spray has a very high 11% concentration of the active ingredient capsaicin, with a degree of spicy 2,500,000 SHU (Scoville Heat Units).

In our case report the symptoms (burning pain, tearing, blepharospasm) are not managed in outpatient period. The signs persist to the fullest extent upon admission to the emergency department on 20th minutes of the incident. The patient is highly agitated, disoriented and incapacitated. The signs of refl ectory, painful and irritative syndrome disappear 5 minutes after repeated decontamination with water combined with a local anesthetic Alcain eye drops. Psycho-emotional stress was overcome and capacity of the patient is recovered.

Oleoresin Capsicum is an oily extract. Typically decontamination of the eye is performed by a strong tangential water stream, which lasts at least 15 minutes. However, this is diffi cult to achieve due to refl ex blepharospasm and swelling that do not allow full volitional opening of the eyelids. OC spray can be removed from the skin using specially developed in recent years decontamination wipes or sprays.

Our observations coincide with those described by other authors that rapidly and permanently taking hold of the symptoms is reported only after application of local anesthetic Alcain eye drops [5, 10, 11]. Proparacaine relieves the pain and also by interrupting the refl ex arc overcomes blepharospasm and supports decontamination with water. The local ophthalmic anesthesia signifi cantly relieves burning, protects swelling of nasopharyngeal mucosa and removes the subjective sensation of a foreign body “grains of sand” in the eyes. It prevents local rubbing of the eyelids of the patient which minimizes the risk of

Page 195: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

193

supplementary contamination and development of destructive injury to cornea, secondary bacterial infection, as well as enhanced swollen syndrome due to mechanical friction.

Conclusion:1. The use of the local anesthetic infl uences symptomatology successfully, shortens

its duration and reduces the risk of complications and permanent damage to the visual analyzer.

2. The Rapid and effectively curb of the pain syndrome is a prerequisite for preventing the psycho-emotional stress in children exposed to Pepper spray.

3. The local ophthalmic anesthesia optimizes treatment, leading to early recovery of the patient and his capability.

Lately in the context of increasing social tension is appropriate to increase public awareness about clinical effects of Pepper spray, as well as using approbated effective therapy that is appropriate for minors exposed to riot control agents.

References:

[1]. Cohen MD, The health effects of pepper spray: a review of the literature and commentary. J Correctional Health Care 1997;4:73-89.

[2]. Forrester MB, Stanley SK, The epidemiology of pepper spray exposures reported in Texas in 1998-2002. Vet Hum Toxicol 2003 Dec; 45(6):327-30. PubMed ID 14640489

[3]. Holopainen JM, Moilanen JA, Hack T, Tervo TM, Toxiccarriersinpepperspraysmaycausecornealerosion. Toxicol Appl Pharmacol 2003 Feb 1; 186(3):155-62. PubMed ID 12620368

[4]. Hui K, Liu B, Qin F, Capsaicin activation of the pain receptor, VR1: multiple open states from both partial and full binding. Biophysical journal, 2003, May; 84(5):2957-68.

[5]. Konov V, Early recovery of the servicemen after training with an OC - spray (PEPPER - spray) and treatment with a local anesthetic. Military Medicine 2009, p. 22-24, ISSN 1312-2746

[6]. RegaPP, Irritants - CS, CN, CNC, CA, CR, CNB, PS.http://emedicine.medscape.com/article/833315-overview Updated: Aug 29, 2011

[7]. Smith G, Stopford W, Health Hazards of Pepper Spray. NCMJ 1999 Sept/Oct; 60(5):268-274. Vol. 60 Number 5

[8]. Tominack RL, Spyker DA. Capsicum and capsaicin: a review: case report of the use of hot peppers in child abuse. Clin Toxicol 1987;25:591-601.

[9]. Vesaluoma M, Mьller L, Gallar J, Lambiase A, Moilanen J, Hack T, Belmonte C, Tervo T, Effects of oleoresin capsicum pepper spray on human corneal morphology and sensitivity. Invest Ophthalmol Vis Sci 2000 Jul; 41(8):2138-47. PubMed ID 10892855

[10]. Zollman TM, Bragg RM, Harrison DA, Clinical effects of oleoresin capsicum (pepper spray) on the human cornea and conjunctiva. Ophthalmology 2000 Dec; 107(12):2186-9. PubMed ID 11097593

[11]. Sharma AN, Management of Ocular Chemical Injuries. J Toxicol Clin Toxicol 2004;42(4):421-2

Page 196: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

194

Chapter 25

Suicidal Self-poisonings and Resilience

Plamen VANEV, Julia RADENKOVA-SAEVAToxicology Clinic, UMHATEM „ N.I.Pirogov”,

21 Totleben Blvd., 1606 Sofi a, Bulgaria,e-mail:[email protected]; [email protected]

Abstract. The aim of the study is to determine the protective factors for prevention of a suicidal activity. According to our ten years study (2002-2012) at the Toxicology Clinic, Emergency University Hospital “Pirogov”, the demographic distribution of self-poisonings indicates that this type of suicidal activity is the most typical for the age group up to 25 years – (32%). The psychological resilience is associated with those protective factors that help overcoming the suicidal thoughts and feelings and serves to prevent the suicidal behavior. Using the adapted for Bulgaria method “Suicide Resilience Inventory – 25” we prove the existence of such three factors: emotional stability, external protection and internal protection. The main features underlying the suicidal behavior are: emotional instability (anxiety, sensitivity, affective disorders), a failure of external support (a loss of a signifi cant person for the loved one, family problems, abuse from the others), internal mental characteristics (cognitive rigidity, a lack of self-control, masochistic tendencies, a low self-esteem). The results of the study aimed to primary and secondary prevention of suicidal activity.

Key words: suicidal self-poisonings, resilience, protective factors

IntroductionSuicide is a phenomenon which clearly manifested the drama of human free choice.

The attention of philosophers since ancient times has been focused on death and dying as a crossroads between existence and non-existence. In this phenomenon is interwoven several aspects – from legal and moral to absolutely pragmatic. For Bulgaria, it is a signifi cant problem because the country is in the top of the world twelfth suicide rating. Man resorts to suicidal acts mostly because it requires a fl exible change but for some reason he can’t fulfi ll it in a different way than suicide.

After 80-th years of the twentieth century through “concept resilience”, in psychology and psychiatry enters one new approach. This concept explains why some people, even under extremely adverse external (e.g. unreliable family) or internal (e.g. disease) conditions, able to cope and even emerge from the crisis stronger and wiser. The resilience concept is related to the future and focuses on the factors, which determine fl exible recovery from external and internal stress events. Concerning to suicidal behavior, it is looking for so called “buffers”, which would help to reduce the risk suicidal thoughts to become actions,

Page 197: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

195

habits and destiny. So, suicidal resilience is: “ ... opportunities, resources or competence to regulate suicidal thoughts, feelings and attitudes.” [5].

Suicide is extremely complex, particularly human phenomenon. We believe that on the suicidal behavior affect fi ve groups of factors and in each case they are uniquely intertwined:

1. Physical environment (space, meteorological, geographical, etc.).2. Instrumental factors for to put an end to the life.3. Biological (structure and functioning of the body).4. Mental functioning of individuals at all levels.5. Social characteristics of different in importance and size communities.The second factor of those listed fi ve factors - the instrumental one, is connected to

a subjectively accepted (or planned) method, manner, or “tool” to put an end to life or realizing parasuicidal action.

A numerous suicidal methods exist. These methods vary in their severity and specifi city, but one of the most widespread of them is the self-poisoning. His psychological characteristics varied from demonstrative parasuicidal gestures to severe destructive acts [6].

These characteristics of the method of self-poisoning, make the research in this area a signifi cant one - a model for the study of suicide.

The aim of the study is to identify resilience (“Buffer”) factors that reduce the risk of suicidal behavior. The study includes 3890 patients, hospitalized in the Toxicology Clinic, Emergency University Hospital “Pirogov,” Sofi a, Bulgaria for ten-year period (2003–2012).

Methods: „Suicide Resilience Inventory – 25” [5]; Demographics method. Studied people:• 3890 persons with intentional self - poisoning, treated at the Toxicology Clinic.• 112 adults, made suicide attempts by self-poisoning (26 of them male).• Parallel control group (112 adults, 26 of them are male) - people with no evidence of

suicidal behavior to date.Results: Factor analysis results, obtained by using the SRI-25 method show the three-factor’s

explanation of the phenomenon (see Table 1).1. Emotional Stability, refl ecting the functioning of the emotional sphere of the

individual (the highest rate).2. External Protection - associated with the ability to obtain help and support in a

diffi cult moment by individuals and groups (with a smaller percentage).3. Internal Protection - refl ecting what gives meaning to one’s life.

Table 1. Results of factor analysis

Factors Rotation Sums of Squared Loadings

eigenvalues % of Variance Cumulative %1. Emotional Stability 6.01 24.06 24.062. External Protection 5.12 20.51 44.583. Internal Protection 4.94 19.79 64.37

Page 198: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

196

The comparison between the group of attempted suicide and those in the control group persons shows that resilience factors have a worse performance in the suicidal group of patients with a high statistical signifi cance (see Table 2).

Table 2. Comparison of studied people in resilience factors

Mean Mean Std. Dev. Std. Dev. Р

FactorsControl group

Suicide group

Control group

Suicide group

1. Emotional Stability 5.38 3.43 0.89 1.36 >0.001

2. External Protection 5.27 3.88 0.88 1.25 >0.001

3. Internal Protection 4.95 3.39 0.75 1.08 >0.001

General Resilience 5.19 3.56 0.68 1.04 >0.001

Demographic analysis of suicidal self-poisoning in 3890 people gives indirect evidence.

Dominate female individuals. The women have a lower emotional stability. This is refl ected in a higher frequency of mood affective disorders among them (see Table 3).

Table 3. Distribution by sex

Gender Number Percent

male 817 21

female 3073 79

total 3890 100

Dominate people at a young age (up to 25 years - 32% of the studied people), which is related to their emotional immaturity (see Table 4).

Table 4. Distribution according to age

Age Number Percentunder 16 320 8.2

16-25 927 23.8

26-35 1119 28.8

36-45 745 19.2

46-55 551 14.2

over 55 228 5.8

total 3890 100

Page 199: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

197

According to the employment it can be concluded that all groups have their specifi cs -for example the unemployed have a problem with understanding their life (“I’m that, whatI work”, as they say in our civilization) which is determined from the defi ciency of the factor “internal protection” (understanding the life and identity).Working people often experience prolonged stress, so their emotional stability is violated. For the pensioners the most important factors are the loss of relatives (external protection), poorer health (internal protection), and as well as emotional instability. For the young people theproblems in today’s schools are associated with increased nervous strain, violence and diffi cultiesin the styles of sense making [1]. This is a reason for the deterioration of all buffer factors (see Table 5).

Table 5. Distribution according to employment

Status of Employment Number Percent

Employed 1471 37.82

In education institutes 897 23.06

Housekeepers 88 2.26

Unemployed 664 17.07

Retired 770 19.79

Total 3890 100

The marital status - (dominated singles) refl ects most common low social support (external protection) (see Table 6).

Table 6. Distribution according to marital status

Family Status Number Percent

Married 1153 29.640

Unmarried 2204 56.658

Divorced 255 6.555

Widowed 278 7.147

Total 3890 100

The monthly distribution of suicide self-poisoning showed an increase of suicidal phenomena in the spring months, mostly associated with emotional instability and more frequent affective disorders in this season (see Table 7. and Figure 1.).

Table 7. Distribution by month of committed suicide action

Months Number Percent

January 330 8.483

February 301 7.738

March 367 9.434

Page 200: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

198

April 340 8.740

May 421 10.823

June 401 10.308

July 298 7.661

August 280 7.198

September 274 7.044

October 298 7.661

November 302 7.763

December 278 7.147

Total 3890 100

Figure 1. Distribution by month of attempted suicide action

DiscussionWe can consider it is proven that there exist some peculiar buffers for overcoming

the suicidal crisis, which defi nitely are worsen for the suicidants. They are: the emotional stability, the external and the internal protection. These buffers are in a theoretical harmony with the rich suicidal literature [2, 9]. Indirectly are found considerable evidences at the analysis of the demographic characteristics of the persons, attempted suicide self-poisoning.

Although too doubtful theoretically, the idea for the so-called “Suicidal disease”1 (See Figure № 2) enables the outlining some preventive measures in accordance with the above described factors of resilience [10].

On the matter of increasing the emotional stability, the purposes and methods of prevention [8] are related to:

• Psycho education since the earliest age for better emotional functioning.• Access to a psychological and psychiatric care for the groups in risk.• Effective treatment and rehabilitation of the affective and other disorders leading to

emotional instability.

Page 201: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

199

The External protection is associated with:• Engagement of the Social Institutions when there is a risk of suicidal behavior.• Creation of “Befriender” societies.• Group Psychotherapy.• Family consulting.Concerning on the internal protection signifi cant aspects for the prevention of suicidal

phenomena are:• Support of all age and social groups for fi nding the meaning of life2.• Engagement of the Institutions with the group’s fi nding the meaning of life: social,

religious, political.• Psychotherapeutic work in existential plan [4].

As a summary of the proposed by us model we offer Figure №3. In each stage of the development of the mental crisis and its escalating into a suicidal one there are acting peculiar resilient factors and their insuffi ciency determines the extending of the crisis. The effective infl uence of all these factors leads to overcoming in all of its stages [3].

Notes:1 These theories originate long ago from the classics of Psychiatry. For example even

Esquirol at the beginning of 19-th century ( in his book Des Maladies Mentales - 1838) considers that the suicide has all the signs of the mental disease – and only in a state of madness the person is able to attempt at his own life and all the suicidants are mentally ill people [10].

2 “Wer ein Warum zu leben hat, erträgt fast jedes Wie” - Nietzsche F. (“Anyone who has something to live for can bear almost everything.”) [4].

Figure 2. Model of the suicidal disease and prevention

Page 202: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

200

Figure 3. Stress, suicidal crisis and resilience

References

[1]. Boor M., Relationship between Employment Rates and Suicide Rates in Eight Countries: 1962-1967. In: Psychological Report, 47, 1980, p. 1095-1101.

[2]. Caplan G., Emotional Crises In: The Encyclopedia of Mental Health, 3, 1963, p. 521-532.[3]. De Shazer S. Putting Difference to Work. W.W.Norton&company, NY, 1991.[4]. Frankl V.E., Logotherapie und Existenzanalyse, Munchen 1987, p. 251,[5]. Gutierrez P., Osman A.Adolescent Suicide: An Integrated Approach to the Assessment of Risk and

Protective Factors. Ilinois, Northern Illinois Univ. Press, 2007.[6]. Kessel N. Selfpoisoning. In: Essays in Self-Destruction. Ed. by Edwin S. Shneidman. N.Y., Science House,

1967, p. 345-372.[7]. Pokorny A. Myths about Suicide. . In: Suicidal Behavior. Ed. By H. L. P. Resnik, N. Y., 1968, p. 57-72.[8]. Shneidman E., Farberrow N. The Suicide Prevention Center in Los Angeles. In: Suicidal Behavior. Ed. By

H. L. P. Resnik, N. Y., 1968, p. 367-380.[9]. Shneidman, E.S. Ten commonalities of suicide and their implications for response.In: Crisis, 1986, 7(2),

p. 88-93.[10]. Williams J.M.G., Cry of Pain – Understanding Suicide and Self-Harm. London, Penguin Books, 1997.

Page 203: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

201

Chapter 26

Toxicity of Amlodipine in Combination with Enalapril – a Case of Suicide

Nikola RAMSHEV a, Severina DAKOVA a, Zorka RAMSHEVAb, Konstantin RAMSHEV a , Kamen KANEV c

a Clinic of Intensive TherapyMilitary; b Department of Clinical Laboratory,c Department Disaster Medicine and Toxicology

MilitaryMedicalAcademy, Sofi a, Bulgaria

Abstract. The purpose of the article is to demonstrate a case with unsuccessful outcome due to delayed hospitalization and resistance to the therapy. It concerns a young woman who had swollen 30 tablets of Enalapril and 30 tablets of Amlodipine for suicidal purpose. She has been brought by the Emergency Ward Team on the next day in condition of shock with mixed genesis, acute cardio-vascular, pulmonal and renal insuffi ciency, dyselectrolytemia, toxic myopericarditis, pulmonal edema, and severe depressive syndrome. The full spectrum of blood, gas and toxico-chemical laboratory analysis as well as functional tests was performed. In spite of the adequately initiated intensive therapy, cardiopulmonary resuscitation and the attempt for detoxifi cation by an exchanged blood transfusion, the patient progressed and developed polyorgan insuffi ciency with a fatal outcome on the forth day of admission to the clinic.

Key words – toxicity, Amlodipine, Enalapril

Introduction:Calcium channel blocking (CCB) agents as well as ACE inhibitors are medications

widely prescribed for treatment of cardiovascular diseases. Amlodipine belongs to the class of dihydropyridine calcium channel blockingagents

and is prescribed for treatment of essential hypertension and angina pectoris. Referring to its mode of action, amlodipine inhibits calcium infl ux into cardiac and vascularsmooth muscle cells resulting in dilation of both arteries and arterioles. Amlodipine is a peripheral arterial vasodilator that acts directly on vascular smooth muscle to cause a reduction in peripheral vascular resistance and reduction in blood pressure. The drug demonstrates slow absorption after oral administrationwith a peak plasma concentration observed after 6 to 9hours [1]. Elimination half-life at fi rst dose is 45.6 +/- 10 hours which is the longest half-life of all calcium channel blockers [2]. Amlodipine seems to have a linear pharmacokinetic profi le; the dose is correlated with the mean peak plasma concentration [3]. There is data available for intoxication with CCB agents like Diltiazem, Verapamil and Nifedipine, however there is limited clinical experience with Amlodipine intoxication [4]. Amlodipine poisoning is very rare, and only a few cases with serious or fatal overdose of

Page 204: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

202

amlodipine poisoning have been reported so far.There is an information summarized in an article published in Basic & Clinical Pharmacology & Toxicology, 2006[5]describing four fatal cases for the period 1995-2005 where all patients developed severe symptoms such as sustained hypotension and coma.

Enalapril is an Angiotensin Converting Enzyme (ACE) inhibitor used in the treatment of hypertension and congestive heart failure as well as for prevention of symptomatic heart failure in patients with asymptomatic left ventricular dysfunction (Ejection Fraction < 35%).

Limited data are available for Enalapril overdose in humans. The most prominent features of overdose reported to data are marked hypotension, beginning in 6 hours after ingestion of tablets in combination with blockade of the renin-angiotensin system, and stupor. Symptoms associated with ACE-I overdose may include circulatory shock, electrolyte disturbances, renal failure, hyperventilation, tachycardia/bradycardia etc.

Enalapril-induced acute hepatotoxicity has been described to be with an unusual morphology characterized by macro- and microvesicular steatosis associated with neutrophil infi ltration and Mallory bodies, occasionally with satellitosis. This morphology may be referred as „predominantly periportal steatohepatitis” [6].

Serum enalaprilat levels 100- and 200-fold higher than usually seen in therapeutic doses have been reported after ingestion of 300 mg and 400 mg of Enalapril, respectively.

The pharmacokinetics of amlodipine after single intravenous and oral doses and its pharmacokinetics after repeated oral doses in healthy male volunteers have been evaluated and summarized in the below tables (Tables 1 and 2) [7]:

Table 1. Pharmacokinetic parameters of 10 mg amlodipine after single intravenous and oral doses (mean +/- S.D.)

Intravenous OralPeak concentration (ng/ml) - 5.9 +/- 1.2Time to peak (hr) - 7.6 +/- 1.8Biovailability (%) - 64 (range 52–88)

Clearence (ml/min./kg) 7.0 +/- 1.3 -Volume of distribution (l/kg) 21.4 +/- 4.4 -Half-life (hr) 33.8 +/- 5.3 35.7 +/- 6.1

Table 2 Pharmacokinetic parameters of 15 mg amlodipine after 14 repeated oral doses given once daily (mean +/- S.D.)

Day 1 Day 14

Peak concentration (ng/ml) 6.9 +/- 2.6 18.1 +/- 7.1Time to peak (hr) 8.9 +/- 3.7 8.7 +/- 1.9Average concentration (ng/ml) 4.5 +/- 1.6 14.5 +/- 5.8Half-life (hr) - 447 +/- 8.6

The special warning for the use of Enalapril is symptomatic hypotension. An excessive hypotension has been observed in patients with severe congestive heart failure, with or

Page 205: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

203

without associated renal insuffi ciency which might be associated with oliguria and/or progressive azotemia, and rarely with acute renal failure and/or death.

The lethal dose Amlodipine is in the range of 40 mg/kg based on animal studies. In most cases severe symptoms of toxicity were seen, however our case shows that intake of 350 mg amlodipine results in a serum concentration of amlodipine 20–30 times higher than peak serum concentration after single intake of 10 mg amlodipine which does not always cause severe symptoms. Amlodipine overdose carries a signifi cant risk of life threatening hypotension and bradycardia and possible refl ex tachycardia. Overdose of Calcium Channel Blockers carries a signifi cant risk of death and potentially life threatening arrhythmias occur with lower doses [8]. The main pharmacokinetic property of amlodipine is its long elimination half-life of 45 hours after repeated doses. The steady state peak and the average concentration are approximately three times higher than the corresponding concentrations observed after a single dose. Bradycardia and hypotension are the most common. The other cardiovascular effects include intraventricular conduction delays, ventricular dysrhythmias and congestive heart failure. Blockade of calcium channels in cardiac muscle results in negative inotropic and chronotropic effects. Respiratory depression, gastrointestinal symptoms, central nervous depression, with or without seizure and coma are also reported [9]. Pulmonary edema not consistent with the degree of myocardial depression has been observed and reported [10]. Amlodipine blocks L-type calcium channels in the pancreatic islets cells which might lead to hyperglycemia and acidosis and, hence requirehyperinsulinaemia-euglycaemia therapy to reverse cardiovascular collapse [11].

The following steps are included in the common therapeutic procedure in case of CCB overdose [10]:

1. Hemodynamic status assessment 2. Airway control 3. Aggressive decontamination with charcoal 4. Whole bowel irrigation (multiple doses of charcoal should be given every 4 hr. in

case the whole bowel irrigation cannot be done)5. Pharmacologic therapy (include atropine for bradycardias, bolus doses of calcium

salts if case of excluded concomitant digoxin toxicity, and hyperinsulinaemia-euglycaemia therapy). In case of persistence of the symptoms of toxicity the continuous calcium therapy is followed by a phosphodiesterase inhibitor, glucagon, and adrenergic agents which are added consequently. Isolation from all hepatotoxic agents should be considered for patients with intoxication of Enalapril. A liver biopsy should be done a few days after admission as well asthe corticosteroids and supportive treatment shell continue until normalization of serum enzyme levels.

6. Mechanical supportive measures (in case of pharmacology therapy failure). There is information about using of transvenous pacing with unclear clinical benefi t as well as balloon pump and bypass but data are limited so far.

The aim of the article is to describe a suicidal case of severe amlodipine overdose with concomitant overdose of Enalapril as well as to demonstrate the fast development of clinical symptoms in term of untimely admission to hospital complicating the therapeutic management.

Page 206: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

204

Clinical case summary:22 years old woman has taken 30 tablets Amlodipine in combination with 30 tablets

Enalapril for suicidal purpose in the evening on 08/June/2012. She has been brought by the Emergency Ward team at 10:00 p.m. on 09/Jun/2012 and admitted to the Clinic of Toxicology wherefrom later transferred to the Clinic of Intensive Therapy. In the clinic she has presented a shock with mixed genesis, acute cardiovascular failure, “shock kidney”, acute renal failure-anuria, dyselectrolytemia, “shock lung”, noncardiogenic pulmonary edema-APF (Acute Pulmonary Failure), toxic myopericarditis, impaired glucose tolerance, and severe depressive syndrome.

Tests, consultations:Analysis of hematology toxicity:Mass Spectrometry/ 10/Jun/2012: detected substances – Phendimetrazine;

Phenmetrazine; Ephedrine; Piracetam; Metoclopramide; Felodipine; Amlodipine; Methylprednisolone acetate

Urine Analysis/ 11/Jun/2012: detected substances – Phendimetrazine; Ephedrine; Piracetam; Felodipine; Metoclopramide; Amlodipine. Blood - Amlodipine 0.14 mg/ml.

Acid-base balance: pH- 7.42, pCO2- 29, pO2- 98,SatO2- 98%.Blood glucose profi le: 20.8-18.6-10.9-15.6-8.2-8.4 mmol/L

Hematologic tests:

Hb. Er. Htc. Leu Pl. MCV Seg. Mo Lymph. ESR

131-110 4.5-3.4 0.38-0.30 25.0-22.3-20.1 255-232-193 86-86.3 84 7.3 8.8 7-6

Hemostasis:

ApTT INR Fibrinogen D-dimer LDH TPI CRP

25-23-23

1.2-1,2-1.4

2.5-1.8

Over 0.5Below 3.0

249-558

0.03-0.03-3.75

1.4

Biochemistry:

Gl. TP Alb. Chol. Tr. HDL LDL Urea Creat. UA Ser. Fer.10.4-9.6

48-44

32-31

3.6 0.94 1.13 2.02 6.3-23.2

194-449-597-565

430-390

11.1

TB OB K Na Cl Ca Amyl. AP GGTP AST ALT CPK8-9-13

5-5 5.3-3.6-5.5

141-135-139

98-101

2.2-2.4

125-327-175

92-60

13 15-30

15-20

54-53-623

Page 207: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

205

Diagnostic methods:ECG/ at admission/: AV tachycardia, frequency: 117/ min., descending ST-depression

and +/- T-waves from V1 to V4. Dynamic monitoring: sinus tachycardia, persisting ST-depression from V1 to V4.

X-ray of lungs and heart:09/Jun/2012: Expanded lungs; strengthened bilateral lung pattern; heart – no pathologic

fi ndings10/Jun/2012: No pathologic fi ndings 11/Jun/2012: Expanded parenchyma of lungs, no shades, strengthened perihilar

pattern, extended hilar shades, normal position of the heart shade, central type congestive alterations

Echocardiography:LA 36-37 mm, LV 52/39 mm, TDV 128 ml, TSV 64 ml, SV 64 ml, EF 50% (Teichholz),

septum 6-7 mm, LVPW 7-8 mm, preserved segment kinetics, MV – prolapse of the front fl ap, eccentric regurgitation – grade 1-1+ , Ao radix 22 mm, ascended Ao 24 mm, AV – tricuspid, preserved function, RV 26 mm, TV – physiologic regurgitation, PV – preserved function, AT 140 m/sec, pericardium – insignifi cant effusion in front of RV without hemodynamic effect – 2-3 mm, thickened pericardium behind RVBW. Heart rate during the examination – 120/min, abnormal position

Echography of abdominal organs:Liver, gallbladder, bile ducts, spleen, uterus – normal echography image; kidneys

within normal size, preserved structure (parenchyma pyelon index), no concretions or congestion, bladder – empty

Consultations with specialists:Therapist – 09/Jun/2012 – defi ned testsPsychiatrist – 09/Jun/2012 – Diagnosis: Depressive episode - severe, suicide attempt,

therapy prescribedToxicologist – 09/Jun/2012 – therapy prescribedHemodialysis Department – 09/Jun/2012 - The patient was not eligible for extracorporal

detoxifi cation due to unstable hemodynamic and BP: 60/30 mmHg. Clinically consulted by the toxicologist being on duty

Hemodialysis Department – 10/Jun/2012; 11/Jun/2012 – persistence of the severe hypotension and HR 120/min which does not allow extracorporal detoxifi cation as of the current moment

Hemodialysis has no effect in case of amlodipine intoxication. Toxicologist – 10/Jun/2012, 11/Jun/2012 – therapy prescribedSurgeon – 10/Jun/2012 – excluded acute abdomenClinic of Anesthesiology, Reanimation and Active Treatment – 11/Jun/2012: placed

central vein catheter

Page 208: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

206

Disease progression and therapeutic approach:Plasma Lyte 1000 ml; Ringer 500 ml; Ser. Glucose 5% 500 ml; Haessterile 500 ml;

Dopamine 5 ml/h, 1 amp./50 ml – 10 ml/h; Nootropil 5-5-5; Vit. B6 3X2 amp.; Vit. B1 4 ml; Humanalbumin 2 vials; Vit. B1 2 amp.; Quamatel 2X1 vial; Fraxiparine 0.4 ml, Ser. Glucose 10% 10 ml 2X1 amp.; Helicid 1 vial; Maxipim 2X1000 mg; Dexamethasone; Na bicarbonate; Urbason 60+60 mg; charcoal theatment; Glucagon 330 mg; Dobutamine 250 mg/50 ml 5 ml/h; Adrenaline 3 amp./30 ml; Degan 1 amp.; Helicid 2X1 vial; Vit. B12 500 mg; Preductal 2X1t.; Plavix 75 mg; Ser. Glucose 5% 500 ml +8E insulin + 3 amp. Ca Gl; Perfalgan 1 vial; 2 sacks of Erythrocytes; 1 sack of blood; Fraxiparine 0.4 ml; Alterenol 5 amp./50 ml - 4 ml/h; tramal; Ringer 500 ml; Adrenaline 14 amp.; Atropin 13 amp.; Lysthenon Ѕ amp.; Arduan Ѕ vial; Urbason 80 mg; Na HCO3 2 amp.

Discussion: It is a case of 22 years old woman presented with severe depressive syndrome who

had made a suicide attempt by swallowing of different medications (as per the anamnesis – Nordipine/ Amlodipine and Enalapril, 30 tablets of each) at 10 a.m. on 09/Jun/2012. She has been brought by the Emergency Ward Team to MMA-Sofi a in the condition of shock and admitted to the Clinic of Intensive Therapy at 07:00 p.m. on 09/Jun/2012.

Different components have been identifi ed by an extensive toxic-hematology analysis, as follows: phendimetrazine; phenmetrazine; efedrin; piracetam; metoclopramide; felodipine; amlodipine; methylprednisolone acetate.

After being admitted to the Clinic of Intensive Therapy the patient has been intensively treated with high doses intravenous catecholamines/dopamine; dobutrex; adrenaline; noradrenaline; stimulation of diuresis; glucagon; correction of alkaline-base and electrolyte disturbances; corticosteroids; gastroprotectors; infusions of aqueous-electrolyte, carbohydrate and high molecular solutions; perfusion of insulin according to the blood glucose profi le; LMWH; humanalbumin and maxipime. Gastric lavage with charcoal and enema were done.

In spite of the intensive therapy during the patient’s stay in the Clinic of Intensive Therapy a refractory shock condition has persisted by developing of “shock kidney” with anuria; increased nitrogen substances and dyselectrolytemia; and “shock lung” with pulmonic congestion. Toxic impairment of the myocardium with ECG alterations and rising of the markers of myocardial necrosis have been identifi ed. Hemodialysis treatment has not been done due to the persistence of the shock condition at the time of admission to the Clinic of intensive Therapy. An attempt for detoxifi cation by exchange blood transfusion has been performed instead. Treatment scheme has been followed by controlling of THA (Toxic-Hematology Analysis) on everyday basis according to the consultations with the toxicologist. All diagnostic and treatment activities have failed. Multiple organ failure has been developed based on the persistence of the shock condition with cardiogenic and non-cardiogenic component. After a long cardiopulmonary resuscitation in full capacity the patient died at 03.46 on 12/Jun/2012.

Conclusions:Different clinical manifestations have been observed in the previously reported cases;

all patients developed severesymptoms such as sustained hypotension and coma. The

Page 209: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

207

symptoms of hypotension and bradycardia further requiredhyperinsulinaemia-euglycaemia therapy in order to reverse the cardiovascular collapse [11].

There are also cases reported for patients with Amlodipine intoxication developing hypertension and renal failure. The long period between the intake of toxicdose enalapril and the fi rst signs of liver damage suggests the possibility of a metabolic idiosyncrasy [12].

All cases of Calcium Channel Blockers’ overdose should be regarded andmanaged as if potentially lethal.In most cases severe symptoms of toxicity were seen,however our case shows that intake of 150 mg Amlodipineresults in a serum concentration of amlodipine 20–30times higher than peak serum concentration after single intakeof 5 mg Amlodipine. The combination with another medication – in our case it was Enalapril, leads to severesymptoms, diffi cult therapeutic management and fatal outcome.

References:

[1]. Haria, M. & A. J. Wagstaff: Amlodipine – a reappraisal of its pharmacological properties and therapeutic use in cardiovascular disease. Drugs 1995, 50, 560–586.

[2]. Donnelly, R., P. A. Meredith, S. K. H. Miller, C. A. Howie & H. L. Elliott: Pharmacodynamic modeling of the antihypertensive response to amlodipine. Clin. Pharmacol. Therap. 1993, 54, 303– 310.

[3]. Clavijo, G. A., I. V. De Clavijo & C. W. Weart: Amlodipine: A new calcium antagonist. Amer. J. Hosp. Pharm. 1994, 51, 59–68.

[4]. Rasmussen, L., S. E. Husted & S. P. Johnsen: Severe intoxication after an intentional overdose of amlodipine. Acta Anaesth. Scand. 2003, 47, 1038–1040.

[5]. R. P. Poggenborg, L. Videbжk and Ib Ab. Jacobsen: A Case of Amlodipine Overdose, Basic & Clinical Pharmacology & Toxicology 2006, 99, 209–212.

[6]. G. H. da Silva, Al. Ribeiro Alves, P. Duques, T. Sevб-Pereira, El.Soares, C. Escanhoela: Acute Hepatotoxicity Caused by Enalapril: a Case Report. J Gastrointestin Liver Dis: June 2010, Vol.19 No 2, 187-190

[7]. Faulkner, J. K, D. McGibney, L. F. Chasseaud, J. L. Perry & I. W. Taylor: The pharmacokinetics of amlodipine in healthy volunteers after single intravenous and oral doses and after 14 repeated oral doses given once daily. Brit. J. Clin. Pharmacol. 1986, 22, 21–25.

[8]. Howarth, D. M., A. H. Dawson, A. J. Smith, N. Buckley & I. M. Whyte: Calcium blocking drug overdose: an Australian series. Hum. Exp. Toxicol. 1994, 13, 161–166.

[9]. Marques, I., E. Gomes & J. Oliveira: Treatment of calcium channel blocker intoxication with insulin infusion: case report and literature review. Resuscitation 2003, 57, 211–213.

[10]. Salhanick, S. D. & M. W. Shannon: Management of calcium channel antagonist overdose. Drug Safety 2003, 26, 65–79.

[11]. Boyer, E. W. & M. Shannon: Treatment of calcium-channel-blocker intoxication with insulin infusion. New Engl. J. Med. 2001, 344, 1721–1722.

[12]. Zimmerman HJ. Hepatotoxicity. The adverse effects of drugs and other chemicals on the liver. Philadelphia: Lippincott Williams & Wilkins, 1999.

Page 210: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

208

Chapter 27

Problems of Effective Clinical Communication in Cases of Acute Intoxication

Marieta YOVCHEVA, Snezha ZLATEVAClinic of Intensive Treatment of Acute Intoxications and Toxoallergy,Military Medical Academy – Sofi a, Naval Hospital – Varna, Bulgaria

Abstract: The communication between the doctor and the patient in cases of acute intoxication is a complex, dynamic and often, diffi cult process. Clinical toxicology is rich in specifi c communication problems. A unifi cation of the rules of clinical communication is a diffi cult task because of the great variety in etiology, pathogenesis and clinical presentation of exogenous intoxications, including patients from practically all age, sex, education, profession, social, intellectual and cultural groups, frequent combination with psychiatric diseases and auto aggressive behavior, prevalence of emergency cases. The toxic cerebral syndrome which has different expression, onset, dynamics and treatment response has a key importance. In connection with it specifi c communication barriers exist at every stage of the poisoning. A number of factors arising from the organization of the toxicological help also infl uence the effectiveness of clinical communication: participation of doctors and medical specialists from different specialties, great intensity and work-loading, 12-hours work-schedule, which creates a treating team instead of one treating doctor, insuffi cient information about the patient at the admittance, lack of enough knowledge and skills in clinical communication with toxicology patients. Possibilities of improving the quality of communication are discussed: differentiated approach according to the kind, urgency, toxic mental changes and treatment stage of an acute intoxication; current education of clinical toxicology communication and etc. An adaptation of the communication to the dynamics of the intoxication and an individualized approach to patients are especially important for the process of informed consent.

Key words: communication, acute intoxication, factors

IntroductionThe effective communication with patients is fundamental to effective clinical care

and good quality of medicine. [1, 2, 3, 4 ] The traditional concept of effective clinical communication is a two-way process of sharing information which involves one party (doctor or other medical specialist) sending a message that should be easily understood and a receiving party (patient), who sends feedback [5, 6]. However the transactional model of communication of Barnlund [7], stating that individuals are simultaneously engaged in the sending and receiving of messages, is more suitable for clinical communication.

Page 211: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

209

Effective verbal and non verbal communication should generate and maintain the desired effect, with the potential to increase the effect of the message. Therefore, effective communication serves the purpose for which it was planned or designed. Communication is an interaction where at least two interacting agents share a common set of signs and a common set of semiotic rules. A communicational noise is called any interference with the decoding of messages sent over a channel by an encoder. Types of noise are environmental, physiological- impairment, psychological, semantic, syntactical, organizational, cultural and etc. Communicational noise leads to barriers to effective communication that retard, distort or prevent the message exchange. Generally they are connected tophysical, physiological or psychological barriers, system design, ambiguity of words or phrases, individual linguistic ability, attitudinal barriers, modes of presentation the information and etc. The result is failure of communication process or an undesirable effect.

Clinical toxicology is rich of specifi c communication problems. The toxic changes in central nervous system as well as additional medical, psychological and social factors create barriers to effective clinical communication, which cause a number of medico-ethical, deontological and legal problems. [8, 9, 10, 11, 12, 13, 14, 15] As a result of this the important process of informed consent or informed refusal of treatment of toxicological patients has specifi c diffi culties in comparison with other clinical specialties. The factors which infl uence communication in clinical toxicology can be divided in three categories [1, 13]: I. Factors, connected with the patient. A. Pre morbid personality: 1. Auto aggressive behavior in the past or presence: suicidal ideas and acts, addictions, abuses. 2. Co morbidity: acute or chronic psychic diseases (with or without psychotropic treatment), serious somatic chronic diseases. 3. Patients that lack knowledge about poisons and/or experience to copy with toxicology situation – children, mentally retarded, senile, etc. 4. Socially weak, poor or isolated patients. 5. Patients who are afraid of a negative impact of the information about their intoxication on their family, professional and social life. B. Intoxication change of the patient’s personality: 1. High percentage of temporary, ‘acute’ toxic change of the mind as a result of direct or indirect cerebral toxicity, psychosis, stress, etc., which lead to disturbed cognitive processes, diffi cult or impossible perception and processing of information, distorted and wrong concepts, ideas, reasoning and decisions. 2. Pre morbid or toxic change of emotional processes. 3. Frequent dissimulation and less frequent - aggravation or simulation. 4. Some toxicological procedures and manipulations are unpleasant for the patient or can be perceived as humiliating, especially in cases of obtunded consciousness. II. Communication factors connected with the doctor or other medical specialist: 1. Different level of toxicology knowledge and experience. The specialists of clinical toxicology in Bulgaria and most of other countries are quite few. Medics from different clinical specialties participate in diagnostics and treatment of toxicology cases: emergency medicine, general medicine, family doctors, internal diseases, pediatrics, anesthesiology and reanimation, image diagnostics, laboratory, etc. 2. Different level of communication knowledge and experience in clinical medicine and in particular – in communication with patients of clinical toxicology. Different level of training of medical students in this area. 3. Different concepts of the proper general model of patient-doctor interrelation (paternalism, autonomy or collaboration) and, in particular – of the applicability of each model in clinical toxicology. 4. Personal characteristics: age, physical health, professional level, work-load, level of exhaustion, co-existing personal, professional

Page 212: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

210

or social problems, etc. III. Communication factors connected with the professional environment and organization: 1. First toxicological help – usually emergency teams and units or specialized toxicology rooms, but in practice can be any medical unit. Acute intoxications have high level of emergency. A delay of urgent diagnostics and treatment for communication purposes is impossible without risk of patient’s life.2. Frequent admission of more than one intoxicated patient at one and the same moment. 3. Team style of work, usually organized in 12-hour work schedule. Usually a toxicology patient communicates not only with one treating doctor, but rather with several different treating doctors on duty, as well as several different nurses. Team style of clinical communication requires a perfect coordination and uniform communicative style from medics. 4. Suffi ciency of staff, work-load, intensity of work. 5. Quality and synchrony of communication between health care workers, ambulatory and hospital units. 6. Technical equipment, especially for urgent diagnostics and treatment; suitable conditions for toxicology help, isolation of noise and outer interference; protection of patient’s confi dentiality. 7. Psychological microclimate in the team on duty.

AimIdentifi cation and discussion of the main communication problems in clinical

toxicology and their impact on the informed consent or refusal.

Tasks1. Characterization of the patients with different intoxications from the point of

effective communication and the impact on informed consent or refusal. 2. Outline of the main communicative tasks of the medics in clinical toxicology and

suggestions of some possibilities for adapting the process of informed consent to the specifi city of toxicological patients, with dynamic assessment of the mental status.

Material and methods1. Survey of 1299 hospital fi les of intoxications, treated at the Department of Toxicology,

Naval Hospital -Varna, Military Medical Academy for a 2-years period – 2006-2008. 2. Survey of 905 cases of refusal of hospital admittance in the same department, of

altogether 12661 ambulatory toxicology patients, for a 5-years period, 2006 – 2011. 3. Inquiry of 118 hospital toxicology patients about their remembrance of the signing

the informed consent.

Results 1. 1299 patients with intoxications were treated during the period 2006-2008 year

in Toxicology Clinic, Naval Hospital-Varna, 52% men and 48% - women, from 13 to 92 years old. Etiological distribution: ethyl alcohol - 332 patients (25.56%); methyl alcohol and ethylene glycol – 6 patients (0.46%), medicaments -378 patients (29.10%); narcotics -54 patients (4.16%) ; carbon monoxide and other gases - 52 patients (4%); ‘technical substances’ - 181 patients (13.9%); poisonous animals – 168 patients (12.93%), mushrooms and other plants – 128 patients (9.85%). The total number of patients, admitted in changed consciousness was 598 (46%). 58 (4.46%) patients were with severe delirium. A strong psycho-motor agitation at some stage of intoxication was observed in 111 (8.5%) patients. 80 (6.16%) patients were in coma, 78 (6.0%) – in sopor, 151 (11.6%) – somnolent, 231

Page 213: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

211

(17.8%) – with obnubilation (obtundation). In many cases different pathologic states of the mind were observed in one and the same patient in the course of intoxication.

Figure 1.

The number of patients with changed mental status varied a lot in different etiological groups. Toxic change of the central nervous system had 265 patients, 79.8% of all alcohol cases, 270 patients, 71.4% of all medicament cases, 34 patients, 62.9% of narcotic cases; 7 patients, 13.4% of CO-cases; 16 patients, 8.84% of poisonings with ‘technical’ common household and industrial substances. Very low percentage of changed mental status was established in the groups of mushroom and plant poisonings – 11 patients, 8.59% of them and in the group of animal poisonings – practically no toxic changed mental states. Nevertheless some emotional instability and stress reactions were observed in these groups too.

In 370 cases (29%), the reason for the intoxication was a suicidal attempt, fi rst – in 76%, second – in 16 % and third or more consecutive – in 8%. In 275 cases (21%) the reason was misuse or abuse. 157 patients (12%), had some kind of addiction. Altogether 62% of the intoxications were intentional. 498 patients (38%), had unintentional, accidental poisonings.

150 patients with initial toxic change of the mind restored normal mental status and clear consciousness to the end of the fi rst hour after the admittance, 146 – in 3 hours, 112- in 6 hours, 90 – in 9 hours, 49 – in 12 hours, 41 – in 24 hours, 15 – in 48 hours, 9 – after 72 or more hours. In 8 cases information about co-existing dementia was received after the admittance.

In 466 cases (35.87%), the psychiatric consultation diagnosed co-existing chronic or acute psychiatric disease or disorder.

Page 214: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

212

Table 1. Summary of the written informed consents:

Kind of intoxication Signature of the patient

Signature of the patient’s relatives or legal reprentatives

Signature ofOther person N Signature Total number

All intoxications 730 399 117 53 1299Medicaments 128 190 140 20 378Alcohols 148 111 57 22 338Narcotics 19 21 10 4 54Carbon monoxide 45 4 2 1 52Common household or industrial ‘technical’ poison 154 22 2 3 181Poisonous animals 153 14 1 1 168Mushrooms and other plants 113 13 1 1 128

2. A survey of 905 (7.14%) refusals of hospital treatment from altogether 12661 ambulatory toxicology patients for a 5-years period was done. During the fi rst examination 6651 patients had been assessed as indicated for hospital treatment, so the percentage of refusals from their number was 13.6%. The reason of intoxication was suicidal attempt in 18% and in 1% the patients refused information about it. Etiological distribution was variegated, including 110 alcohol intoxications, 171 medicament intoxications, 57 narcotic intoxications, 35 CO intoxications and etc. Clinical presentation was assessed as severe in 23 cases (3%), moderate in 199 cases (22%) and light in 683 cases (75%). After initial refusal 29 patients (3%) were admitted in Toxicology clinic later, from 2 hours to 3 days after the fi rst examination.

Table 2. Verifying of the informed refusal of hospital admittance

Written refusal by the patientWritten refusal by

relatives or other legal representative

Written refusal by the patient

and the relatives

No signature, the refusal

described by the medical team

No written refusal, no description

Only signature

Hand-written whole sentence+

signature

Only signature

Sentence+signature

Signatures of the team on

duty

106 532 42 13 86 107 19

11,8% 58,8% 4,6% 1,4% 9,5% 11,8% 2,1%

3. An inquiry was made among 118 patients concerning their remembrance of communication with the doctor at the admittance and understanding of the informed consent. 10 patients (8%) refused to participate in the inquiry. As it is a kind of opinion they were not excluded from the analysis. 3 questions were asked in oral conversation on the last day of their hospital treatment:

Page 215: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

213

� Do you remember the content of the offi cial form of informed consent that you/your relatives have signed at the admittance? 26 patients (22%) were satisfactory acquainted with the content; 28 (24%) remembered signing particular document for consent without reading it; 29 (25%) remembered signing ‘a lot of documents’, could not say what is the meaning of informed consent. 25 patients (25%) did not sign informed consent, their relatives or friends did it for them. Only 5 of them had read the informed consent later. � 2. Do you remember the explanation of your admitting doctor about the reasons

why you need hospital treatment? 66 patients (56%) answered: ‘Yes, he/she explained me well.’ 23 patients answered: ‘I do not remember explanations, but I was told that hospital treatment is necessary.’ 12 patients (10%) did not remember anything from the admittance, but later had received explanations. 7 patients (6%) claimed that they had not been told anything at all. � 3. Did you want to be admitted in Toxicology Clinic for treatment? 21 patients

answered with ‘yes’. 55 (47%) patients were hesitating, but had trusted the opinion of the doctor. 18 patients did not want hospitalization at fi rst, but had been persuaded by the doctor. 14 (12%) patients answered that they did not want hospital treatment and had been treated compulsory.

DiscussionThe analysis of the hospital cases shows that from communication viewpoint

toxicological patients are not a uniform group. They highly varied in age, etiology and cause of intoxication, onset, severity and dynamics of toxic changes and especially- of toxic changes of the mind, co morbidity, social and psychological state and etc. These data varied widely in different etiological groups. A high total percentage of initially changed mental status of the patients was established – 46 %. It was very high in the group of medicament intoxications (71.4%), alcohol intoxications (78.4%) and narcotic intoxications (69%), while in the other groups it was much lower. However even in the group of animal envenomations, where it was zero, there were patients with stress, agitation and fear. From 603 patients with initially toxic changes of the mind 170 (28%) restored normal conscience in one hour. In the other 433 cases the restoration of normal mental state was delayed: from 1-2 hours to 72 hours. A signifi cant part of the informed consent forms were signed not by the patients but by their relatives – 399 (30.7%) or, rarely, by other person like friend, neighbor or colleague – 117 (9.0%). 53 (4%) patients left the clinic willfully after restoring clear state of mind without signing the consent. The analysis of the informed refusal of hospital admission also showed a signifi cant problem - 7.14% from the total number of ambulatory patients and 13.6% of those with indication for hospital treatment. 22% of them were estimated as moderate severe and 3% - as severe clinical forms. The suicidal patients (18%), reluctant to treatment had always been a great challenge to clinical toxicologists. The presence of medicament, alcohol or narcotic overdose presumes some level of toxic change of mind even in the lightest cases. Therefore the quality of communication and validity of the informed refusal can be questionable. From 10 years in such cases a written refusal in a sentence with free text and a signature, instead of single signature is preferred in Toxicology Clinic - Varna. Although not legally mandatory, the common sense of this act helps sometimes the patients or relatives to think over the decision and at the same time shows the ability of patient to think and write.

Page 216: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

214

The results of the inquiry are quite contradictive and refl ect some of the communication problems in clinical toxicology. Only 22% of the patients had read and remembered well the content of the informed consent form. 25 patients, 21% had not possibility to give informed consent at the admittance and only 5 of them read it later. On the other side, 56% of the patients considered, that initial information, given by the doctor, was enough and another 23% had trusted to doctor’s opinion without additional information. The other patients did not remember well the admittance, so communication and information exchange had to be delayed. Unlike other clinical disciplines, in clinical toxicology the percentage of hesitating or refusing treatment patients is high, which was confi rmed by the answers of the second and third question. Special attention should be paid to those 12% of the patients who were not persuaded in the benefi t of their hospital management during the whole treatment course as well as to those 8%, who refused to participate in the inquiry possibly as a refl ection of negative attitude. In such cases clinical communication skills of the toxicologist, psychologist and psychiatrist should be combined.

The mental state of the patient is of crucial importance for the effective communication. The perception, processing and returning back of information, taking decision on the base of this process and realizing the consequences from this decision can be disturbed by toxic changes of the conscience. The presence of toxic cerebral syndrome at any stage of the intoxication creates specifi c communicative barriers which lead to total communication impossibility or partial communication diffi culties. As Prof. Monov writes: ’First we should asses the possibility to create a contact with the patient. Between the light obnubilation and the deep coma we fi nd a whole spectrum of symptoms of disturbed contact of the patient with the surrounding world, expressed in changed perception of the reality and changed behavior.’[9]. As duration of cerebral toxic syndrome is different and often hard to predict, it is often hard to establish the fi rst suitable moment for effective communication. The toxic change of mind is not the only reason for communication problems from a patient. Emotional reactions as anger, sorrow, fear or stress can precede the intoxication and express as agitation or stupor.

All the components of the informed consent are specifi cally infl uenced in many toxicological cases in connection to communicative disorders. The most important change lies in the base component – the competency of the patient, but serious problems exist also in the information components - giving information by the doctor and understanding of this information by the patient. As a result obstacles appear in the consent components –the requirement of lack of duress on the patient and the expressing consent on patient’s own free will. A legally valid informed consent should be ‘concrete, voluntary, preliminary and informed’ [12, 13]. Many toxicology patients cannot sign valid informed consent at the admission from legal viewpoint. The same is valid for the informed refusal. The fact that informed consent is not just a one-moment decision and signature, but a process is especially important in clinical toxicology. [13] A special form for dynamic assessment of the mental status and of the right moment of restoring the competency of the patient to sign the informed consent was proposed by the authors with the purpose of individual approach to this problem. [14].

Conclusions:1. Unifi cation of the rules of clinical communication in toxicology is rather diffi cult

Page 217: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

215

because of the great variety of causes, etiology, pathogenesis and clinical presentation of the exogenous intoxications. The toxicological patients are highly varied group: they include practically all ages, sex, intellectual, cultural and social levels, professions and etc. Special attention should be paid to the numerous cases with toxic change of the mind.

2. The high level of emergency and quick clinical dynamics of many toxicological cases requires from the treating doctors and teams specifi c knowledge and skills in communication with toxicological patients. 3. As a result from communication barriers, mainly pathologic from toxic origin, the process of informed consent or informed refusal in toxicological cases is often diffi cult, with individual dynamics.

References:

[1]. Vodenicharov C, S. Popova,Medical Ethics. Sofi a. 2003. p. 55-169.[in Bulgarian][2]. Radanov St. Medical Deontology.Siela. 2005. p. 83-140; p. 162-240. [inBulgarian][3]. Michael Simpson, Robert Buckman, Moira Stewart, et al. Doctor-patient communication: the Toronto

consensus statement. BMJ, VOL 303, 30 NOVEMBER 1991, p.1385-1387[4]. Stoev V. Clinical Communication.Softtrade. 2011. [in Bulgarian][5]. Berlo, D. K. (1960). The process of communication. New York, New York: Holt, Rinehart, & Winston[6]. Daniel Chandler , “The Transmission Model of Communication”, Aber.ac.uk[7]. Barnlund, D. C. (2008). A transactional model of communication. In. C. D. Mortensen (Eds.), Communication

theory (2nd ed., pp47-57). New Brunswick, New Jersey: Transaction. <http://en.wikipedia.org/wiki/Communication

[8]. Lisaev P. Medical Deontologyand Medical Law. Pleven. 2008. [inBulgarian][9]. Monov, Alexander – Clinical Toxicology, Sofi a, Venel OOD, 1995.[10]. Donchev P. Medical Law andDeontology. Medicina i fi zkultura.Sofi a. 1992. p. 7-77; p. 129-142.

[inBulgarian][11]. Law of Health. Prom. SG 70/10Aug 2004. In force from 1st of January2005. [in Bulgarian][12]. Zinovieva D. Medical Law. Sofi a.2004. P. 23-29; p. 151-168; p. 223-231; p.264-269. [in Bulgarian][13]. Dimitrova S. The InformedConsent in Medical Practice.Alfamarket. Stara Zagora. 2003. p. 6-47. [in

Bulgarian][14]. Yovcheva M. Deontological Problems of Clinical Toxicology in Bulgaria, Connected to Communication

Diffi culties, during the Decade 2000-2010. Journal of IMAB- Annual Proceedings (Scientifi c Papers) 2012, vol. 18, book 3, p. 308-310.

[15]. Yovcheva M., Zlateva Sn. Marinov P. Deontological Problems of Clinical Toxicology in Bulgaria during the Decade 2000-2010. Bulgarian medicine vol.II, 2/2012, p. 16-20.

Page 218: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

216

Chapter 28

Diagnostic and Therapeutic Problems of Toxoallergic Reaction after Scolopendra Bite –

10-Years Experience

MarietaYOVCHEVA, Petko MARINOV, Snezha ZLATEVAClinic of Intensive Treatment of Acute Intoxications and Toxoallergy,Military Medical Academy – Sofi a, Naval Hospital – Varna, Bulgaria

Abstract. Scolopendra envenomation is a relatively new toxicological problem in Varna region. The experience of consulting and treatment of 130 cases during the period 2003-2013 year is summarized. 63 patients were admitted in Toxicology clinic (48.5%) and 67 were treated as out-patients (51.5%). 25 of the ambulatory patients refused hospital treatment. A local toxoallergic syndrome was observed in all the cases, with different severity. In 55 patients the local syndrome was combined with a general toxoallergic reaction, usually light and short lasting. 6 atypical and more severe cases were observed. Secondary infection was found in patients who delayed fi rst medical help. The diagnosis is based on the history and the clinical presentation. Diagnostic diffi culties result from the lack of enough information about scolopendra envenomation, self- treatment, delay of medical consultation and help, refusal of hospital treatment and observation, possibility of mistake with poisonous snake bite or Hymenoptera sting. The treatment includes early appliance of corticosteroids, H1-blockers, effective analgesics, anti-tetanus prophylaxis, local compresses, and antibiotics, if necessary. There are signifi cant differences in the therapeutic schemes, volume and duration. Especially great differences exist in the approach to antibiotic treatment of this envenomation. A special ambulatory therapeutic scheme is discussed because of the great percentage of out-patients.

Key words: Scolopendra, toxoallergic reaction, envenomation, treatment.

Introduction Scolopendra bite and envenomation is known all over the world. There are more

than 550 species from the genus Scolopendra, class Chilopoda (centipedes), subphylum Myriapoda, phylum Arthropoda. Scolopendra cingulata Latr. , or Mediterranean scolopendra is typical for Europe. However other, more toxic scolopendras like Sc. Subspinipes or Sc. Heros could be transported accidentally from other continents. [1, 2, 3]. There are few reports of toxic effects of centipede bite in Bulgaria. Some cases were reported in South Bulgaria before 2000 year. [4, 5]. Scolopendra envenomation is a relatively new toxicological problem in North East Bulgaria. However, since 2002-2003 the frequency of these envenomations has increased in Varna region. [5, 6]. S. Cingulata is 10-15 cm

Page 219: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

217

long. It moves fast and often the bitten cannot succeed to see the centipede. Its body has 21-23 segments, each with a pair of legs with sharp claws. Thefi rst pair of legs next to the mouth is called maxillipedes.They are modifi ed into hollow claws connected with poison gland in the base. The bite leaves double puncture oftenwith a small hemorrhage. Some authors prefer to use the term ‘sting’ [3]. These centipedes are active mainly at night and prefer dark and moist places under logs, foliage leaves or stones. Destroying of their habitat or climatic changes (too dry or wet years) force them to seek dark and moist places in human homes. They can be aggressive to man in certain conditions. All scolopendras produce venom, studied by many scientists during the last years. Its composition is still not examined and specifi ed precisely. [1, 3, 7, 8]. Like other animal venoms, it is a complex mixture of different biologically active compounds: enzymes (esterases, acid and alkaline phosphatases, hemolytic phospholipase A2- only in some species, proteases and especially, metalloproteases, hyaluronidases), non-enzymatic proteins ( cardiotoxins, so called toxin S-cardio-depressant factor, disintegrins, haemolysins, myolysins, neurotoxins), other non-peptidic active components – histamine and serotonin, acetylcholine, etc. The venom of different scolopendra species varies in composition. Recent studies show that the centipede venom apparatus differs substantially from that of other arthropods in both functional morphology and venom pharmacology. [3]. There is not a valid routine laboratory test for scolopendra venom. It is generally accepted that a bite by scolopendra is relatively benign for man. DL for mice is 0.01venom secretion per gram body weight. Clinical presentation in humans is dominated by the local toxic allergic syndrome with an instant local burning pain that ranges in intensity from excruciating to moderate and sometimes radiates or spreads to other parts of the victim’s body, double puncture wound, bleeding at fi rst, sometimes with later small central necrosis, localized swelling, and erythema. Complications of the local syndrome were reported: necrosis, paresthesias, great swelling of the whole limb, lymphangitis, lymphadenitis, secondary infl ammation. The general toxic allergic syndrome is usually mild to moderate and is expressed immediately or soon after the bite: transitory weakness, dizziness, sickness, headache, chills, and fever. More serious anaphylactoid reaction with angioedema of distant parts of the body, itching, urticaria and arterial hypotonia is possible. Some rare, but severe complications have been described in literature: myocardial ischemia and infarction, haemoglobinuria and haematuria, rhabdomyolysis, hemorrhage, lethargy, severe anaphylaxis, edema of the larynx, eosinophyllic cellulitis, necrotizing cellulitis and fasciitis. [1, 3, 9, 10, 11, 12, 13]. Patients with allergies and small children are at greater risk of severe clinical forms. [1, 3]. Although it is thought that centipede bite is not fatal for man, several fatalities have been reported. Only three of them were confi rmed as directly connected to scolopendra envenomation [1, 3]. The rest reports were connected to secondary infections commonly associated with these bites or the cause of death was not clear. [3].

As a whole scolopendra envenomations produce transient symptoms that last several days. General symptoms disappear quickly – from 1-4 hours to 24 hours. Local symptoms last from 1-3 days to more than 1 month. Cases complicated with necrosis and secondary infection need management longer than a month. Thus, while generally benign for humans, centipede bite is still able to produce toxic effects and trauma from the wounds. [3].

There are some controversies about the treatment of these bites. Some authors think that non-complicated cases should not be treated as they tend to heal on their own. [9]. However

Page 220: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

218

most of the authors recommend treatment with antihistamines, anti tetanus prophylactics and low dose corticosteroids. There is not specifi c antidote for this envenomation. [1, 3, 14]. Attempts have been made to make specifi c anti-scolopendra serum but they are at early phase. [15]. It is reasonable to differentiate the treatment according to clinical expression and course. A diagnostic and treatment algorithm was suggested by the authors in previous article [6].

AimSurvey of the clinical experience of diagnosis and treatment of scolopendra bites

and envenomation in Varna region for a 10-years period. Summary of the most important problems.

Material and methodsStudy of 63 hospital fi les and 67 ambulatory fi les of 130 patients with toxic allergic

reactions after centipede bite consulted and treated in Toxicology Clinic- Naval Hospital-Varna from 2003 to 2013 year.

Results 130 patients with scolopendra envenomation were consulted and treated in Toxicology

Clinic, Naval Hospital- Varna from 2003 to 2013 year – 87 men and 43 women at the age from 17 to 79 years. 63 of them were hospitalized and the other 67 were ambulatory cases. In the group of ambulatory patients 25, 19.2% patients had refused hospital admission.

Figure 1.

51 of the patients declared co morbidity: arterial hypertension, ischemic disease of the heart, sugar diabetes, and etc. 2 patients had insect-allergy and 10 - non-insect allergy. In 82 (63.08%) cases the patient reported of centipede bite during evening or night hours and in 48 (36.92%) cases - during the day. 54 (76%) patients saw the centipede and either recognized or described it well. In 30 (23.07%) cases the patient did not see the animal that had bitten him. These cases were diagnosed according to the typical history and clinical presentation. Another 15 cases with suspicion of centipede envenomation were excluded from this study, because of very unclear etiology and clinical presentation. The

Page 221: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

219

most common reported circumstances of the envenomation were: work in garden, yard or orchard, moving wood logs to another place, cleaning the cellar or other part of the home, working with manure, sleeping in newly built house.

All 130 patients had a local toxic allergic syndrome. 55 patients, 42.3%, had also some kind of general toxic allergic reaction.

Figure 2.

Local symptoms always included sharp local pain, starting almost immediately after the bite, with burning or piercing character, often irradiating along the bitten limb, lasting from 30-40 minutes to 2-3 days. In 2 cases the pain lasted more than a week- 9 days and 10 days. 22 patients reported that transitory short strong pain in the bitten extremity had appeared during the fi rst 2 -3 weeks, long after the swelling had disappeared. The other local symptoms were burning sensation of the skin or other kind of paresthesia, local swelling with different size – from 3-4 cm to edema of the whole bitten forearm and arm, erythema, double puncture wound, well distiguishable on thin skin, in singular cases –with prolonged bleeding from the wound, small local necrosis.

Table 1. Local toxic symptoms.

Pain EdemaDouble

puncture wound Paresthesia Local necrosis

Local hemorrhage

Secondary infl ammation

130 110 108 98 36 6 21

100% 84.6% 83.07% 75.38% 27.69% 4.61% 16.15%

Extended local reactions were observed in 53 patients, 40.76%. Clinically important local necrosis was seen in 36 cases, usually small-sized. In 4 cases it needed surgical management. In several cases near and more distant skin haemorrhages were observed (Picture 1). Atypical late appearance of long lasting edema and hemorrhages fi rst on the bitten limb and later-symetrically, on the opposite limb, was observed in 2 patients.

Secondary local infl ammation with different severity was observed in 21 patients, 16.15%. (Picture 2). 7 of these patients had also general infl ammatory symptoms.

General toxic syndrome was observed in 55 cases, 42.3%. When questioned thoroughly many patients report some kind of general symptoms. Minor general reactions were expressed with short lasting, about 10-40 minutes, transitory headache, sickness and

Page 222: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

220

weakness. Moderate general reactions were complicated with fever, chills, itching and generalized urticaria, nausea, sometimes vomiting, but without serious hemodynamic instability. Anaphylactoid reactions with clinically signifi cant hemodynamic instability, chest pain, abdominal pain, distant angioedema, were considered for severe general reactions. No ECG data of myocardial ischemia was observed. During the period there were not fatal cases of scolopendra envenomation.

Table 2. General toxic symptoms.

No general reaction

Minor general reaction

Moderate general reaction

Severe general reaction

Minor general reaction +severe local

infl ammationTotal number

75 32 10 6 7 130

57.7% 24.6% 7.7% 4.6% 5.4% 100%

40 patients, 30.7%, did not seek medical help immediately, but used self- treatment and applied different kinds of folk medicines. All the cases of secondary infection were from this group.

5 cases of scolopendra bite were misdiagnosed with snake bite and treated with snake antivenom before the consultation in Toxicology Clinic.

Table 3. Treatment of scolopendra envenomation.

Medicament Treated patients Percentage from the total number of treated patients

Antihistamines-H1-blockers 130 100%Corticosteroids 122 93.84%TAT-tetanus prophylaxis 119 91.54%Analgesics 85 65.38%Antibiotics 76 58.46%Low molecular heparin 20 15.38%Local compresses 102 78.46%Local blockade 2 1.53%Other symptomatic medicines 45 34.61%

. Picture 1 Picture 2

Page 223: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

221

The treatment included H1 blockers, non-opioid analgesics, local compresses, TAT, uplift position of the bitten limb, single or multiple doses of corticosteroids IV. Average doze of Methylprednisolone 40-60 mg or equivalent dose Dexamethasone was prescribed. In case of abnormal local reaction or general anaphylactoid reaction corticosteroids were given in larger dose and for several days- IV and PO. In 8 cases corticosteroids were not prescribed because of late admittance for secondary complications. Antibiotics were not used routinely. Antibiotic was used in 76 cases with prolonged edema, including 21 secondary infections. The following antibiotics were given: Ciprofl oxacin or other chinolones, Clindamycin, Metronidazol, penicillines or cephalosporines, IV, IM or PO. In 20 cases of prolonged edema of the bitten limb low molecular heparin was made in low dose. The treatment with H1-blockers IM and PO and corticosteroids was enough effective for pain-management in 45 cases. The rest 85 were treated with additional analgesics – mainly non-steroid anti infl ammatory medicaments PO or IM or IV. Opiate analgesics were necessary in 25 cases to cope effectively with the pain syndrome.

Discussion:The survey of 130 cases of scolopendra toxic effects confi rms the data of other

authors that the main clinical presentation is a transitory toxic allergic local syndrome, often accompanied by a mild or medium general toxic allergic syndrome [1, 2, 3, 5]. The envenomation as a whole has benign nature in humans, so a lot of cases are never reported to hospitals or physicians. Information about centipedes is quite insuffi cient. Many people in North East Bulgaria have not heard about their toxic effects so after they have been bitten they do not seek immediately medical help. It is one of the reasons for the great percentage of refusals from hospital treatment -19%. As we do not have specifi c laboratory test and the centipede is not brought to be verifi ed by a biologist, the diagnosis relies on the case history and the physical fi ndings. The typical history includes description of a ‘strange, fast moving centipede’ , very painful bite with double puncture wound, sudden strong local pain with irradiation along the limb, local swelling and hyperemia, hyperesthesia, paresthesia, possible regional lymphangitis and lymphadenitis, transitory muscle stiffness, itching, possible small painful necrosis round the bite. Unlike Hymenoptera stings, there is not a standart for ‘normal’ local reaction after centipede bite. We accept that a local swelling over 10 cm in diameter , with or without necrosis and with a tendency to grow bigger after the fi rst hour, is an abnormal, extended reaction [6]. Extended local reaction should not be mistaken with infl ammatory reaction after secondary infection of the wound. General symptoms are not rare, 42%, but transient. The patient should be questioned specially about them. Usually transitional, short lasting general weakness, anxiety, nausea, vomiting, palpitations, fear, oppression, were reported. In some patients the general toxic allergic syndrome disappeared quickly (about 30-60 min) even without treatment. Some of these symptoms perhaps were due to the strong pain and stress and others – real toxic effects. In rare cases serious anaphylactoid reactions occurred. During the period some cases of severe and atypical clinical presentation of the scolopendra envenomation were observed. Two of them were complicated with distributed necrosis on the bitten limb. Another atypical complication was observed in two patients: late and extended local reaction with bullas with hemorrhagic content was observed fi rst in the bitten limb and several days later – in the opposite limb, perhaps as a result of late immune response to scolopendra venom.

Page 224: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

222

Although not so frequent, cases with secondary infection are important. The venom of centipedes itself is considered antiseptic because of some components. The maxillipedes of scolopendra, which make the double puncture, however, are often contaminated with aerobic or anaerobic bacteria. Late seeking for medical help, late and insuffi cient disinfection of the place, self- treatment with strange methods (like cutting the edematous skin with a knife), refusal to take antibiotics are some of the common reasons for severe secondary infl ammation.

Differential diagnosis was made with: A. Snake bite and viperine envenomation. In cases with double puncture when the victim had not seen the animal, anamnesis is important. We suppose a scolopendra bite under the following circumstances: evening and night hours, wet and damp places, human dwellings, especially cellars and yards. In non-confi rmed cases the treatment with anti-snake serum is a diffi cult decision and it is best to admit the patient in hospital for further observation. [4]. B. Bee, wasp or hornet sting. C. Mechanical double puncture wound.

The treatment of centipede bites was symptomatic. The standard treatment was with antihistamines, low dose methylprednisolone ( 40-60 mg) or dexamethasone and tetanus prophylactics. Only in 20 cases of more severe reaction the dose of methylprednisolone exceeded 80 mg the fi rst day and corticosteroid treatment lasted more than 3 days. In 65.38% additional analgesia was necessary with metamizol, paracetamol, non-steroid anti-infl ammatory medicaments, in single cases- with opiate analgesics. Cold local compresses during the fi rst 24 hours and Rivanol compresses next days, uplift position of the limb also relieved the pain syndrome. Unlike other reports, during the period local blockade was made only in 2 cases. Antibiotic treatment was prescribed in 58% of the cases. To our opinion, antibiotic prophylactic is not mandatory in every case of centipede bite. It is necessary when the fi rst medical help and disinfection of the wound had been delayed or when the patient had treated himself with non-sterile devices. Surgical treatment of necrotic zones was made in 4 cases.

The described scolopendra toxic allergic reactions varied in severity, complications, lasting and necessity of hospital treatment. In previous article we have suggested some differentiation of the treatment schemes according to clinical expression and course of the envenomation. During the period the ambulatory cases outnumbered hospital cases – 67(52%) to 63(48%). Patients who had come on time, with minor local reaction without general reaction can be treated as out-patients. A common problem during the period was the refusal of hospital treatment. 25 patients, 19% from all, refused admission in Toxicology Clinic and insisted to be ambulatory patients. The treatment approach in ambulatory cases still needs some unifi cation. Even the minor reactions need at least 2 hours clinical observation. In most out-patients the following scheme was used: disinfection, H1-blocker IM or PO, early single dose of Methylprednisolone 40-60 mg IV or equivalent dose Dexamethasone, TAT, cold local compress fi rst 12-24 hours, prescription of oral antihistamine and in some cases- oral antibiotic, advice of uplift position of the bitten limb and Rivanol compress next days, instruction to observe the bitten place for 10 days. Telephone number for urgent consultation with toxicologist, if necessary, was given.

Page 225: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

223

Conclusions:1. Scolopendra envenomation is a relatively new toxicological problem in Varna

region. 130 cases were treated in Toxicology Clinic –Naval Hospital - Varna, from 2003 to 2013 year. The real frequency of these cases is diffi cult to estimate as many of the bitten by this centipede do not seek medical help. More public information about the toxic effects of scolopendra bite would be useful in the future.

2. The main clinical presentation of centipede envenomation was a well expressed toxic allergic local syndrome in all the patients, dominated by pain and edema around double puncture wound. Although rare, severe local complications are possible, with necrosis, hemorrhage, extended and long lasting edema and secondary infl ammation.

3. 55 patients, 42.3%, had also light or moderate severe general toxic allergic syndrome. Severe anaphylactoid reactions are possible, but during the period they were very rare. There were not lethal cases.

4. Differential diagnosis was made with poison snake bites and Hymenoptera stings.5. The management was symptomatic, with different therapeutic schemes according to

the stage of severity. An algorithm for diagnosis and treatment was suggested for ambulatory patients, who prevailed during the period. Special attention is necessary to those patients, who need hospital treatment, but insist to be treated as ambulatory patients.

6. All the cases of sure or suspect scolopendra bites should be consulted and treated by a doctor. Consultation with a toxicologist is preferable.

References:

[1]. Robert L. Norris, Prof., Dep. Of Surgery, Div. of Emergency medicine, Standford University Medical Center- Centipede Envenomation, E-medicine, Nov 19, 2008, updated Sep 5, 2013..

[2]. Bush SP, King BO, Norris RL, Stockwell SA. Centipede envenomation. Wilderness Environ Med 2001;12:93-9. [PUBMED]

[3]. Eivind A.B. Undheim, Glenn F. King – On the venom system of centipedes (Chilopoda), a neglected group, Toxicon 57, 2011, 512-524.

[4]. Monov, Alexander – Clinical Toxicology, Sofi a, Venel OOD, 1995.[5]. M. Iovcheva, S. Zlateva, P. Marinov, Yu. Sabeva- Toxoallergic reactions after a bite from Myriapoda,

genus Scolopendra in Varna region during the period 2003-2007- Journal of IMAB- AnnualProceeding ( Scient. Papers), 2008, book I, p. 7982.

[6]. M.Yovcheva, Sn. Zlateva - Diagnosis and Treatment of Scolopendra Bites. Toxicological Problems. Third National Congress of Clinical Toxicology with International Participation. Chapter 27. Military Medical Academy, Sofi a, Irita, 2011.

[7]. Rates B.-Prospect of new bioactive molecules from the venom of Scolopendra sp. From Serra de Cipo, Brazil, Symposium of the Pan-American section of International society of Toxinology, Aug., 2004.

[8]. Gutierrez M. del C., Abarea C.-A toxic fracture of scolopendra venom. Comp. Biochem. Physiol., Toxicol.. and Pharmacol., 2003, June,135/ 2, p. 205-214.

[9]. Sean P. Bush, Bradley O. King, Loma Lueta University, USA, - Wildness and Environmental medicine, 2003, vol. 12/2, p. 93-99.

[10]. Ozsarac M, Karcioglu O, Ayrik C, Somuncu F, Gumrukcu S. Acute coronary ischemia following centipede envenomation: Case report and review of the literature. Wilderness Environ Med 2004;15:109-112. [PUBMED]

[11]. Yildiz, S. Biceroglu, Spec. Gazi hospital, Izmir, Turkey,-Acute myocardial infarction in a young man caused by centipede sting. Emerg med J 2006, 23, p. 30.

Page 226: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

224

[12]. Senthilkumaran S, Meenakshisundaram R, Michaels AD, Suresh P, Thirumalaikolundusubramanian P. Acute ST-segment elevation myocardial infarction from a centipede bite. J Cardiovasc Dis Res [serial online] 2011 [cited 2013 Sep 18];2:244-6.

[13]. M. Serinken, B. Erdur, S. Sener, B. Kabay, A.A. Cevik: A Case of Mortal Necrotizing Fasciitis of the Trunk Resulting From a Centipede ( Scolopendra moritans) Bite. The Internet Journal of Emergency Medicine. 2005 Volume 2 Number 2.

[14]. WHO, Weekly epidemiology record, 1.09.2001/ 38, 2001, p.289-300.[15]. Pedro Parrilla-Alvarez, Luis F. Navarrete, Marнa E. Girуn, Irma Aguilar and Alexis Rodrнguez-Acosta-

Use of hen egg derived immunoglobulin against Scolopendra ( Scolopendra gigantea) venom, Evista Cientнfi ca, Rev. Cient. (Maracaibo) vol.18 no.4 Maracaibo Aug. 2008.

Page 227: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

225

Chapter 29

Characteristics of Acute Chemical Poisonings in Ukraine: Morbidity and Mortality

Levchenko Oleg E. - MD, Professor, Colonel of Medical Service, chief of the Toxicology, radiology and medical protection department of Ukrainian Military Medical Academy. Ukraine, 04050, Kyiv, str. Melnikova 24, tel./Fax: +38 (044) 489 16 34. E-mail: [email protected] Natalia V. - PhD, assistant professor of Toxicology, radiology and medical protection department of Ukrainian Military Medical Academy. . Ukraine, 04050, Kyiv, str. Melnikova 24, tel./Fax: +38 (044) 248 10 92, E-mail: [email protected]

Key words: acute poisonings, morbidity, mortality.

IntroductionThe number of acute chemical poisoning in Ukraine increases over the past 10 years.

There is a tendency to increase the number of acute poisoning among children (0 to 18). Today, the overall prevalence of acute poisoning ranges from 15 to 20 cases per 10 thousand populations. Acute poisonings occupy 15-20 % in the structure of all emergency cases that require admission to hospital. The problem of reducing the incidence of acute poisoning, especially among children, is an important task of preventive and clinical toxicology in Ukraine.

AimAnalysis of prevalence and patterns of acute chemical poisoning among the adult and

child population in different regions of Ukraine.

MethodsOffi cial statistical data Center for Health Statistics and the General Bureau of Forensic

Science Ministry of Health of Ukraine were used in the data. Also, the International Classifi cation of Diseases-ICD – 10 was used.

ResultsComparative analysis of acute chemical poisonings among other classes of diseases

found that over the past 10 years, “injury and poisoning” consistently stayed in the fi fth

Page 228: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

226

place in morbidity and in seventh place on the prevalence of all diseases. It should be noted that according to the State Statistics Committee of Ukraine, in 2012, the deaths from acute poisoning took the 5th place among the causes of deaths. The data presented in Figure 1.

Figure. 1. The structure of the main causes of deaths among the population of Ukraine (State Statistics Committee of Ukraine, 2012).

Prevalence in the group of “injury and poisoning” in recent years in Ukraine amounted to 40.4 - 44.7 cases/10000 population, the incidence was between 39.6 - 51.4 cases /10000 population.

Over the past 10 years, the incidence of acute poisoning increased among the group of children 0-14 years, on average, by 10.4 % (from 13.8 ± 0.05 cases/10000 to 15.4 ± 0.1 cases/10000) among children 0-4 years - from 31.7 ± 0.1 to 37.1 ± 0.2 cases/10000 children.

Among children aged 15-17 years, the frequency of acute poisoning was 11.9 cases/10000 population of the age. Among children aged 15-17 years, the severe course of acute poisoning recored 1.8-2.1 times more than among children under the age of 14.

The above demonstrates the high intensity of external risk factors for children and adults on the one hand and on the other hand - a lack of effectiveness of primary prevention. The total number of patients (adults and children) with acute poisoning in some regions of Ukraine is presented in Figure 2.

Analysis of mortality rates, according to Central Bureau of Forensic Science Ministry of Health shows that on average the lethal poisonings of more than 9000 people record in Ukraine each year.

The main causes of fatal accidents are often acute alcohol poisoning (55.6 %) and carbon monoxide - 31.6 %. The common causes of deaths from poisoning are: surrogate alcohol, drugs, medicines and pesticides, - it doesn’t exceed the overall structure of 2%. The structure of main causes of deaths as a result of acute chemical poisoning is shown in Figure 3.

Page 229: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

227

Figure. 2. The total number of patients with acute poisoning (adults aged 18 and older and children ages 0-17) in 2012, in areas of Ukraine

(f.20 addition to t.3220 “Injury, poisoning”)

Figure 3. The structure of main causes of the lethal poisoning (State Statistics Committee of Ukraine, 2012)

In an average year the hospital delivered about 27,000 persons with severe poisoning, around 1.5-2.0% of patients die. But the concern is the fact that over 8,000 people die due to severe poisonings outside the hospitals.

Analysis of cases of acute ethanol poisoning (T51.0) (adults aged 18 and older and children ages 0-17) in regions of Ukraine (f.20 addition to t.3220 “Injury and Poisoning”) showed that on average there are more than 10,000 cases of poisoning with ethanol in hospitals each year.

.

0

500

1000

1500

2000

2500

3000

Auto

nom

ous

Che

rkas

y

Che

rnih

iv

Che

rniv

tsi

Dni

prop

etro

vsk

Don

etsk

Obl

ast

Ivan

o-

Kha

rkiv

Obl

ast

Khe

rson

Khm

elny

tsky

i

Kie

v O

blas

t

Kiro

vohr

ad

Luh

ansk

Lvi

v O

blas

t

Myk

olai

v

Ode

ssa

Obl

ast

Pol

tava

Obl

ast

Riv

ne O

blas

t

Sum

y O

blas

t

Ter

nopi

l Obl

ast

Vin

nyts

ia

Vol

yn O

blas

t

Zak

arpa

ttia

Zap

oriz

hia

Zhy

tom

yr

Kyi

v C

ity

Sev

asto

pol

Administrative territory

Acu

te p

oiso

ning

, cas

es

Adults '18 and older Children 0-17

Page 230: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

228

Forensic anatomical dissections confi rmed 5,000 deaths from acute ethanol intoxication. In this case, only 60-80 people died in hospital, and about 5000 cases – didn’t get to the hospitals and died outside. Thus, in an average year 15,000 people (3.85 cases/10000 populations) suffer cause of the damaging effect of ethanol (confi rmed by court chemical- toxicological studies) in Ukraine.

Acute drugs poisoning (T40) are fi rst presented by opioids, cocaine, cannabinoids and hallucinogens. According to the data, each year 500-600 patients with drug poisoning receive the treatment in the emergency hospitals. Forensic medical research confi rms 150 deaths due to poisoning drugs. 10% of patients die in the hospitals, 90 % of them - outside the hospitals.

Thus, in an average year 700 people (0.18 cases per 10 thousand populations) suffer from the toxic effects of drugs in Ukraine.

Acute poisoning with acids, alkalis, solvents, oxidants (T54) are recorded mainly among the adults. The group of the poisons cauterants presented acetic acid, nitric acid, household chemicals, solvents. According to the data, the total number of patients who were admitted to hospitals with acute poisoning by acids, alkalis, solvents, oxidants, is 500-550 people.

According to the Central Bureau of Forensic Science (Kyiv) each year forensic anatomical dissection are done for 120 people who died after severe poisoning by acids, alkalis, solvents, oxidants. Only 7% of cases are the hospital deaths, the rest are the outside ones.

Thus, in an average year more than 600 people suffer from damaging effects of acids, alkalis, solvents, oxidants in Ukraine (0.16 cases per 10000).

Acute poisoning with heavy metals (T56 - T57) occupy a small percentage of acute chemical poisoning (2 %). The most common cause of heavy metal poisoning is lead. According to the data the total number of patients who were admitted to hospitals after acute heavy metal poisoning is 200-250 people. In an average year there are about 100 autopsies of people who died cause of acute heavy metal poisoning. 5 % of patients died in the hospitals, the rest patients – outside the hospitals. Each year approximately 300-350 people (0.08 cases per 10000) suffer from acute heavy metal poisoning in Ukraine.

Acute mushrooms poisonings (T62.0) are recorded every autumn. The total number of patients suffering from the acute mushrooms poisoning is 550-600 people (0.14 cases per 10000).

According to the Central Bureau of Forensic Science (Kyiv) each year forensic anatomical dissection are done for 55-60 people who died from acute mushrooms poisonings. 30% of patients died in the hospitals, the rest patients – outside the hospitals. The Ministry of Health reported that about 80 species of mushrooms in Ukraine are potentially dangerous for health, and 20 out of them are even dangerous for life. Most of the cases occurred due to consumption of death caps, which are being mistaken for champignons or russules. Edible mushrooms may cause poisonings as well, when they are not correctly processed or when they accumulate different contaminants, especially along the roads. The death cap does not grow in some regions of Ukraine, for example in the south. However, the poisonings occur mostly in the southern region.

Page 231: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

229

ConclusionsThe epidemiological indicators of acute chemical poisoning range from 15 to 20 cases

per 10 thousand of population in Ukraine. The indicator of frequency of acute poisoning averages 8.0 cases per 10 thousand of

population (7.0 cases - in adults and 10.0 cases - in children).The total number of acute poisonings per year exceeds 36,000 cases, lethal poisonings –

9,000 cases. About 500 people die in hospitals ( mortality among hospitalized - 1.5-2.0%), more than 9,000 die outside hospitals ( mortality – 24.0-25.0%). Overall mortality rate from acute chemical poisoning is 24.87% (including patients treated in hospitals and people who died outside hospitals). The main causes of deaths from acute chemical poisoning are: alcohol poisoning (33.14%), carbon monoxide poisoning (17.02%). The total mortality from acute chemical poisoning is 1.55%. Deaths among patients who were treated in hospital due to acute poisoning: acids, alkalis, solvents, oxidants - 4.38%, heavy metals -2.08 %; mushrooms - 4.55%, opioids, cocaine, cannabinoids, hallucinogens - 1.75% ethanol - 0.65%.

In this case, victims of acute poisoning are on average 15-20% of the total number of patients who are hospitalized each year due to occurrence of emergency conditions.

Bibliography

1. ESPAD.2. State Statistic Service of Ukraine. http://www.ukrstat.gov.ua/3. Ministry of Health of Ukraine. http://www.moz.gov.ua/ua/portal/SI “General forensic medical examination Ministry of Health of Ukraine”. http://www.moz.gov.ua/ua/

portal/oth_ecsp.html.

Page 232: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

230

Page 233: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

231

Part 3

DRUG TOXICITY

Page 234: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

232

Page 235: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

233

Chapter 30

Toxicology – Faculty of Pharmacy, Medical University, Sofi a – in the Years

Mitka MITCHEVA 1, Nikolai DANCHEV, Henry ASTROUG, Virginia TZANKOVA, Rumiana SIMEONOVA, Magdalena KONDEVA-BURDINA, Vessela VITCHEVA Laboratory “Drug metabolism and drug toxicity”, Department of Pharmacology,

Pharmacotherapy and Toxicology, Medical University-Sofi a, Faculty of Pharmacy, Sofi a, BULGARIA

Abstract. Some forty fi ve years ago the toxicology lectures have entered the curriculum of the Faculty of Pharmacy with the Medical Academy in Sofi a with the idea that the pharmacy students need a broader knowledge in the medical-biological sciences. The pioneers of these days understood that the alumni in Pharmacy should be familiar not only with the activity, but with the mechanisms of toxic drug effects and with the risks in their use, also, improving their capacity to support the optimising and individualising the proper drug therapy. The present achievement in implementation of these demanding, but noble tasks is a result of the pioneers’ pledging efforts, of their enthusiasm, dedication and responsibility. Since then the toxicology as a discipline in the Faculty had developed and today it is an essential element of the pharmaceutical education. It really interacts with Pharmacology, Pharmacotherapy and other disciplines and structures with the Faculty. The Department of Pharmacology, Pharmacotherapy and Toxicology since the beginning created a favourable, friendly and cosy environment for creating of considerable scientifi c and academic resources in Toxicology. One can distinguish the Toxicology teaching with its tradition and dynamics, with its constant actualizing and harmonizing with vanguard tendencies in the education and the formation of students, master-thesis’ students, specialists and PhD students. The toxicologists in the department have a reach palette of scientifi c studies and publications in the fi elds of characterizing of new molecules, of toxicokinetics, of drug metabolism and drug toxicity, of factors affecting it, of bioactivation and deactivation, of drug abuse, of nanotoxicology etc. The in vivo and the in vitro studies refer the latest trends in the different fi elds of Toxicology and Pharmacy in collaboration with the leading Bulgarian and European study centres. The activities in the fi eld of toxicology contribute to the highest accreditation rating of the Faculty of Pharmacy.

The foundations of modern toxicology teaching in the Faculty of Pharmacy in Sofi a were laid more than forty fi ve years ago.

Tzanko Stoychev,1 coming back from his UK specialisation in Toxicology, as an established and leading scholar in the fi eld, organized the fi rst systemic drug toxicology

1 Tzanko Stoychev (1922 – 1996) was a professor in Toxicology, later Corresponding Member of the Acad-emy of Sciences

Page 236: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

234

course in 1968 with the Faculty. Why this happened in the Faculty of Pharmacy? The medical and social signifi cance of the growing problem of drug toxicity related to self-medication and polypragmasia, on the one hand and on the other hand, the requirement of the modern pharmacists should know the drug related risks – their use and abuse – imposed the need to introduce the toxicology teaching in the Faculty. The faculty management and personally the Dean then - professor Isaev, warmly welcomed Tzanko Stoychev to start the course “Drug Toxicology” in the Department of Organic and Pharmaceutical Chemistry. Its head, professor Damyan Danchev (1921 – 1995) actively and with enthusiasm supported him. Later a call for an assistant professor to organise and conduct the practical studies was appointed.

We should underline the dedication and responsibility of Tzanko Stoychev and Damyan Danchev the two competent Bulgarian scientists and public fi gures, brings together in their efforts to develop successfully this scientifi c fi eld in the Faculty of Pharmacy.

The fortieth anniversary of the independent Department of Pharmacology and Toxicology in the Faculty of Pharmacy was celebrated on the 3rd of July 2013. The fi rst head of the department was one of the greatest Bulgarian pharmacologists professor Dushka Staneva (1921 - 2011). Gradually the department grows and becomes stronger and stronger. Professor Staneva attracted and invited in her team some of the famous Bulgarian medical scientists: professors: Radi Ovcharov, Tzvetan Boyagiev and Yossiv Ilarionov. The department was assisted by three assistant professors, four technical assistants and a zoo-technician, responsible for the vivarium. This is the natural environment for the development of teaching and scientifi c development of drug toxicology.

The years of joint efforts will stay in the memory of collaborators and followers of Academy corresponding member prof. Tzanko Stoychev and prof. Dushka Staneva. This is a period known with its success, creative work and academic atmosphere.

The Toxicology since then grew up. The created Laboratory of Toxicology, Drug Metabolism and Drug Toxicitywas signifi cantly developed.

The Toxicology now is an essential discipline in the education of pharmacy students. It interacts with Pharmacology and Pharmacotherapy as well as with other departments and structures in the Faculty of Pharmacy. The Department of Pharmacology, Pharmacotherapy and Toxicology since the beginning created a favourable, friendly and cosy environment for creating of considerable scientifi c and academic resources in Toxicology. Nowadays, the team involves: professor, two associate professors and three assistant professors. Three of them were ERT.

One can distinguish the Toxicology teaching with its tradition and dynamics, with its constant actualizing and harmonizing with vanguard tendencies in the education and the formation of students, master-thesis’ students and PhD students as well as continuous education and specialization of specialists in toxicology and toxicology analysis and in lthe pharmaceutical industry. The students can follow the optional courses “Drug abuse” and “Drug metabolism and toxicity” yearly. Recently the Faculty Council approved the new program “Cosmetic Toxicology”.

Five of the laboratory members – two associate professors and three assistant professors received their education in the Faculty of Pharmacy. Five PhD students from this laboratory received the educational and scientifi c degree in toxicology. Four of the laboratory members

Page 237: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

235

fi nish their higher education with successful defended Master Thesis of Toxicology. After successful competition they become assistant professors in toxicology.

More than hundred master theses were made in the toxicology laboratory and are connected with more advanced tendency in toxicology. Students’ interest in toxicology is constant. Many of the themes were prepared in close collaboration with departments of Faculty of Pharmacy and other academic and scientifi c units in our country. Some of them of the master theses were made in collaboration with departments in Spain, Italy, France and Belgium according to EU Programs Erasmus and Tempus.

Most of our alumni are employed in teaching, research and service activities within universities, consulting fi rms, governmental agencies, pharmaceutical and chemical fi rms, toxicology laboratories, or other private or public agencies.

Toxicologists from the department actively participated in the publication of numerous textbooks, manuals, reference books etc., independently and in collaboration.

Number of publications: 110. Number of citations: 251 Journals:Method and Findings Experimental Clinical Pharmacology (IF = 0,774)Drug metabolism and pharmacokinetics (IF = 0,474)Bioorganic and Medicinal Chemistry (IF = 2,286)Redox Report (IF = 1,593)European Journal Medicinal Chemistry (IF = 2,187)World Journal of Gastroenterology (IF = 2,081)Pharmaceutical Biology (IF = 0,448)Arhiv za Higijenu Rada I Toksikologiju (IF = 1,048)Farmacia (IF = 0,669)Phytomedicine (IF = 2,972)Phytotherapy Research (IF = 2,086)Biotechnology and Biotechnology Equipment (IF = 0,760)Pharmacognosy Magazine (IF = 1,525)BioMed Research International (IF = 2,880)Toxicology Letters (IF = 3,666) etc...The scientifi c investigations and publications are connected to toxicological researches

of newly perspective molecules, toxicokinetics, drug metabolism and toxicity, the processes of bioactivation and deactivation, oxidative stress, drug abuse, factors infl uencing toxicity as well as nanotoxicology. The scientifi c researches in vitro/ in vivo are connected with the modern tendency in different area in Toxicology and Pharmacy. The investigations are in close collaboration with leading, scientifi c institutes in our country and in Europe.

Many important scientifi c results in the fi eld of drug toxicology are achieved:� Studies on the enzyme mechanisms of biotransformation and the infl uence of

biologically active compounds of natural and synthetic origin. Mechanisms of hepatotoxicity and hepatoprotection.

Page 238: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

236

Newly synthesized and isolated from plants compounds as well as approved drugs have been analyzed for their effect on liver metabolism and toxicity. Experimental models for liver injury have been used too.

In vitro as well as in vivo methods have been used to characterized the metabolizing activity at different levels – subcellular (microsomes and mitochondria), cellular (isolated hepatocytes) and whole animals.

Lots of methods for assessing toxicity have been introduced or modifi ed. For example, a method for isolating hepatocytes aiming increasing of cell viability and reducing the time for isolation and used reagents have been optimized [1].

Biologically active substances (BAS) of synthetic originThe effects of BAS with proven pharmacological effects and presumed hepatic

metabolism have been investigated for their toxicity on isolated hepatocytes.As a result of a screening for hepatotoxicity newly synthesized benzimidazole

derivatives with similar to albendazole anti-helminthic effect were found to have statistically signifi cant lower hepatotoxicity compared to albendazole. The difference in the effects is thought to be due to a difference in the structure and metabolism [2, 3].

Experiments on isolated rat hepatocytes showed that two newly synthesized coumarin derivatives with anticoagulant activity had lower cytotoxicity, compared to that of warafarin. QSAR analysis revealed that one of the compounds had a higher electron density and lower electrophilic activity compared to warfarin. This could explain the weaker reducing effect on glutathione.

The hepatotoxicity of valpromide derivatives were also investigated [4].

Biologically active substances (BAS) of natural originThe effects of BAS of natural origin with presumed antioxidant hepatoprotective

activity were tested on hepatotoxicity models. In conditions of carbon-tetrachloride intoxication (a model of metabolic bioactivation)

benzophenones Hd15, Hd21 and Hd22, isolated from Hypericum annulatum, revealed statistically signifi cant cytoprotective and antioxidant effects, comparable to those of silymarin. These effects were probably due to the infl uence on carbon tetrachloride metabolism [1].

In vitro and in vivo experiments on diosgenin, isolated from Asparagus offi cinalis, revealed that this compound had an antioxidant as well as a cytoprotective effects probably due to diosgenin membrane stabilizing activity (effect found also by Yamaguchi et al. [5]). Western blot analysis revealed that diosgenin affected CYP3A expression in a comparable to phenobarbital extend [6]. A competitive mechanism on a metabolic level could explain diosgenin protective effect on hepatocytes in the experiment with carbon tetrachloride-induced hepatotoxicity.

In conditions of oxidative stress, induced by tert-butyl hydroperoxde, diosgenin revealed higher cytoprotective and antioxidant activity, compared to silymarin in isolated hepatocytes. This suggested that diosgenin could act as scavenger of ROS, formed by tert-butyl hydroperoxide.

Page 239: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

237

In 6-OH-dopamin-induced oxidative stress on isolated synaptosomes accompanied with a damaged mitochondrial function, diosgenin revealed a protective effect, probably due to its antioxidant activity [7].

� Toxicological aspects of clinically signifi cant drug interactions on the level of metabolism. Factors affecting drug metabolism in modeled pathological conditions like hypertension, drug abuse etc.

Toxicological aspects of clinically signifi cant drug interactions on the level of metabolism

The drug metabolism can be a key determinant of drug toxicity. A non-toxic parent drug may be transformed by drug metabolizing enzymes to a toxic metabolites (metabolic bioactivation). Conversely a toxic drug may be transformed to a non-toxic metabolites ( detoxication). The microsomal metabolizing reactions involved cytochrome P450 family of enzymes (CYPs), where a few are responsible for the majority of metabolic reactions, involving drugs. This includes the isoformes CYP 1A2, 2C9, 2C19 (15%), 2D6(20%), 3A4(50%). Many drug interactions are result CYP-enzymes’ inhibition or induction. This why the drug-drug interactions, particularly on the liver level metabolizing system, can result either in the toxicity or in the loss of effi cacy. We performed our scientifi c studies in cellular and subcellular models either in vitro and in vivo. Some macrolides’ administration like Troleandomycine, Erythromycin or Erythromycin derivatives induces cytochrome P450 isoenzyme(s) in rats. The induced isoenzimes’ activity, demethylate and oxidise the drugs into metabolites - probably the nitroso derivatives - which form some stabile, inactive complexes with iron(II) of cytochrome P450. Due to both (induction and inhibition) there is a progressive accumulation of complexed, inactive cytochrome P450 in the liver and the oxidative metabolism of several other drugs may be impaired. This explains the mechanism of adverse drug reactions and also of accidents in Troleandomycine or Erythromycin co-administration with ergotamine, oral contraceptives, carbamazepine, theophylline etc.[10] These effects contrast, absent or are negligible during the josamycine and spiramycin treatment, probably because of their different metabolism.

The hepatotoxic agent Naphthalene undergoes metabolic activation to dyol-epoxides - reactive intermediate metabolites, responsible for a toxic stress in the cell. As a result of our study, based on the investigation of Paracetamol and Naphthalene interaction, we found out that in co-administration of Paracetamol and Naphthalene, Paracetamol decreased the toxicity of Naphthalene. This effect probably due to a competition for one the same cytochrome P450 isoformes.

Multiple co-administration of Grapefruit juice and Paracetamol, led to changes in some parameters, connected with drug metabolism, compared to their alone application. These changes correlate with the observed increased plasma level of Paracetamol.

Effects of biologically active substances (BAS) on the processes of drug metabolism and toxicity in experimental pathological models

Model of essential hypertensionThe studies concerning a description of the metabolic capacity of the liver in biological

Page 240: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

238

models, including in the experimental model of essential hypertension in animals in both sexes has long been the subject of investigations in the laboratory of Drug metabolism and drug toxicity.

Genetically determined hypertension – SH (spontaneous hypertension) proved to be an adequate model for scientifi c researches related to the impact of new promising biologically active substances (BAS) of synthetic and natural origin and of known drugs and other xenobiotics of the drug metabolizing enzyme systems (DMES), drug toxicity and drug-drug interactions in this pathology.

Our laboratory has made comparative assessment of pathophysiological and biochemical parameters in untreated male and female spontaneously hypertensive rats ( SHR) and their normotensive controls (NTR). In female rats (NTR and SHR) we established a lesser amount of CYP 450 as well a lower activity of some monooxygenase systems. We registered a more intensive process of lipid peroxidation too [8].

The experiments revealed that biotransformation processes are less pronounced in both NT and SH female rats, expressed in smaller Cytochrome P450 quantities as well as lower activities of the monooxigenases.

The established higher rate of ethylmorphine demethylation (EM) – a substrate for CYP3A in male hypertensive rats followed by that in male NTR is most likely due to the biotransformation of testosterone, a substrate of the same isoform, which place a role in the pathogenesis in hypertension.

Lower activity of the antioxidant enzymes catalase (CAT), superoxidedismutase (SOD) and glutathione peroxidase was established in male SHR. Enzyme activity correlated with highest values of arterial hypertension.

The infl uence of inducers of drug metabolism like phenobarbital and ethanol has also been a subject of investigation. Phenobarbital exerted its inducing effect on the studied enzymes to a lesser extend in SHR then in NTR, the induction being more pronounced in female rats from both lines. Ethanol induces the enzymes responsible for drug metabolism particularly pronounced in female SHR and NTR rats. The induction is more pronounced in SHR females, compared to NTR.

Ethanol induced the antioxidant enzymes (CAT) and (SOD) in SHR from both sexes compared to NTR. Glutathione and malondialdehyde quantities were not signifi cantly changed.

We found out that cytisine does not affect the parameters of drug biotransformation. Cytisine administration led to an increased brain toxicity only in SHR of both sexes, probably due to increased permeability of the blood-brain barrier, changed blood perfusion and hypoxia, typical for hypertensive animals [9].

Psychoactive substances like amphetamine and cocaine can provoke hypertension, in cases of unknown or untreated hypertension in young patients. This requires study on the impact of these substances on the biotransformation in SHR from both sexes. The parameters of drug metabolism were not affected by amphetamine in female rats from the two lines; whereas in male rats the suppression in NTR and activation of enzyme system in SHR was registered [11].

Cocaine inhibited the activity of the tested enzymes and reduced the amount of

Page 241: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

239

cytochrome P 450 in male NTR, while in all other groups an activation of the process involved in drug metabolism was established.

Amphetamine and Cocaine infl uenced to a lesser extent the amount of GSH and MDA in SHR of both sexes, which correlate in our in vitro studies [12’].

The effects of calcium-channel blockers – nifedipine and nitrendipine induced the studied enzymes of phase I and II of drug metabolism to the same extent in SHR and NTR. The compound L-114 (the newly synthesized 1,4-dihydropyridine derivative), with proven antiarrhythmic effects reduced the amount of cytochrome P450 in both strains of rats and inhibited the enzyme activity of the I phase. This substance induces the enzyme UDPGT, suggesting a different pathway of biotransformation, compared to nifedipine and nitrendipine.

Experimentally determined differences in drug metabolism related to sex and pathological status suggest interaction on a metabolic level which required monitoring in order to optimize the therapy and prevention of the health risk.

Experimental models in vivo/in vitroof cholesterol metabolismAn important research area is the regulation of cholesterol metabolism. The alternative

pathways to eliminate excess cholesterol from the liver cells are studied using in vitro (mitochondrial membranes, isolated hepatocytes, isolated macrophages hepatic cell line HepG2) and in vivo experimental techniques (experimental model of hepatic porphyria). The studies are related to the elucidation of the role of mitochondrial protein, called peripheral benzodiazepine receptor (PBR), located on outer mitochondrial membrane on cholesterol transport and metabolism in the liver cells. The in vivo effects were investigated in normal and in pathological conditions - in a model of hepatic porphyria. The results showed that the binding to PBR is related to cholesterol translocation between mitochondrial membranes and the interaction of ligands with PBRs play a role in the complex mechanism of cholesterol traffi c between mitochondrial membranes in liver cells.

Calcium antagonist nifedipine and nitrendipine are known to inhibit experimentally induced atherosclerosis. Possible antiatherosclerotic mechanisms include alteration of lipid metabolism. The effects of nifedipine and nitrendipine on cholesterol 27-hydroxylation are studied in vivo. A correlation between the affi nity constants of the dihydropyridine derivatives nifedipine and nitrendipine for PBR and increased levels of the metabolite 27 - hydroxycholesterol is found. Dihydropyridine calcium antagonist nifedipine and nitrendipine stimulate 27-hydrohycholesterol production in liver mitochondria in vivo by a mechanism coupled to PBR.

� NanotoxicologyThe development and commercialization of products containing nanoparticles raises

many of the same issues as with introduction of any new technology, including concerns about the toxicity and environmental impact of nanomaterial exposures. In general, with the current technological capabilities, engineering new nanoparticles is a far more rapid task

Exposure to nanoparticles for medical purposes involves intentional contact or administration; therefore, understanding the properties of nanoparticles and their effect on

Page 242: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

240

the body is crucial before clinical use. A new approach in our department are investigational studies on the effects of multiple factors (physiochemical properties, external factors) on toxicity of nanoparticles of different origin (metal nanoparticles, silica and polymeric nanoparticles) in vitro and in vivo.

The in vivo and the in vitro studies refer the latest trends in the different fi elds of Toxicology and Pharmacy in collaboration with the leading Bulgarian and European study centres. The activities in the fi eld of toxicology contribute to the highest accreditation rating of the Faculty of Pharmacy.

References

[1] M. Mitcheva, M. Kondeva, V. Vitcheva., P. Nedialkov, G. Kitanov, Effect of benzophenones from Hypericum annulatum on carbon tetrachloride-induced toxicity in freshly isolated rat hepatocytes, Redox Report11(1) (2006), 1-8.

[2] A. T. Mavrova, K. K. Anichina, D. I. Vuchev, J. A. Tsenov, M. S. Kondeva, M. K. Mitcheva, Synthesis and antitichinellosis activity of some 2-substituted-[1,3]thiazolo[3,2-a]benzimidazol-3(2H)-ones, Bioorg Med Chem13 (2005), 5550-5559.

[3] A. T.Mavrova, K. K. Anichina, D. I. Vuchev, J. A. Tsenov, P. S. Denkova, M. S. Kondeva, M. K. Mitcheva, Antihelminthic activity of some newly synthesized 5(6)-(un)substituted-1H-benzimidazol-2-ylthioacetylpiperazine derivatives, Eur J Med Chem41 (2006), 1412-1420.

[4] M. Kondeva-Burdina, H. Astroug, E. Bechar, V. Vitcheva, I. Valkova, M. Mitcheva, Comparative studies of two new valproic acid derivatives on rat hepatocytes, Pharmacia57(1-4) (2010), 51-55.

[5] A. Yamaguchi, S. Tazuma, H. Ochi, K. Chayama, Choleretic action of Diosgenin is based upon the increases in canicular membrane fl uidity and transporter activity mediating bile acid independent bile fl ow, Hep Res25 (2003), 287-295.

[6] A. Kosters, R. J. J. Frijters, C. Kunne, E. Vink, M. S. Schneiders, F. G. Schaap, C. P.Nibbering, S. B. Patel, A. K. Groen, Diosgenin-induce biliary cholesterol secretion in mice requires Abcg8, Hepatology41 (2005), 141-150.

[7] M. Kondeva-Burdina. In vitro investigation for hepatotoxicity and hepatoprotection of Biologically active compounds from natural and synthesized origin. PhD thesis, 2007.

[8] R. Simeonova, M. Mitcheva, V. Vitcheva, Sexual dimorphism in some drug-metabolizing enzyme activity in SHR vs NTR, Arch Balkan Med Union44(1) (2009), 35-37.

[9] R. Simeonova, V. Vitcheva, M. Mitcheva, Effect of Cytisine on some brain and hepatic biochemical parameters in SHR, Interdisc Toxicol3(1) (2010), 21-25.

[10] Pessayre D, Mitcheva MK, Descatoire V, Cobert B, Wandscheer JC, Level R, Feldmann G, Mansuy D, Benhamou JP. Hypoactivity of cytochrome P-450 after triacetyloleandomycin administration. Biochemical Pharmacology Vol. 30, (1981): 559-564

[11] R. Simeonova, V. Vitcheva, M. Mitcheva, Effects of multiple D-Amphetamine administration on some hepatic biochemical parameters in SHR, In: Medical management of Chemical and Biological Casualties (S. Tonev, K. Kanev, C. Dishovski – Eds.) Sofi a, Publishing House IRITA (2009), 252-259.

[12] V. Vitcheva, M. Kondeva-Burdina, M. Mitcheva, D- Amphetamine toxicity in freshly isolated rat hepatocytes: a possible role of CYP3A, Arh Hig Rada Toksikol60 (2009), 139-145.

Page 243: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

241

Chapter 31

7-Nitroindazole, a Selective Inhibitor of Neuronal Nitric Oxide Synthase ( Nnos)

Attenuates Cocaine and Amphetamine Withdrawal and Brain Toxicity an Rats

Vessela VITCHEVAa,b1, Rumyana SIMEONOVAa, Mitka MITCHEVAa

aLaboratory of Drug Metabolism and Toxicity; department of Pharmacology, Pharmacotherapy and Toxicology; Faculty of Pharmacy,

Medical University – Sofi a, 2, Dunav str., Sofi a-1000b Center for Food Safety and Nutrition (CFSAN), U.S. FDA,

Paint Branch Parkway, College Park, MD

Abstract: The effect of the selective neuronal nitric oxide inhibitor 7- nitroindazole (7-NI) on cocaine and amphetamine withdrawal symptoms and brain parameters such as activity of neuronal nitric oxide synthase ( nNOS), glutathione (GSH) levels and malondialdehyde (MDA) quantity was examined in rats made dependent on cocaine and amphetamine by repeated administration for fi ve consecutive days. Twenty four hours after the last administration behavioral changes were evaluated and the withdrawal symptoms, such as changes in spontaneous locomotor activity, increased level of anxiety, excessive salivation, were observed. Co-administration of 7-NI attenuates these symptoms, which correlates with the biochemical assay of nNOS. The data on nNOS activity showed that both cocaine and amphetamine administered alone signifi cantly increased enzyme activity by 55 % and by 59 %, respectively. Co-administration of 7-NI decreased nNOS activity, induced by the multiple administration of the psychostimulants. Cocaine and amphetamine treatment also resulted in brain toxicity, witnessed by increase in production of MDA and deletion of the cell protector GSH. Our results also showed a benefi cial effect of 7-NI on these parameters. On the basis of these data we can conclude that 7-NI plays an important role not only in ameliorating withdrawal symptoms but also mitigated cocaine and amphetamine-induced brain toxicity.Corresponding Author: Vessela Vitcheva, Center for Food Safety and Nutrition (CFSAN), U.S. FDA, Paint Branch Parkway, College Park, MD USA, e-mail address:[email protected]

Key words: amphetamine, cocaine, 7-nitroindazole, nNOS, toxicity, rats, brain

IntroductionAmphetamine and cocaine belong to the group of psychostimulants that have

been recognized as one of the most signifi cant example of drug abuse due to intense feelings of euphoria, increased concentration and energy, sociability, appetite reduction. Pharmacologically, the psychostimulant effects of cocaine and amphetamine appear to be

Page 244: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

242

mediated by their ability to enhance dopaminergic activity within the mesocorticolimbic circuit through binding to the dopamine, serotonin and noradrenaline transport proteins, and directly prevent the re-uptake of these neurotransmitters into pre-synaptic neurons [1]. Their repeat intake is related to the development of tolerance and dependence accompanied by serious injuries of the central nervous system (CNS). Several studies have indicated that NO is involved in the development of tolerance to opioids [2], psychostimulants [3], ethanol [4] etc. due to its ability to modulate the release of various neurotransmitters such as dopamine, glutamate, and norepinephrine. The mechanisms involved in psychostimulant-induced neurotoxicity have been subject of numerous studies. The role of nitric oxide (NO) and the related NO-N-methyl-D-aspartate (NMDA) cascade in neurotoxicity and the development of tolerance and withdrawal to psychostimulants have also been confi rmed. Activation of NMDA-type glutamate receptor stimulates the release of NO by neuronal NO synthase ( nNOS) [5]. Although NO plays an important physiological role as a neurotransmitter and/or neuromodulator in the CNS, excessive NOS-dependent NO release during high levels of NMDA receptor stimulation result in production of hydroxyl (HO.) and peroxynitrite (ONOO-) radicals that are responsible for oxidative injury [6]. There are a number of studies showing the benefi cial effect of inhibiting nNOS activity as a means of reducing NO-NMDA-induced neurotoxicity and attenuating tolerance and withdrawal to psychoactive agents [4], [7], [8], [3].

Regarding this information, the objective of the following study was to investigate the effect of 7-nitroindazole (7-NI), selective nNOS inhibitor on withdrawal symptoms and neurotoxicity induced by multiple administration of cocaine and amphetamine.

1. Materials and methods1.1. AnimalsMale Wistar rats (body weight 200 g -250 g) were housed in Plexiglas cages (3 per cage)

at 20±2 °C and 12/12-hour light/dark cycle. Food and water were provided ad libitum. The animals were purchased from the National Breeding Centre, Sofi a, Bulgaria. All procedures were approved by the Institutional Animal Care Committee and performed strictly following the principles stated in the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientifi c Purposes (ETS 123) (1991) [9].

1.2. Design of the ExperimentMale Wistar rats were randomized into six experimental groups with six animals in

each (n=6). Group 1 was control animals treated with physiological saline administered i.p. for 5

days.Group 2 was animals treated with 7-NI at a dose of 25 mg/kg bw/day, i.p. for 5 days [10]Group 3 – animals treated with cocaine at a dose of 15 mg/kg bw/day, i.p., 5 days) [11] Group 4 – animals treated with amphetamine at a dose of 5 mg/kg bw/day, i.p., 5 days [12]Group 5 – animals treated with 7-NI in combination with cocaine, administered for 5

days at the respective dosesGroup 6 – animals treated with 7-NI in combination with amphetamine, administered

for 5 days at the respective doses

Page 245: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

243

1.3. Chemicals and reagentsAll the reagents used were of analytical grade. The drugs used in this study, 7-NI,

cocaine and amphetamine, as well as other chemicals, sucrose, Tris, DL-dithiotreitol, phenylmethylsulfonyl fl uoride, potassium phosphate, calcium chloratum (CaCl2), mag-nesium chloratum (MgCl2), L-arginine, L-valine, hemoglobin bovine, beta – Nicotinamide adenine dinucleotide 2`-phosphate reduced tetrasodium salt (NADPH), ethylenediaminetetraacetic acid (EDTA), bovine serum albumin (fraction V) were perchaised from Sigma Chemical Co (TaufkirchenGermany). 2,2’-dinitro-5,5’dithiodibenzoic acid (DTNB) was obtained from MERCK (Darmsstadt, Germany).

1.4. Test for physical dependence in ratsPhysical dependence capacity was recorded 20 hours after the last administration of the

psychostimulants, alone and in combination with 7-NI, using the Observation behavioral test, described by Itzhak [13].

1.5. Assessment of brain biochemical parametersRats were decapitated, the brains were taken out, measured and divided into 3 small

parts – one for measurement of nNOS activity, one – for assessment of MDA quantity and one for GSH levels assessment

1.6. Preparation of brain tissue extracts and assessment of nNOS activityThe brains were minced and homogenized in 10 volumes of buffer, containing 320 mmol

L-1 sucrose, 50 mmol L-1 Tris, 1 mmol L-1 DL-dithiotreitol, 100 μg/L phenylmethylsulfonyl fl uoride (pH=7.2) according to the method used by Knowels and Moncada [5].

The homogenates were then centrifugated at 17 000 x g for 60 min. The protein content was measured by the method of Lowry [14] with bovine serum albumin as a standard. nNOS activity was measured spectrophotometrically using the oxidation of oxyhemoglobin to methemoglobin by NO (18) with slight modifi cations. The incubation medium contained 40 mM potassium phosphate buffer, pH=7.2, 200 μmol L-1 CaCl2, 1 mmol L-1MgCl2, 100 μmol L-1 L-arginine, 50 mmol L-1L-valine, 2.6 μmol L-1 oxyhemoglobin, 100 μmol L-1 NADPH and brain extract. The change in the difference in absorbance at 401 nm and 421 nm was monitored with a double split beam spectrophotometer (Spectro UV-VIS Split), at 37 °С. The activity of the enzyme was expressed in nmol/min/mg, using the milimolar extinction coeffi cient of methemoglobin 77.2 М-1cm-1.

1.7. Preparation of brain homogenate for assessment of malonedialdehyde (MDA)The brains were homogenized with 0.1М phosphate buffer and EDTA, рН = 7.4

(1:10) [15]. Lipid peroxidation was determined by measuring the rate of production of thiobarbituric acid reactive substances (TBARS) (expressed as malondialdehyde (MDA) equivalents) as described by Deby and Goutier[16] with slight modifi cations. Briefl y one volume of the brain homogenate was mixed with 1 mL 25% trichloracetic acid (TCA) and 1 mL 0.67% thiobarbituric acid (TBA). Samples were then mixed thoroughly, heated for 20 min in a boiling water bath, cooled and centrifuged at 4000 rpm for 20 min.The absorbance of supernatant was measured at 535 nm against a blank that contained all the reagents except the tissue homogenate. MDA concentration was calculated using a molar extinction coeffi cient of 1.56 x 105 M-1 cm-1 and expressed in nmol/g wet tissue.

Page 246: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

244

1.8. Preparation of brain homogenate for GSH assessment [15]GSH was assessed by measuring non-protein sulfhydryls after precipitation of proteins

with trichloracetic acid (TCA), using the method described by Bump et al. [25]. Briefl y, Brains were homogenized in 5% TCA (1:10) and centrifugated for 20 min at 4 000 x g. The reaction mixture contained 0.05 mL supernatant, 3 mL 0.05 M phosphate buffer (pH = 8) and 0.02 mL DTNB reagent. The absorbance was determined at 412 nm and the results expressed as nmol/g wet tissue

1.9. Statistical analysisStatistical program ‘MEDCALC’ was used for analysis of the data. The data are

expressed as mean ± SEM of six rats in each group. The signifi cance of the data was assessed using the nonparametric Mann–Whitney U test. Values of P ≤ 0.05 were considered statistically signifi cant.

2. ResultsThe effect of 7-NI on cocaine and amphetamine withdrawal symptoms is shown

on Table 1. Psychostimulants’ withdrawal was manifested by moderate decrease in spontaneous locomotor activity, excessive salivation and enhanced breathing. In the animals experiencing amphetamine deprivation the food consumption was moderately decreased. In the animal groups treated with 7-NI in combination with either psychostimulant the withdrawal symptoms were attenuated.

The changes in brain biochemical parameters are shown in Table 2. Being an inhibitor of nNOS 7-NI administered alone led to a signifi cant decrease in the enzyme activity by 42% (p>0.05). Cocaine and amphetamine multiple administration resulted in statistically signifi cant (p>0.005) increase in nNOS activity by 60% and by 64 % and in MDA production – by 22 % and 27 %, respectively. Both compounds led to GSH depletion by nearly 48 % (p>0.05). 7-Nitroindazole co-administration decreased nNOS activity by 46% and by 60 %, compared to the cocaine and amphetamine group, respectively. It also restored GSH levels in both groups and decreased MDA production in the combined 7-NI+ cocaine group. MDA production was not infl uenced by 7-NI in the animals treated with 7-NI+ amphetamine.

Table 1. Infl uence of 7-NI on cocaine and amphetamine withdrawal measured by the observation test of Itzhak

Behavioral changes Control 7-NI Cocaine 7-NI + Cocaine Amphetamine 7-NI +

AmphetamineDecreased spontaneous

locomotors activity- - ++ - ++ -

Changes in coordination - - - -

Salivation - - +++ + +++ +

Enhanced breathing - - +++ + +++ +

Decreased food consumption

- - - - ++ -

Decreased water consumption

- - - - - -

+++ severe;++ moderate; + slight; - no effect

Page 247: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

245

Table 2. Changes in brain biochemical parameters after multiple administration of 7-NI, cocaine and amphetamine, alone and in combinations

Group nNOSactivity (nmol/ mg) MDAquantity (nmol/g) GSHlevels (nmol/g)

Control 0.604 ± 0.04 3.58±0.30 1.72 ± 0.18

7-NI 0.353 ± 0.06a 3.65 ± 0.16 1.13± 0.05a

Cocaine 0.964 ± 0,12a 4.35 ± 0,32a 0.91 ± 0.05a

7-NI + Cocaine 0.523 ± 0.10b 3.54 ± 0,18b 1.42 ± 0.06ab

Amphetamine 0.992 ± 0.17a 4.54 ± 0.19a 1.03 ± 0.10a

7-NI + Amphetamine 0.393 ± 0.07ac 4.25 ± 0.14a 1.35 ± 0.04ac

Data are expressed as mean ± SEM of six rats. aр< 0.05 vs control, b р < 0.05 vs cocaine, cp>0.05 vs amphetamine (Mann–Whitney U test, P < 0.05)

3. DiscussionIn the present study the infl uence of 7-NI, a selective inhibitor of nNOS on behavioral

and biochemical changes in the brain of rats treated with cocaine and amphetamine, was investigated. Both cocaine and amphetamine are powerful addictive drugs of abuse, causing tolerance and dependence development and withdrawal syndrome. One of the neurobiological mechanisms underlying these processes is thought to be activation of NMDA/NO cascade that results in an increase of nNOS activity and excessive production of NO that plays an important role both as neurotransmitter and neurotoxicant [6]. These changes in the brain are considered responsible for the behavioral effects, caused by abrupt discontinuation of these psychostimulants after repeated administration.

In the present study, 20 hours after the last application of cocaine or amphetamine, the animals showed withdrawal symptoms – decreased spontaneous locomotors activity, salivation, enhanced breathing (see Table 1). On the biochemical level, this corresponds with signifi cantly increased nNOS activity (see table 2). The heterocyclic compound 7-nitroindazole, which inhibits NOS by competing with both L-arginine and tetrahydrobiopterine [17] has been used extensively as selective inhibitor of nNOS. Several studies have indicated that 7-NI affects different physical processes and behaviors, related to drug abuse, such as tolerance, withdrawal, neurotoxicity, psychomotor stimulation and reward [13], [10]. Our results showed that 7-NI, administered along with cocaine or amphetamine, attenuated the withdrawal, which is probably due to the detected decrease in nNOS activity, enhanced by multiple administration of the psychostimulants (Table 2). These data support the behavioral studies carried out by Itzhak [13] and Itzhak and Ali [3] in which the authors proved that administration of 7-NI reduced the hyperactivity and attenuated the induction of behavioral sensitization to cocaine and amphetamines.

In our study we also measured the product of lipid peroxidation MDA and the levels of the cellular protector GSH in brain homogenate. Both amphetamine and cocaine, administered alone, increased the MDA production and deplete GSH levels (see table 2). 7-NI co-administartion restored GSH levels in both groups that could be partly due

Page 248: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

246

to the inhibition of nNOS activity. Production of the lipid peroxidation product MDA in the combined 7-NI+ cocaine group was also decreased compared to cocaine only group. Erdinз et al. [18] in one of their studies reported that 7-NI statistically signifi cant decreased the MDA levels under conditions of cerebral ischemia. Its benefi cial effect on decreasing lipid peroxidation in the brains, taken from cocaine treated rats could be explained by its inhibitory effect on nNOS activity and related decreased formation of hydroxyl and ONOO-

radicals. MDA production, however, was not infl uenced by 7-NI in the animals from the combined 7-NI+ amphetamine group. These results can fi nd their explanation in the data reported by Bashkatova et al. [19]. The authors suggested that the lipid peroxidation caused by amphetamine in rat’s brain is NO-independent.

Under the conditions of this study we could conclude that 7-NI plays an important role not only in ameliorating withdrawal symptoms but also mitigated cocaine and amphetamine-induced brain toxicity.

References

[1] K.F. Foley. Mechanism of action and therapeutic uses of psychostimulants.Clin Lab Sci18 (2005), 107-13.[2] W.M. Lue, M.T. Su, W.B. Lin, P.L. Tao. The role of nitric oxide in the development of morphine tolerance

in rat hippocampal slices. Eur J Pharmacol 383 (1999), 129-35.[3] Y. Itzhak, S.F. Ali. Role of nitrergic system in behavioral and neurotoxic effects of amphetamine analogs.

Pharmacol Ther109 (2006), 246-62.[4] I.T. Uzbay, B.F. Erden, E.E. Tapanygit, S.O. Kayaalp. Nitric oxide synthase inhibition attenuates signs of

ethanol withdrawal in rats. Life Sci 61 (1997), 2197-2209.[5] R. G. Knowles, S Moncada.Nitric oxide synthases in mammals.Biochem J298 ( 1994), 249–258[6] F.X. Guix, I. Uribesalgo, M. Coma, F.J. Mun˜oz. The physiology and pathophysiology of nitric oxide in the

brain.Progress in Neurobiology (2005), 76: 126–152.[7] S.L. Collins, K.M Kantak. Neuronal nitric oxide synthase inhibition decreases cocaine self-administration

behavior in rats. Psychopharmacology159 (2002), 361-9.[8] M.T. Santamarta, I. Ulibarri, J. Pineda. Inhibition of neuronal nitric oxide synthase attenuates the

development of morphine tolerance in rats. Synapse 57 (2005), 38-46.[9] Council of Europe. European Convention for the Protection of Vertebrate Animals used for Experimental and

other Scientifi c Purposes. CETS No. 123, 1991. [displayed 30 May 2007] Available at http://conventions.coe.int/ Treaty/Commun/QueVoulezVous.asp?NT=123&CM=1&CL=ENG

[10] S.F. Ali, Y. Itzhak. Effects of 7-nitroindazole, an NOS inhibitor on methamphetamineinduced dopaminergic and serotonergic neurotoxicity in mice, Ann NY Acad Sci884(1998), 122-130

[11] Y. Itzhak, S.F. Ali, J.L. Martin, M.D. Black, P.L. Huang. Resistance of neuronal nitric oxide synthase-defi cient mice to cocaine-induced locomotor sensitization.Psychopharmacol140 (1998), 378-86.

[12] H.S. Yin, C.T. Chen, T.Y Lin.. Age- and region-dependent alterations in the GABAergic innervation in the brain of rats treated with amphetamine. Int J Neuropsychopharmacol7 (2004), 35-48.

[13] Y.Itzhak. Modifi cation of cocaine- and methamphetamine-induced behavioral sensitization by inhibition of brain nitric oxide synthase.J Pharmacol Exp Ther282 (1997), 521-527.

[14] O.H. LowryProteinmeasurementwiththeFolinphenolreagent. JBiolChem193 ( 1951), 265-75.[15] D. Fau , D. Eugene, A. Berson, P. Letteron, B. Fromenty, C. Fisch, D. Pessayre. Toxicity of the antiandrogen

fl utamide in isolated rat hepatocytes.J Pharmacol Exp Ther269 (1994), 1-9[16] C. Deby, R. Goutier. New perspectives on the biochemistry of superoxide anion and the effi ciency of SOD

Biochem Pharmacol 39 (1990), 399–405.

Page 249: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

247

[17] F.B. Wittwveen, J. Giovanelli, S. Kaufman. Reduction of quinonoid dihydrobiopterin to tetrahydrobiopterin by nitric oxide synthase.J Biol Chem 271(1996), 4143-4147.

[18] L. Erdinз, M. Erdinз, M. Deniz. Effect of 7-nitroindazole on lipid peroxidation induced by cerebral ischemia and reperfusion injury in rats. Dlcle TIP DERGISI (J of medical school) C:27 S:1 (2000), 1-6.

[19] V. Bashkatova, M.M Kraus, A. Vanin, A. Kornick, H. Prast. Comparative effects of NO-synthase inhibitor and NMDA antagonist on generation of amino acids and acetylcholine neurotoxicity.Ann N Y Acad Sci1025 (2004), 221-230.

Page 250: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

248

Chapter 32

Cytotoxic Effects оf Zn/Ag Complex оn Cultured Non-Tumor Cells

Radostina ALEXANDROVAa, Abdulkadir ABUDALLEHa,b, Tanya ZHIVKOVAa, Lora DYAKOVAc, Stoyan SHISHKOVb, Marin ALEXANDROVa,

Gabriela MARINESCUd, Daniela Cristina CULITAd, Luminita PATRONd

aInstitute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences,

Tel. +359 2 979 3678Fax. +359 2 8710 107

E-mail. [email protected] of Biology, Sofi a University “St. Kliment Ohridski”,

cInstitute of Neurobiology, Bulgarian Academy of SciencesdInstitute of Physical Chemistry “Ilie Murgulescu”, Bucharest, Romania

Abstract. It has been found in our previous investigations that Zn/Ag complex (Zn-Ampy-Ag) with Schiff-base ligandexpress signifi cant cytotoxic and cytostatic effects on cultured human and animal tumor cells. The aim of our present study was to evaluate the infl uence of the same compound on viability and proliferation of cultured non-tumor cells. The following permanent cell lines were used as model systems in our experiments: Lep-3 (3-month old human embryo), MDBK (bovine kidney) and BALB/c 3T3 (mouse fi broblasts). The investigations were performed using methods with different cellular targets and mechanisms of action such as thiazolyl blue tetrazolium (MTT) test, neutral red uptake cytotoxicity assay (NR), crystal violet staining (CV), double staining with acridine orange and propidium iodide (AO/PI). The results obtained revealed that applied at a concentration range of 0.5 to 100 μg/ml for 72 h the compound examined decreases signifi cantly viability and proliferation of the treated cells. Key words: zinc, silver, Schiff bases, non-tumor cell lines, cytotoxicity

IntroductionThe Schiff-bases are widely employed as ligands in coordination chemistry [1]. These

ligands are versatile and allow a good control over the stereochemistry of the metallic centers, as well as over the nuclearity of the complexes, by the appropriate selection of the starting materials (carbonyl precursors and primary amines) [2]. All these advantages make Schiff bases suitable candidates in the effort to synthesize metal complexes of interest in bioinorganic chemistry, catalysis, encapsulation, transport and separation processes, etc. [3]. In a recent paper, Marinescu and coworkers showed that the self-assembly process between binuclear [Zn2L1]2+ complex cation and complex anion [Ag(CN)2]- generates

Page 251: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

249

one-dimensional coordination polymer with the following formula: 1∞[{L1Zn2(μ3-OH)}2(H2O){μ-[Ag(CN)2]}](ClO4)3•THF•0.5MeOH (H2L1 is bicompartmental Schiff-base ligand resulting from condensation reaction of 2,6-diformyl-p-cresol with 2-aminomethyl-pyridine) [4]. It has been found in our previous investigations that this complex (Zn-ampy-Ag) expresses signifi cant cytotoxic and cytostatic activities on cultured human and animal tumor cells (data in press). In order to continue research in this area the aim of the present study was to evaluate the effect of Zn-Ampy-Ag on viability and proliferation of cultured non-tumor cells.

1. Materials and Methods1.1. Chemicals and other materialsDulbecco’s modifi ed Eagle’s medium (DMEM) and fetal bovine serum (FBS) were

purchased from Gibco-Invitrogen (UK). Dimethylsulfoxide (DMSO) neutral red, crystal violet, acridine orange, propidium iodide and trypsin was obtained from AppliChem (Germany); thiazolyl blue tetrazolium bromide (MTT) was from Sigma-Aldrich Chemie GmbH (Germany). All other chemicals of the highest purity commercially available were purchased from local agents and distributors. All sterile plastic ware and syringe fi lters were from Orange Scientifi c (Belgium).

1.2. CompoundThe newly synthesized compound 1∞[{L1Zn2(μ3-OH)}2(H2O){μ-[Ag(CN)2]}](ClO

4)3•THF•0.5MeOH (noted as Zn-ampy-Ag) (where H2L1 is bicompartmental Schiff-base ligand resulting from condensation reaction of 2,6-diformyl-p-cresol with 2-aminomethyl-pyridine) was evaluated for biological (cytotoxic, antiproliferative) activity in our study. The synthesis of the compound and its structure are presented in scheme 1.

Scheme 1.THF = tetrahydrofuranMeOH = methanol

OH

CH3

O O

+ N

NH2

-2H2O

THF/MeOHOH

CH3

N N

N N

+ 2 Zn(ClO4)2THF/MeOH

O

CH3

N N

N N

Zn Zn

OH

2+

2 ClO4-2

HL1

+ K[Ag(CN)2]THF / MeOH

1 [{L1Zn2(μ3-OH)}2(H2O){μ-[Ag(CN)2]}](ClO4)3·THF·0.5MeOH8

Page 252: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

250

The compound investigated was dissolved in dimethylsulfoxide and diluted in culture medium. The fi nal concentration of DMSO in the stock solutions (where the concentration of the tested compound was 1 mg/mL) was 2%.

1.3. Cell cultures and cultivationThe following permanent cell lines were used as model systems in our study: Lep-3 (3

month old human embryo),MDBK (bovine kidney) and BALB/c 3T3 (murine fi broblasts). The cell lines were obtained from Cell Culture Collection of IEMPAM-BAS.

The cells were grown as monolayer cultures in DMEM medium, supplemented with 5-10% FBS, 100 U/mL penicillin and 100 g/mL streptomycin. The cultures were maintained at 37 єC in a humidifi ed CO2 incubator (Thermo scientifi c, Hepa class 100). For routine passages adherent cells were detached using a mixture of 0.05% trypsin and 0.02% EDTA. The experiments were performed during the exponential phase of cell growth.

1.4. Cytotoxicity assaysThe cells were seeded in 96-well fl at-botommed microplates at a concentration of 1Ч104

cells/well. After the cells were grown for 24 h to a subconfl uent state (~ 60-70%), the culture medium was removed and changed with medium modifi ed by different concentrations (5, 10, 20, 50, 100 and 200 μg/mL) of the compounds tested of the compounds tested. Each solution was applied into 4 to 6 wells. Samples of cells grown in non-modifi ed medium served as controls. After 24h, 48h and 72h of incubation, the effect of the compounds on cell viability and proliferation was examined by thiazolyl blue tetrazolium bromide (MTT) test, neutral red uptake cytotoxicity assay (NR) and crystal violet (CV) staining.

The MTT colorimetric assay of cell survival was performed as described by [6]. The method consisted of three hours incubation with MTT solution (5 mg MTT in 10 mL DMEM) at 37 єC under 5% carbon dioxide and 95% air, followed by extraction with a mixture of absolute ethanol and DMSO (1:1, vol/vol) to dissolve the blue MTT formazan.

The NR assay was based on the method of [5]. Briefl y, to each well medium containing 50 μg NR/mL (0.1 mL) was added. The plate was placed in the incubator for 3 h for the uptake of the vital dye. Thereafter, the medium with NR was removed and the cells were washed with phosphate-buffered saline (0.2 mL/well), followed by the addition of 0.1 mL 1% acetic acid solution containing 50% ethanol to extract the dye from the cells.

The CV assay was based on the method of [7]. After each well was washed with PBS, the cells were fi xed and stained with 0.4% crystal violet solution in methanol for 30 min.

Optical density was measured at 540 nm (MTT, NR, CV) using an automatic microplate reader (TECAN, SunriseTM, Austria). Relative cell viability, expressed as a percentage of the untreated control (100% viability), was calculated for each concentration. “Concentration –response” curves were prepared and the effective concentrations of the compounds - CC50 and/or CC90 (causing a 50% and 90% reduction of cell viability, respectively) were estimated (where possible) from these curves using Origin 6.1TM. All data points represent an average of three independent assays.

1.5. Double staining with acridine orange (AO) and propidium iodide (PI)The cells were grown on sterile cover slips in 6-well plates in the presence of the

compounds tested. Non-treated cells served as controls. After 24, 48 and 72h of incubation, the coverslips were removed and the ability of the compounds to induce cytopathological

Page 253: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

251

changes was assessed using double staining with acridine orange (AO) and propidium iodide (PI) according to the standard procedures [14].

The coverslips were washed with PBS for 2 min. Equal volumes of fl uorescent dyes containing AO (10 μg/mL inphosphate-buffered saline, PBS) and PI (10 μg/mL in water) were added to the cells. Fresh stained cells were placed on a glass slide and examined under fl uorescence microscope (Leika DM 500B, Wetzlar, Germany) within 30 mun before the fl uorescent color started to fade.

1.6. Statistical analysisThe data are presented as mean ±standard error of the mean. Statistical differences

between control and treated groups were assessed using one-way analysis of variance (ANOVA) followed by Dunnett post-hoc test and Origin 6.1TM .

2. ResultsThe results obtained by us reveal that:1. Applied at concentrations of 0.5, 1, 10, 20, 50, 100 μg/ml for 72 h the compound

investigated decreases signifi cantly viability and proliferation of the treated cells (Fig 1, Table 1), cytopathological changes are also found (Fig. 2);

2. The cell-specifi c response has been observed (Fig. 1, Table 1) – the Lep-3 human embryonic cells show higher sensitivity to cytotoxic/cytostatic activity of the tested complex as compared to bovine kidney cells and murine fi broblasts;

3. A positive correlation between the data obtained by assays with different cellular targets and nechanism(s) of action was observed (Fig. 1, Table 1);

4. Tested independently, the same amount of DMSO as those in the solutions of the compounds tested, has no signifi cant cytotoxic effect as compared to the control – the cell viability was > 94%.

Table 1. Cytotoxicity (CC50 and CC90, μg/ml) of Zn-ampy-Ag against human (Lep-3), bovine (MDBK) and murine (BALB/c 3T3)

Cell line Lep-3 BALB/c 3T3 MDBK

Assay MTT NR CV MTT NR MTT

Period 72h 72h 72h 72h 72h 72h

CC50 13.50 25.30 27.00 35.61 32.75 49.00

CC90 19.70 45.00 45.30 48.07 n.d. 57.27

CC50 (CC90) – concentrations (μg/ml), that reduce cell viability by 50% (90%); n.d. - CC90 was not determined because at all concentrations administered the cell viability was > 10%.; MTT = thiazolyl blue tetrazolium bromide (MTT) test, NR = neutral red uptake cytotoxicity assay; CVS = crystal violet staining

Page 254: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

252

Fig.1. Effect of Zn-ampy-Ag on viability and proliferation of Lep-3(A) and BALB/c 3T3 (B). Thecompound was applied at concentrations of 0.5, 1, 10, 20, 50, 100 μg/ml. (A) The investigations were carried out by thiazolyl blue tetrazolium bromide (MTT) test, neutral red uptake cytotoxicity assay (NR)

and crystal violet staining (CVS) on 72 h. (B) The evaluation was performed by thiazolyl blue tetrazolium bromide (MTT) test and neutral red uptake cytotoxicity assay (NR) on 72 h.

Fig.2. Fluorescence micrographs of AO and PI double-stained BALB/c 3T3 cells untreated (a) or incubated for 72 with Zn-ampy-Ag (0.5 μg/ml) (b) and Zn-ampy-Ag (20 μg/ml) (c).

Moderate (b) and severe loses of cells (c) following the treatment.Bar = 50 μm.

3. DiscussionThe development of anticancer drugs based on metals is a very active fi eld [11], [12].

On the other hand, biological activities of zinc (essential element required for normal functioning of 300 mammalian enzymes) and silver (especially its antimicrobial activity) are very attractive and it is not surprising that compounds containing both metals deserve special interest [8], [10], [13]. It has been found in our previous investigations that Zn-ampy-Ag exhibits promising anticancer properties in cultured human and animal tumor cells (unpublished data). The study presented here reveals that the same compound decreases signifi cantly viability and proliferation of the treated human (Lep-3), bovine (MDBK) and murine (BALB/c 3T3) non-tumor cells. Additional investigations are needed to clarify better the putative anticancer activity and toxicity of Zn-ampy-Ag. It has to be mentioned here that the construction and application of delivery systems (liposomes and other nanoparticles) make it possible to reduce the side effects of the drugs encapsulated even when applied at toxic doses [9].

Acknowledgement: Supported by Grant BG051PO001 – 3.3.06 -0048/ 04.10.2012 and a bilateral project between Bulgarian Academy of Sciences and Romanian Academy.

1.A

0 25 50 75 100 1250

50

100

150ControlMTTNRCV

Concentration μg/ml

Cel

l via

bilit

y,%

of t

heC

ontr

ol

1. B

0 25 50 75 100 1250

50

100

150ControlMTTNR

Concentration μg/ml

Cel

l via

bilit

y, %

of t

heC

ontr

ol

Page 255: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

253

References

[1] R. Hernandez-Molina, A. Mederos, ComprehensiveCoordinationChemistryII, Elsevier, Amsterdam, 2004, 411.

[2] M. Andruh, F. Tuna, M.A. Cato (Ed.), FocusonOrganometallicChemistryResearch, NovaPublishers, Hauppauge, 2005,144.

[3] P.A. Vigato, S. Tamburini, L. Bertolo, Coord.Chem. Rev. 251(2007), 1311.[4] G. Marinescu, A.M. Madalan, C. Tiseanu, M. Andruh, Polyhedron 30 (2011) 1070.[5] E. Borenfreund, J. Puerner, Toxicity determination in vitro by morphological lterations and neural red

absorption, Toxicol. Lett.24 (1985), 119-124.[6] T. Mosmann, Rapid colorimetric assays for cellular growth and survival: application to proliferation and

cytotoxicity assays, J. Immunol. Meth.65 (1983), 55-63.[7] K. Saotome, H. Morita, M. Umeda, Cytotoxicity test with simplifi ed crystal violet staining method using

microtitre plates and its application to injection drugs,Toxicol in Vitro3 (1989), 317-21.[8] H. Tapiero, K.D. Tew , Trace elements in human physiology and pathology: zinc and metallothioneins,

Biomed Pharmacother.57(9), (2003), 399-411. [9] N.P. Barry, P.J. Sadler , Challenges for metals in medicine: how nanotechnology may help to shape the

future, ACS Nano.7(7) (2013), 5654-9.[10] S. Chernousova, M. Epple, Silver as antibacterial agent: ion, nanoparticle, and metal, Angew Chem Int Ed

Engl.52(6) (2013), 1636-53[11] B. Desoize, Metals and metal compounds in cancer treatment, Anticancer Res. 24 (2004), 3-18.[12] D. Chen, V. Milacic , M. Frezza,Q.P. Dou, Metal complexes, their cellular targets and potential for cancer

therapy, Curr Pharm Des.15(7) (2009), 777-91.[13] R. Alexandrova, G. Rashkova, D. Salkova, I. Sainova, Something more about zinc, Exp. Pathol.

Parasitol.5(8) (2002), 17-24.[14] A.S.I. Wahab, A.B. Abdul, A.S. Alzubairi, M.M. Elhassan, S. Mohan, In vitro Ultramorphological

Assessment of Apoptosis by Zerumbone on (Hela), J.Biomed Biotechnol. (2009), 46-55.

Page 256: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

254

Chapter 33

Effects of D- amphetamine on brain and hepatocytes isolated from male and female spontaneously

hypertensive rats ( SHR)

Rumyana SIMEONOVA1, Magdalena KONDEVA-BURDINA, Vessela VITCHEVA, Mitka MITCHEVA

Laboratory of drug metabolism and drug toxicity, Department of pharmacology, pharmacotherapy and toxicology, Faculty of pharmacy, MedicalUniversity – Sofi a

Abstract. D-Amphetamine is widely used drug of abuse, known to cause enhancement in blood pressure, neurotoxicity and hepatotoxicity, both in humans and rats.Spontaneously hypertensive rats ( SHR) are generally used for studies in cardiovascular research. It is well known that SHR are more sensitive to liver injury, provoked by some toxic compounds than their normotensive controls. Regarding this information, the aim of the following study was to investigate D- amphetamine brain toxicity and hepatotoxicity in isolated hepatocytes from SHR, compared to normotensive Wistar-Kyoto rats (WKY). In the fi rst series of our experiments male and female SHRs and WKY rats were treated with D- amphetamine (5mg/kg bw/day, 5 days) and brain toxicity of the compound was assessed by measuring nNOS activity, MDA quantity and GSH levels. Neuronal NOS activity was statistically signifi cant increase in the brains taken from WKY rats but not in SHRs. MDA quantity increased and GSH levels depleted in a similar way in both strains. Liver toxicity of D- amphetamine was investigated in vitro in isolated rat hepatocytes, incubated with 100 microM of the compound. D- amphetamine hepatotoxicity judged by decrease in cell viability and GSH depletion was more pronounced in SHRs. However, MDA production was less increased when compared to WKY rats. On the basis of this study we might suggest that the observed differences are probably due to pathophysiology of SHR and it was speculated that this strain possess some compensatory mechanisms to hepatoprotection and neuroprotection.

Key words. D- amphetamine, SHR, brain toxicity, hepatocytes

IntroductionAmphetamine is a powerful stimulant of the central nervous system (CNS), leading to

the development of physical and psychological dependence. As indirect sympathomimetic it causes both central and peripheral effects through the release of noradrenaline, serotonin and

1 Corresponding Author: Rumyana SIMEONOVA, Laboratory of drug metabolism and drug toxicity, Department of pharmacology, pharmacotherapy and toxicology, Faculty of pharmacy, Medical University, 2, Dunav str. Sofi a, Bulgaria; E-mail: [email protected]

Page 257: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

255

dopamine frompresynaptic terminals.Sympathetic excitation leads to rapid and sometimes irregular heartbeat, sweating, dilation of the pupils, hypertension, and hyperthermia.

Along with the potential for the development of tolerance and dependence, the repeated intake of amphetamines led to cardiovascular toxicity [1], hepatic toxicity [2, 3] and neurotoxicity [4].

The main hepatotoxic mechanism of amphetamine is its liver bioactivation to toxic reactive metabolites [5]. Neurotoxic effects of amphetamine are due to increased production of NO and reactive oxygen species, triggering processes of lipid peroxidation [4].

D- amphetamine is well known substance whichcauses cardiac problems [1] and an elevation of blood pressure.On the other hand essential hypertension is associated with oxidative stress, lipidand protein damage [6] which leads to development of some complications, such ascardiovascular disease, arteriosclerosis and fatty liver.

As for all pathological conditions, the use of animal models is of a great importancefor the study ofpathophysiological changes of hypertension, including the effects of differenttoxic substances on this state.The spontaneously hypertensive rats ( SHR) are a model of genetic hypertension, sharing common characteristics with primary hypertension in man.

On the other hand Sagvolden [7] conceders SHR as the best validated model of cognitive-behavioral disorder associated with attention defi cit hyperactivity disorder (Attention-Defi cit/Hyperactivity Disorder - ADHD). First choices of treatment of this pathology are amphetamine–like drugs.

Some researchers found sex-determined differences in the behavior and toxicity of male and female rats [8, 9].To achieve equivalent brain concentrations in male and female rats it is necessary to apply higher doses of amphetamine in male subjects [10] because of their accelerated hepatic metabolism.

Gender differences were observed even when the application of amphetamine is dosed, to cause the same concentrations in the brain in male and female rats [11].

On the basis of this information we uses male and female SHRs to assess amphetamine brain toxicity after multiple administration and its toxicity in isolated rat hepatocytes. The effects were compared with the normotensive control rats from WKY strain.

1. Materials and Methods1.1. Experimental AnimalsMale and female SHRs strain Okamoto-Aoki and their age-matched normotensive

controls Wistar-Kyoto rats were obtained from Charles River breeding center in Germany. The rats were housed in plexiglass cages (3 per cage) in a 12/12 light/dark cycle, under standard laboratory conditions (ambient temperature 20°C ± 2°C and humidity 72% ± 4%) with free access to water and standard pelleted rat food 53-3, produced according ISO 9001:2008. A minimum of 7 days acclimatization was allowed before the commencement of the study and their health was monitored regularly by a veterinary physician. All performed procedures were approved by the Institutional Animal Care Committee and the principles stated in the European Convention for the Protection of Vertebrate Animals

Page 258: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

256

used for Experimental and other Scientifi c Purposes (ETS 123) [12] were strictly followed throughout the experiment. The number of rats wasreduced, as much as possible, depending on statistical signifi cance.

Blood pressure was measured in conscious animals using a semi-automated tail-cuff device (LE5002, Letica S/A, Spain). Before the experimental period, the rats were conditioned to the restraining cylinders. Rats were pre-warmed for 10 min using a temperature – controlled warming holder (37oC) to facilitate tail blood fl ow before their blood pressure was measured. The mean of tree tail-cuff readings was used as the systolic and diastolic blood pressure value.

1.2. ChemicalsAll the reagents used were of analytical grade. Pentobarbital sodium (Sanofi , France),

KH2PO4 (Scharlau Chemie SA, Spain), MgSO4.7H2O (Fluka AG, Germany), trichloroacetic acid (TCA) (Valerus, Bulgaria), lactate-dehydrogenase (LDH) kit (Randox, UK). HEPES, collagenase from Clostridium histolyticum type I V, albumin, bovine serum fraction V, minimum 98 %, EGTA, 2-thiobarbituric acid (4,6-dihydroxypyrimidine-2-thiol; TBA) were purchased from Sigma Aldrich, (Germany). Sucrose, Tris, DL-dithiotreitol, D-glucose, phenylmethylsulfonyl fl uoride, potassium phosphate, calcium chloratum (CaCl2), magnesium chloratum (MgCl2), L-arginine, L-valine, hemoglobin bovine, beta –Nicotinamide adenine dinucleotide 2`-phosphate reduced tetrasodium salt (NADPH), ethylenediaminetetraacetic acid (EDTA), 2,2’-dinitro-5,5’dithiodibenzoic acid (DTNB) were obtained from MERCK (Germany).

1.3. Isolation and incubation of hepatocytesRats were anesthetized with 0.24 M sodium pentobarbital (0.2 mL/100 g). In situ liver

perfusion and cell isolation were performed as described by Fau et al. [13]. Cells were counted under the microscope and cell viability was assessed by Trypane blue exclusion (0.05%).

In order to choose appropriate amphetamineconcentration for the in vitro experiments, hepatocytes were incubated with D-amphetamineat four concentrations (50 μmol/ L, 100 μmol/L, 150 μmol/Land 200 μmol/L) for one hour.Average toxic amphetamine concentration (TC50) was determined to be 100 μmol/L. Hepatocytes isolated fromthe animals of all groups were incubated withD- amphetamine at TC50 for one hour. Incubations were performed in a 5 % CO2 +95 % O2 atmosphere. For all experimental groups, non-treatedhepatocytes were used as controls.

The following parameters were measured to assess the functional status of hepatocytes: cell viability, lactate dehydrogenase (LDH) activity, reduced glutathione (GSH) levels and malonedialdehyde (MDA) quantity. Cell viability was assessed by Trypane blue exclusion method [13]. The dye was used at a fi nal concentration of 0.05 % and cells were counted under light microscope (x 100). At the end of incubation, the cells were recovered via centrifugation at 400 x g at 4˚C. The supernatant was used for LDH [14] and MDA [13] assessment. The level of GSH was measured in the sediment [13].

Page 259: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

257

1.4. Experimental design for brain toxicity assessmentThe normotensive and hypertensive rats were divided in 8 experimental groups. Group 1: control male SHRGroup 2: male SHR treated with D- amphetamine (5mg/kg bw/day, 5 days)Group 3: control female SHRGroup 4: female SHR treated with D- amphetamine (5mg/kg bw/day, 5 days)Group 5: control male WKY ratsGroup 6: male WKY treated with D- amphetamine (5mg/kg bw/day, 5 days)Group 7: control female WKY ratsGroup 8: female WKY treated with D- amphetamine (5mg/kg bw/day, 5 days)

1.5. Preparation of brain tissue extracts and assessment of nNOS activityRats were decapitated and the brains were minced and homogenized in 10 volumes of

buffer, containing 320 mmol L-1 sucrose, 50 mmol L-1 Tris, 1 mmol L-1 DL-dithiotreitol, 100 μg/Lphenylmethylsulfonylfl uoride (pH=7.2) [15].The homogenates were then centrifugated at 17 000 x g for 60 min. The protein content was measured by the method of Lowry [16] with bovine serum albumin as a standard.

Neuronal NOS activity was measured spectrophotometrically using the oxidation of oxyhemoglobin to methemoglobin by NO [16]. The incubation medium contained 40 mM potassium phosphate buffer, pH=7.2, 200 μmol L-1 CaCl2, 1 mmol L-1MgCl2, 100 μmol L-1 L-arginine, 50 mmol L-1L-valine, 2.6 μmol L-1 oxyhemoglobin, 100 μmol L-1 NADPH and brain extract.The change in the difference in absorbance at 401 nm and 421 nm was monitored with a double split beam spectrophotometer (Spectro UV-VIS Split), at 37 °С. The activity of the enzyme was expressed in nmol/min/mg, using the milimolar extinction coeffi cient of methemoglobin 77.2 М-1cm-1.

1.6. Lipid peroxidation in brain homogenateLipid peroxidation was determined by measuring the rate of production of thiobarbituric

acid reactive substances (TBARS) (expressed as malondialdehyde equivalents) [17]. One volume of brain homogenate was mixed with one volume 25 % trichloracetic acid (TCA) and one volume 0.67 % thiobarbituric acid (TBA). Samples were then mixed thoroughly, heated for 20 min in a boiling water bath, cooled and centrifuged at 4000 rpm for 20 min. The absorbance of supernatant was measured at 535 nm against a blank that contained all the reagents except the tissue homogenate. Malondialdehyde concentration was calculated using a molar extinction coeffi cient of 1.56 x 105 M-1 cm-1 and expressed in nmol/g wet tissue.

1.7. GSH assessment in brain homogenate GSH was assessed by measuring non-protein sulfhydryls after precipitation of proteins

with TCA [18]. Brain tissues were homogenized in 5 % trichloracetic acid (TCA) and centrifugated for 20 min at 4 000 x g. The reaction mixture contained 0.05 mL supernatant, 3 mL 0.05 M phosphate buffer (pH = 8) and 0.02 mL DTNB reagent. The absorbance was determined at 412 nm and the results expressed as nmol/g wet tissue.

Page 260: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

258

1.8. Statistical analysisStatistical analysis was performed using the statistical software ‘MEDCALC’. For

the in vitro experiments the results are expressed as mean ± SEM of four animals per group and for each of the examined parameters, three parallel samples were used. For the in vivo experiments the data are expressed as mean ± SEM of six rats in each group.The signifi cance of the data was assessed using the nonparametric Mann–Whitney U test. Values of P ≤ 0.05, P ≤ 0.01 and P ≤ 0.001 were considered statistically signifi cant.

3. Results3.1. In vitro experiments in isolated hepatocytesTable 1.Effect of amphetamine (100 μM) on cell viability, GSH level, amount of MDA

and LDH leakage in hepatocytes isolated from male and female SHR, compared with male and female WKY rats.

Parameters Groups Male WKY Female WKY Male SHR Female

SHRCell viability%

Control 79% ± 3,3 76% ± 4,2 80% ± 2,1 84% ± 3,5Amphetamine 100 μМ 31% ± 5,8* 40% ± 3,6# 52% ± 1,1+ 60% ± 2,7&

GSH nmol/106 Control 28,1 ± 2,1 22,6 ± 3,1 22,2 ± 3,1 24,1 ± 4,4Amphetamine 100 μМ 6,2±1,5* 8,6 ± 2,9# 20,4±2,1 21,6 ± 3,2

MDAnmol/106

Control 0,203±0,05 0,173± 0,09 0,353±0,03* 0,304 ± 0,07#

Amphetamine 100 μМ 0,387±0,04* 0,369± 0,06#

0,374±0,04 0,327 ± 0,06

LDH μmol/106/min

Control 0,117 ± 0,06 0,124 ± 0,07

0,195 ± 0,09 0,189 ± 0,02

Amphetamine 100 μМ 0,186 ± 0,03* 0,163 ± 0,04#

0,242 ± 0,05+ 0,218 ± 0,07

*p < 0,05 vs male WKY; +p < 0,05 vs male SHR; #p < 0,05 vs female WKY; &p < 0,05 vs female SHR

Incubation of isolated hepatocytes with 100 μM D- amphetamine leads to the following changes:

Cell viability – statistically signifi cant (p < 0.05) decreased by 35 % and by 29 % in hepatocytes isolated from male and female SHR, respectively and by 61 % and by 47 % in hepatocytes from male and female WKY.

GSH levels were decreased as follows: in male WKY by 78% (p < 0.05), in female WKY by 62% (p < 0.05), in male SHR by 9% and in female SHR by 10%.

MDA quantity was increased statistically signifi cant (p < 0.05) by 91 % and by 113 % only in hepatocytes isolated from WKY – male and female rats, respectively. LDH leakage was increased by 59 % (p < 0.05) in male WKY, by 31 % (p < 0.05) in female WKY, by 24% in male SHR (p < 0.05) and by 15% in female SHR.

3.2. In vivo experiments in brain from male and female SHR and WKY rats

Table 2.Effect of D- amphetamine (5mg/ kg bw/ day, 5 days, i.p. administration)

Page 261: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

259

on GSH level, MDA quantity and nNOS activity in brain from male and female SHR, compared with respective genders of WKY rats.

Parameters Groups Male WKY Female WKY Male SHR Female SHR

GSH nmol/g Control 1,58±0,03 1,28±0,07* 0,98±0,05* 1,27±0,02+Amphetamine 1,03±0,04* 0,93±0,02# 0,83±0,02 0,99±0,03&

MDAnmol/g Control 3,52 ±0,05 5,08±0,07* 4,76 ±0,04* 5,26±0,09Amphetamine 4,53±0,08* 6,93±0,07 5,71±0,13+ 6,32±0,04&

nNOS nmol/mg/min Control 0,608± 0,009 0,656 ± 0,012 0,762 ± 0,011* 0,861 ± 0,008#

Amphetamine 1,281± 0,012* 1,216± 0,011# 0,840± 0,014 0,971± 0,007

*p < 0.05 vs male WKY; +p < 0.05 vs male SHR; #p < 0.05 vs female WKY; &p < 0.05 vs female SHR Amphetaminestatistically signifi cant (p < 0.05) reduced GSH levels as follows: in the

brain of WKY by 35% in male and by 27% in female rats; in SHR - by 15% and by 22% in male and female rats, respectively.The amount of brain MDA increased after repeated administration of amphetamine as follows: in male WKY by 29% (p < 0.05), in female WKY by 36%, in male and female SHR by 20%. The activity of brain nNOS increased in male WKY by 111% (p < 0,05), in female WKY, by 85% (p < 0,05), in male SHR by 10% and in female SHR by 13%.

4. DiscussionAbuse with amphetamines affects both men and women. Gender differences were

observed at all levels: beginning, development, onset of dependence, withdrawal symptoms, relapse and biochemical level. In recent years there has been an increase in the number of women abusing psychoactive stimulants [11].

Repeated administration of D- amphetamine is accompanied by pronounced hepatotoxicity in humans [19, 20], as well as in experimental animals [21, 5].

The studies about hepatocellular damage and brain toxicity of D- amphetamine were carried out mainly in normotensive Wistar rats. However, it is well known that the liver organelles are more susceptible to some hepatotoxicants in hypertensive animals than in normotensive [22].

Thus the aim of this study was to investigate the possible differences of toxic effects of D- amphetamine in brains after in vivo multiple adminidtration and in hepatocytes, isolated from male and female SHR and compare these effects with toxicity in normotensive male and female WKY rats.

In hepatocytes isolated from SHR it was observed lesser depletion of GSH in both genders compared to their normotensive matched controls. Amphetamine (100 μM) reduced a cellular GSH - by 37% in male SHR, whereas the reduction in WKY rats was bigger, by 78% (Table 1). Similar results were obtained for female rats – 29% depletion in SHR vs 62% depletion in WKY rats.

The level of MDA was not changed statistically signifi cant in SHR after incubation with amphetamine (100μM) while the amount of MDA in male and female normotensive WKY rats was increased by 91% and 113 % respectively (Table 1).

Page 262: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

260

Similar results were obtained for cell viability and LDH leakage. Our results are in good correlation with the results obtained by Carvalho et al. [5] in isolated hepatocytes from normotensive rats, incubated with amphetamine. It has to be noted that under the conditions of our study more pronounced changes in the examined parameters were observed in both genders of normotensive than in hypertensive rats. One possible explanation might be a compensatory increase in antioxidant protection in SHR, after additional application of toxic compounds [23].

The control amount of MDA and LDH in untreated hepatocytes, isolated from both male and female SHRs was higher, compared to the control levels in hepatocytes isolated from WKY rats. A plausible explanation could be the accelerated processes ofl ipid peroxidation in hypertensive animals [24].Many authors reported that the oxidative stress in this animal model is characterized by lower GSH level and higher MDA formation [25].

Salminen et al. [26] suggested that amphetamine could protect liver from paracetamol- and bromobenzene-induced hepatotoxicity through induction of Heat Shock Proteins (hsps). They found that the application of amphetamine increased levels of hsp25 and hsp70 in the liver and brain of mice as the result of increase of the body temperature. They hypothesized that elevated levels of hsp25 and hsp70 served as an alternative protein targets for binding of reactive metabolites produced by acetaminophen and bromobenzene and thereby the amount of GSH was preserved and the process of lipid peroxidation was slowed. In this manner the formation of MDA was reduced.

Chan et al. [27] demonstrated increased activation of heat shock transcription factor in SHR and increased expression of hsp70. We suggested that the weaker changes of MDA and GSH in both genders SHR, in comparison with their normotensive controls, after administration of amphetamine, could be also due to a protective effect of hsp70

Brain toxicity of amphetamine was also assessed by the level of MDA, the amount of GSH and the activity of the neuronal NOS. The results were similar.

As it was shown for hepatocytes in the brain the initial GSH level in control groups SHR was lower and MDA quantity was higher especially in male SHR, compared to WKY. Thus hypertensive animals would be more sensitive to brain toxicity, induced by D- amphetamine.

Repeated administration of the investigated compound resulted in a statistically signifi cant reduction of GSH levels in male and female WKY and non-signifi cant decrease in the level of GSH in male and female SHR (Table. 2).

Cerebral MDA increased signifi cantly in male and female WKY, but not in SHR. These results obtained for both sex WKY correlated and could be explained by the increased activity of nNOS. NO, produced by nNOS, is related to some neurotoxic effects of amphetamine described in the literature. Bashkatova et al. [28] found that systemic administration of amphetamine resulted in increased formation of NO in the striatum and cortex of rats, and was linked with generation of free radicals. In support of these studies and researches Itzhak and Ali [29] found that inhibition of nNOS activity by certain drugs reduced amphetamine neurotoxicity.

In both sexes SHR nNOS activity was not altered after administration of amphetamine (Table 2). It was found that for untreated male and female SHR this activity was higher compared to untreated male and female WKY. This effect was most likely related to the

Page 263: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

261

pathogenetic mechanism of oxidative stress in hypertensive rats. The increased activity of nNOS is related to the processes of dependence to psychoactive substances and to increased release of dopamine and other neurotransmitters by the reward system in the brain. Yoshimoto et al. [30] found dysfunction in the reward system in the CNS in SHR and Hoskins et al. [31] established decreased levels of dopamine in this strain. These fi ndings could explain to some extent the lack of activation of nNOS in SHR after administration of amphetamine.

In brain as well in hepatocytes, under the infl uence of amphetamine the parameters of antioxidant protection GSH and MDA changed to a lesser extent in hypertensive animals of both sexes, which was most probably due to a compensatory increase of antioxidant enzyme activity mechanisms in hypertensive condition.

On the basis of this study we might suggest that the differences in brain and hepatic toxicity of D- amphetamine in hypertensive rats are probably due to their special pathophysiological characteristics and compensatory protective mechanisms.

References

[1] M. T. Ferreira et al. Effect of physical exercise on markers of acute cardiotoxicity induced by d- amphetamine in an animal model, Rev Port Cardiol25 (2006), 983-96.

[2] M. Filip et al. The serotonergic system and its role in cocaine addiction, Pharmacol Rep 5 (2005), 685-700.[3] F. Carvalho et al. Repeated administration of d-Amphetamine results in a time-dependent and dose-

independent sustained increase in urinary excretion of p-hydroxyamphetamine in mice, J Health Sci53 (2007), 371-377.

[4] C. Advocat. Literature Review: Update on Amphetamine Neurotoxicity and Its Relevance to the Treatment of ADHD, J of Att Dis11 (2007), 8-16.

[5] F. Carvalho et al. D-Amphetamine interaction with glutathione in freshly isolated rat hepatocytes, Chem Res Toxicol9 (1996), 1031-1036.

[6] J. Ren. Infl uence of gender on oxidative stress, lipid peroxidation, protein damage and apoptosis in hearts and brains from spontaneously hypertensive rats, Cli Exp Pharmacol Physio 34 (2007), 432-438.

[7] T. Sagvolden, T. Xu. Amphetamine improves poor sustained attention while d- amphetamine reduces overactivity and impulsiveness as well as improves sustained attention in an animal model of Attention-Defi cit/Hyperactivity Disorder (ADHD) Behav Brain Funct4 (2008) 1-12.

[8] Robinson et al. Sex differences in amphetamine elicited rotational behavior and the lateralization of striatal dopamine in rats, Brain Res Bull 5 (1980), 539–545.

[9] C. A. Brass and S. D. Glick. Sex differences in drug-induced rotation in two strains of rats, Brain Res223 (1981), 229–234.

[10] A. Milesi-Hallй. Sex Differences in (+)-Amphetamine- and (+)-Methamphetamine induced Behavioral Response in Male and Female Sprague-Dawley Rats, Pharmacol Biochem Behav 86 (2007), 140–149.

[11] J. B. Becker et al. Sex differences in drug abuse, Front Neuroendocr29 (2008), 36–47.[12] Concil of Europe (1991) European Convention for the Protection of Vertebrate Animals Used for

Experimental and Other Scientifi c Purposes (ETS123) http://conventions.coe.int/treaty/en/treaties/html/123.htm

[13] D. Fau, A. Berson, D. Eugene, B. Formently, C. Fisch, D. Pessayre. Mechanism for the hepatotoxicity of the antiandrogen Nilutamide. Evidence suggesting that redox cycling of this nitroaromatic drug lead to oxidative stress in isolated hepatocytes, J Pharm Exp Ther 263 (1992), 69-77.

[14] H. U. Bergmeyer, K. Gawehn, M. Grassl. Methods of Enzymatic Analysis, Verlag Chemie, Weinheim, (1974) 481-482.

[15] R. G. Knowles, S. Moncada. Nitric oxide synthases in mammals. Biochem J298 (1994), 249–258.[16] O. H. Lowry. Protein measurement with the Folin phenol reagent. J Biol Chem193 (1951), 265–75.[17] A. H. Polizio, C. Pena. Effects of angiotensin II type 1 receptor blockade on the oxidative stress in

spontaneously hypertensive rat tissues, Regulatory Peptides128 (2005), 1-5.

Page 264: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

262

[18] E. A. Bump, Y.C. Taylor, M. J. Brown. Role of glutathione in the hypoxic cell cytotoxicity of misonidazole. Cancer Res43 (1983), 997 – 1002.

[19] A. L. Jones, K. J. Simpson. Review article: Mechanisms and management of hepatotoxicity in ecstasy (MDMA) and amphetamine intoxications, Aliment Pharmacol Ther13 (1999), 129-33.

[20] H. H. Maurer et al. Chemistry, pharmacology, toxicology, and hepatic metabolism of designer drugs of the amphetamine (ecstasy), piperazine, and pyrrolidinophenone types: a synopsis, Ther Drug Monit26 (2004), 127-31

[21] F. Carvalho et al. Effect of d- amphetamine repeated administration on rat antioxidant defences, Arch Toxicol73 (1999), 2

[22] C.T. Hsu. The role of the autonomic nervous system in chemically-induced liver damage and repair - using the essential hypertensive animal model (SHR ), J Auton Nerv Syst 70 (1998), 79-83.

[23] C. Csonka et al., Effects of oxidative stress on the expression of antioxidative defense enzymes in spontaneously hypertensive rat hearts. Free Rad Biol Med29 (2000), 612-619.

[24] J. Ren. Infl uence of gender on oxidative stress, lipid peroxidation, protein damage and apoptosis in hearts and brains from spontaneously hypertensive rats Clinical and Experimental Pharmacology and Physiology34 (2007), 432 – 438.

[25] G. Amores et al., Antioxidant activity of propionyl-L-carnitine in liver and heart of SHR. Life Sci78 (2006), 1945-52.

[26] Salminen et al. Protection against Hepatotoxicity by a Single Dose of Amphetamine: The Potential Role of Heat Shock Protein Induction, Toxicol Appl Pharmacol147 (1997), 247–258.

[27] S. H. Chan et al. Altered Temporal Profi le of Heat Shock Factor 1 hosphorylation and Heat Shock Protein 70 Expression Induced by Heat Shock in Nucleus Tractus Solitarii of Spontaneously Hypertensive Rats, Circulation107 (2003), 339-345

[28] V. Bashkatova, M. Kraus, H. Prast, A. Vanin, K. Rayevsky, A. Philippu. Infl uence of NOS inhibitors on changes in ACH release and NO level in the brain elicited by amphetamine neurotoxicity, Neuroreport 10 (1999), 3155-8.

[29] Y. Itzhak, S. F. Ali. Role of nitrergic system in behavioral and neurotoxic effects of amphetamine analogs. Pharmacol Ther109 (2006), 246-62.

[30] K. Yoshimoto et al. Pharmacological studies of alcohol susceptibility and brain monoamine function in stroke-prone spontaneously hypertensive rats (SHRSP) and stroke-resistant spontaneously hypertensive rats (SHRSR). Tohoku J Exp Med201 (2003), 11-22.

[31] B. Hoskins et al. Effects of cocaine on tyrosine hydroxylase activity in brain areas from SHR and WKY. Brain Res Bull25 (1990), 639-41.

Page 265: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

263

Chapter 34

An Update on the Signifi cance of Pharmacovigilance in Patient Safety

Eren OZCAGLIa,1, Dionysios VYNIASb, Buket ALPERTUNGAa and Aristidis M. TSATSAKISb

aDepartment of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University 34116, Beyazit- Istanbul, Turkey,

bLaboratory of Toxicology, School of Medicine, University of Crete, Voutes 71003, Heraklion, Greece,

Abstract. The aim of pharmacovigilance activities is to make best use of medicines for the treatment or prevention of disease while improving patient/public health and safety. Consequently, drug related risks and risk factors can be identifi ed by good pharmacovigilance practices so that adverse reactions can be avoided or minimized for the further use of medicines. As pre-authorization term clinical trials demonstrate many limitations, it is very important to collect and communicate post marketing pharmacovigilance information effectively, which will provide intelligent evidence-based use of medicines and will prevent a number of adverse reactions.

Ke ywords. pharmacovigilance, adverse drug reactions, clinical trials

IntroductionThe word “pharmacovigilance” derived from Greek “pharmakon-drug/medicine” and

Latin “vigilans-careful/watchful”. Pharmacovigilance has been defi ned as “science and activities relating to the detection, assessment, understanding and prevention of adverse effects or any other drug-related problem” by World Health Organization (WHO).

First awareness for the need of safety testing in drugs goes to year 1937. Over 100 people including children died because of elixir of sulfanilamide which contained diethylene glycol. In 1937 regulations did not require safety testing for drugs and therefore the company had done none [1]. Next major event is thalidomide disaster occurred in early 60s. Thalidomide was patented by Grunenthal in Germany in 1954 and it was launched in October 1957 as a sedative, a pain killer and an anti-emetic suitable for treating morning sickness in pregnancy. The following year it was licensed in many countries including UK. In USA, Frances Kelsey–the Inspector at the Federal Drugs Authority (FDA)—was not satisfi ed with pre-clinical studies and USA did not license thalidomide. In the pre-clinical testing, no tests had been performed on pregnant animals to check the effects on the foetus. Such testing was not usual at that time as it was generally not believed that drugs would cross the placenta and have harmful effects on the foetus [2]. In the late 50s and early 60s, more than 10,000 children in 46 countries were born with deformities such as phocomelia as a consequence of thalidomide use. It is not known exactly how many worldwide victims

Page 266: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

264

of the drug there have been, although estimates range from 10,000 to 20,000. The Australian obstetrician William McBride and the German pediatrician Widukind Lenz suspected a link between birth defects and the drug, in 1961 thalidomide withdrawn in UK [3].

Many preventative actions have been taken in the light of the lessons learned from thalidomide. Thalidomide has undoubtedly had the greatest impact on shaping the regulations in pharmaceutical Industry and pharmacovigilance activities as we know it today [4]. WHO established its Programme for International Drug Monitoring together with the WHO Collaborating Centre for International Drug Monitoring-Uppsala in response to the thalidomide disaster detected in 1961.

Pharmacovigilance activities become more important by preventing patients from harmful effects of drugs. This development has been driven by recognizing the role of pharmacovigilance, the investigation and marketing of a wider range of diverse medicinal products and more stringent and detailed regulatory requirements.

1. Aim of Pharmacovigilance Programmes

In regulatory agencies, the major aims of pharmacovigilance are the early detection of unknown adverse drug reactions, their frequencies and the identifi cation and quantifi cation of risks for the rational and safe use of medicines. The aims of pharmacovigilance within the industry are similar to those of regulatory agencies; that are to protect patients from unnecessary harm by identifying previously unrecognized drug hazards, elucidating pre-disposing factors, refuting false safety signals and quantifying risk in relation to benefit. Today, pharmaceutical companies and the regulatory authorities work even closely together and share information in terms of patient safety [5]. For rational and safe use of medicines each party has to collect data, evaluate and manage possible risks (Figure 1).

Figure 1. Workfl ow for rational and safe use of medicines

2. Pharmacoeconomics and pharmacovigilance activities

The huge increase in both time and cost in bringing medicines to market is increasing drug development budgets to unsupportable levels; and only big multinational pharmaceutical companies are now able to do so [6]. However, several requirements should be met in order to maintain medicines on the market. The only way to avoid drug withdrawals drugs from the market is the rational use of medicines through good pharmacovigilance practices.

Page 267: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

265

Pharmacoeconomic studies on the costs of adverse drug reactions suggest that governments pay considerable amounts from their health budgets in covering the costs associated with them. In a meta-analysis of prospective studies from hospitals in the United States, it was shown that Adverse Drug Reactions (ADRs) ranked from the fourth to sixth leading cause of death [7]. ADRs account for up to 106 000 deaths in the United States annually. The national cost of preventable in-hospital events alone has been estimated to be more than $US 2 billion [8]. Adverse drug reactions has economic burden on national health systems. There are also costs associated with adverse drug reactions in primary health care or by using over the counter drugs, with these costs being diffi cult to estimate.

Interactions must also be considered as potential safety hazards and therefore it is essential to obtain pharmacovigilance data from the use of over-the-counter medicines, including herbal remedies when they require prescription medication or present with an adverse reaction. There is evidence that taking herbal remedy St John’s wort preparations can result in pharmacokinetic or pharmacodynamic interactions that present a risk to patients who are under pharmaceutical treatment. The use of different St John’s wort products and dosages may produce different outcomes in terms of adverse reactions. It is also necessary to report all suspected adverse reactions and interactions associated with herbal remedies in order to establish information about their safety [9]. Escalation in self medication with herbal remedies will increase the importance of pharmacovigilance data collection about these products.

3. Stakeholders in pharmacovigilance

Health Authorities, Marketing Authorization Holders, healthcare providers and patients are the stakeholders in pharmacovigilance activities. Each party has its own unique role for the purpose of drug safety.

Several new processes have been developed for strengthening pharmacovigilance (Figure 2) for both sides Health Authorities and Marketing Authorization Holders. On a regulatory level, these include conditional approval and risk management plans; on a scientifi c level, transparency and increased patient involvement are two important and effective elements of these processes [10].

Figure 2. Key points for strengthening pharmacovigilance

4. Pre-authorization termAt the pre-authorization term, drug safety information can be obtained via animal

experiments and clinical trials. For authorization procedure a series of clinical trials

Page 268: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

266

required which are conducted on humans. The extent of the trials depends on several factors such as the type of drug, disease, indication etc. All conducted clinical trials should have a human safety as a primal objective. In the early phase studies primary goal is often safety and tolerability. However, in later phase studies primary objective is usually effi cacy with safety and tolerability also being included as a secondary objective. Clinical trials are strictly regulated and precautions have been developed to protect individuals involved in trials as regulatory authorities and pharmaceutical companies have similar approaches on the human safety.

5. Limitations of clinical trialsIn clinical trials there are a number of limitations regarding safety. The major limitations

of randomized clinical trials are narrow population (which does not provide suffi cient data on special groups), narrow indications and short duration. Another limit is related to the diffi culty to interpret or generalize the results since the studied population is very different to the population treated in real life [11]. For example, a major limitation of drug related risk assessment is the lack of human data in immunotoxicity. Immunological end points and clinical criteria could be incorporated into clinical trials and epidemiological studies in order to obtain appropriate results [12]. However, this action will not be a solution for the limitations of clinical trials. As the released drug will be used by huge number of patients with different pharmacogenetic variations, ethnicity, dietary habits, concomitant diseases and medications, then it is not possible to assess limitations of clinical trials with single parameters. Therefore, fi ne running of dosage recommendations, reappraisal of indications, safety information assessment and risk management activities are key points which should be further studied for drug safety after licence approval.

6. Post-marketing periodDue to the limitations of clinical trials, post-marketing period is a critical period for

assessing the safety profi le of a drug. Spontaneous adverse event reportings are a common source for identifi cation of signifi cant safety concerns and are important to obtain data from post-marketing period. Post-marketing drug safety surveillance has traditionally been carried out by systemic manual review of reports of suspected adverse drug reactions sent by healthcare professionals, consumers, pharmaceutical companies or regulatory authorities. It is in the best interest of public health to integrate and understand evidence from all possibly relevant information sources on drug safety [13]. These data are used for causality assessment, signal detection and risk management activities (Figure 3) in order to ensure rational usage of drugs.

Figure 3. Risk management in pharmacovigilance

In order to obtain more data, consumer reportings are becoming more important day by day by providing fi rst hand information with less bias on event reported due to lack of medical knowledge. Patients have the potential to report adverse drug reactions directly in

Page 269: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

267

a number of countries including US, UK, Netherlands and New Zealand [14]. However, more motivation is required for consumer reportings.

7. Current status in pharmacovigilance*In different countries, health requirements and the use of medicines considerably vary

due to many reasons, which include different burdens of disease, cultural, ethnic, dietary and economic factors, and also the system developed for the regulation of drugs.

Effectiveness and safety of a medicine need to be considered in each country’s specific context and therefore, there is a need of local pharmacovigilance system for each country to govern rational and safe use of medicines. On the contrary, regulations vary from one country to another and there is a need of pharmacovigilance harmonization in terms of increasing qualifi ed data fl ow and better management of drug safety in all over the world.

Pharmacovigilance activities have a major part to play in clinical practice and in the development of public health policy. In developed countries, the scope of activities in national pharmacovigilance system has expanded to include risk management and communication of medicines to healthcare providers, patients and the public. There is a lack in resources to undertake training and capacity building and to invest in systems for monitoring drug efficacy and safety in most developing countries. As a limited number of these countries have a well-established pharmacovigilance system, the major resources are often allocated to reduce disease morbidity and mortality instead of establishing effective pharmacovigilance system.

References

[1] A. Gupta, L.K. Waldhauser. Adverse drug reactions from birth to early childhood..Pediatr Clin North Am. 44 (1997), 79-92.

[2] A.Dally. Thalidomide: was the tragedy preventable? The Lancet 351 (1998), 1197–1199.[3] J. Botting. The History of Thalidomide. Drug News Perspect15 (2002), 604-611.[4] J.E. Ridings. The thalidomide disaster, lessons from the past,.Methods Mol Biol 947 (2013), 575-86.[5] J. C. C. Talbot, B. S. Nilsson. Pharmacovigilance in the pharmaceutical industry. Br J Clin Pharmacol45

(1998), 427–431.[6] P.J. Lachmann. The penumbra of thalidomide, the litigation culture and the licensing of pharmaceuticals.

Q J Med105 (2012), 1179–1189.[7] J. Lazarou,B.H. Pomeranz, P.N. Corey. Incidence of adverse drug reactions in hospitalized patients: a

meta-analysis of prospective studies. JAMA279 (1998), 1200-1205.[8] A. Neubert, H. Dormann, H.U. Prokosch, T. Bьrkle, W. Rascher, R. Sojer, K. Brune, M. Criegee-Rieck.

E-pharmacovigilance: development and implementation of a computable knowledge base to identify adverse drug reactions. Br J Clin Pharmacol76 (2013), 69–77.

[9] L. Henderson, Q.Y. Yue, C. Bergquist, B. Gerden, P. Arlett. St John’s wort (Hypericum perforatum): drug interactions and clinical outcomes. Br J Clin Pharmacol 54 (2002), 349-356.

[10] L. Hдrmark, A.C. van Grootheest. Pharmacovigilance: methods, recent developments and future perspectives. Eur J Clin Pharmacol 64 (2008), 743-752.

[11] J.P. Collet. Limitations of clinical trials.Rev Prat 50 (2000),833-837.[12] J. Descotes. Methods of evaluating immunotoxicity.Expert Opin Drug Metab Toxicol 2 (2006), 249-59.[13] P.M. Coloma, G. Trifi rт, V. Patadia, M. Sturkenboom.Postmarketing safety surveillance: where does signal

detection using electronic healthcare records fi t into the big picture? Drug Saf 36 (2013), 183-197.[14] F. van Hunsel, L. Hдrmark, S. Pal, S. Olsson, K. van Grootheest. Experiences with adverse drug reaction

reporting by patients: an 11-country survey. Drug Saf 35 (2012), 45-60.

* A part of this manuscript was submitted to the Innovative Medicine Initiative under the scientifi c section WEBAE – Leveraging Emerging Technologies for Pharmacovigilance with the proposal entitled “Mobile Application Based Reporting for ADverse DRug Reactions

Page 270: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

268

Chapter 35

Effects of multiple ethanol administration on some brain and hepatic biochemical

parameters in male and female spontaneously hypertensive rats ( SHR)

Rumyana SIMEONOVA1, Vessela VITCHEVA and Mitka MITCHEVA

Laboratory of drug metabolism and drug toxicity, Department of pharmacology, pharmacotherapy and toxicology, Faculty of pharmacy, Medical University – Sofi a

Abstract. Ethanol addiction, developed after multiple ethanol administration, is characterized with neurotoxicity and hepatotoxicity. Hypertension is a pathophysiological status that additionally contributes to damages in central nervous system and to a liver disfunction. The aim of our study was to investigate the effects of mutiple ethanol administration on some brain and hepatic biochemical parameters in male and female SHRs, compared to their age-matched normotensive controls Wistar Kyoto rats (WKY). The animals from both strains were treated with ethanol (3g/kg p.o. 14 days). Twenty four hours after the last administration, the samples were taken for measuring the following parameters: nNOS activity (brain), MDA quantity and GSH level (both in brain and liver) and antioxidant enzymes CAT, SOD, GPx, GR and GST in liver spectrometrically. In SHRs multiple ethanol treatment resulted in less pronounced effect on the assessed parameters, both in brain and liver, especially in female SHRs - nNOS activity was not changed and GSH level was less affected, compared to male SHR and to female WKY. Ethanol administration enhanced MDA level in both strains, in both genders and in both organs, but less pronounced in SHRs. Antioxidant enzymes were less affected in both genders of SHRs. Catalase activity increased only in SHRs and decreased in WKY after alcohol intake.The difference in the effect of ethanol between the strains might be due to the changed reactivity and pathophysiological characteristics of the hypertensive model.

Key words. Ethanol, SHR, brain toxicity, hepatotoxicity

IntroductionEthanol addiction, developed after multiple ethanol administration, is a complex

phenomenon that apart of tolerance and dependence development is characterized with neurotoxicity and hepatotoxicity. Repeated administration of ethanol damages organs and

1 Corresponding Author: Rumyana SIMEONOVA, Laboratory of drug metabolism and drug toxicity, Department of pharmacology, pharmacotherapy and toxicology, Faculty of pharmacy, Medical University, 2, Dunav str. Sofi a, Bulgaria; E-mail: [email protected]

Page 271: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

269

systems, affects some pathological conditions in the organism and changes drug metabolism. Main organ whose function the alcohol violates is the liver. Liver injuries have complex mechanism which includes reactive metabolites (acetaldehyde, ROS, etc.) of alcohol. They possess direct cytotoxicity and form adducts with certain proteins in hepatocytes [1, 2] and increase the reduced form of nicotine-amide-adenine-dinucleotide-phosphate (NADPH). This leads to the accumulation of fat in the liver [2] and free radical induced oxidative stress which provokes the peroxidation of lipids, and infl ammatory response [3].

It is well known that women are more sensitive to ethanol toxicity than men [4, 5, 6]. Cirrhosis occurs earlier in women, although they consume smaller amount of alcohol than men [6]. The relationship between alcohol consumption and the increase in blood pressure is established as early as 1915 by Lian [7]. Excessive alcohol consumption causes hypertension, heart attack and stroke [8], which faces hypertensive patients at huge risk.

Spontaneously hypertensive rats ( SHR) are a genetic model of essential hypertension in human. They are used from many laboratories to characterize some aspects of this pathology, including an infl uence of some toxic compounds as alcohol.

The aim of the following study was to investigate the effects of multiple alcohol administration on some biochemical brain and hepatic parameters in SHRs, compared to normotensive Wistar-Kyoto rats (WKY).

1. Materials 1.1. Experimental AnimalsMale and female SHRs strain Okamoto-Aoki and their normotensive controls Wistar-

Kyoto rats were obtained from Charles River breeding center in Germany. The rats were housed in plexiglass cages (3 per cage) in a 12/12 light/dark cycle, under standard laboratory conditions (ambient temperature 20°C ± 2°C and humidity 72% ± 4%) with free access to water and standard pelleted rat food. A minimum of 7 days acclimatization was allowed before the commencement of the study. All performed procedures were approved by the Institutional Animal Care Committee and the principles stated in the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientifi c Purposes (ETS 123) [9] were strictly followed throughout the experiment. The number of rats wasreduced, as much as possible, depending on statistical signifi cance.

Blood pressure was measured in conscious animals using a semi-automated tail-cuff device (LE5002, Letica S/A, Spain). Before the experimental period, the rats were conditioned to the restraining cylinders. Rats were pre-warmed for 10 min using a temperature – controlled warming holder (37oC) to facilitate tail blood fl ow before their blood pressure was measured. The mean of tree tail-cuff readings was used as the systolic and diastolic blood pressure value.

1.2. ChemicalsAll the reagents used were of analytical grade. Pentobarbital sodium (Sanofi , France),

KH2PO4 (Scharlau Chemie SA, Spain), MgSO4.7H2O (Fluka AG, Germany), trichloroacetic acid (TCA) (Valerus, Bulgaria), lactate-dehydrogenase (LDH) kit (Randox, UK). HEPES, collagenase from Clostridium histolyticum type I V, albumin, bovine serum fraction V, minimum 98 %, EGTA, 2-thiobarbituric acid (4,6-dihydroxypyrimidine-2-thiol; TBA)

Page 272: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

270

were purchased from Sigma Aldrich, (Germany). Sucrose, Tris, DL-dithiotreitol, D-glucose, phenylmethylsulfonyl fl uoride, potassium phosphate, calcium chloratum (CaCl2), magnesium chloratum (MgCl2), L-arginine, L-valine, hemoglobin bovine, beta –Nicotinamide adenine dinucleotide 2`-phosphate reduced tetrasodium salt (NADPH), ethylenediaminetetraacetic acid (EDTA), 2,2’-dinitro-5,5’dithiodibenzoic acid (DTNB) were obtained from MERCK (Germany).

2. Experimental design The normotensive and hypertensive rats were divided in 8 experimental groups. Group 1: control male SHRGroup 2: male SHR treated with ethanol (3g/kg p.o. 14 days) [10]Group 3: control female SHRGroup 4: female SHR treated with ethanol (3g/kg p.o. 14 days)Group 5: control male WKY ratsGroup 6: male WKY treated with ethanol (3g/kg p.o. 14 days)Group 7: control female WKY ratsGroup 8: female WKY treated with ethanol (3g/kg p.o. 14 days)The animals in all groups were sacrifi ced by decapitation on the day fourteenth of

the experiment, blood and livers were taken for biochemical assay. For all following experiments, livers were removed, washed with ice-cold saline solution (0.9% NaCl), blotted dry, weighted and divided into three pieces (stored on ice), 1 g each for assessment of MDA quantity, glutathione (GSH) levels and preparation of liver microsomes for evaluation of phase I of biotransformation. Tissues were homogenized in ice-cold buffers using glass homogenizer (PX-OX 2000).

3. Methods3.1. Biochemical parameters in liver tissue3.1.1. Preparation of liver homogenate for lipid peroxidation (LPO) assessment. LPO was determined by measuring the rate of production of thiobarbituric acid

reactive substances (TBARS), expressed as malondialdehyde (MDA) equivalents [11]. One volume of homogenate was mixed with 1 mL 25% trichloracetic acid (TCA) and 1 mL 0.67% TBA. Sampleswere then mixed thoroughly, heated for 20 min in a boiling water bath, cooled and centrifuged at 4000 rpm for 20 min. The absorbance of supernatant was measured at 535nm against a blank that contained all the reagents except the tissue homogenate. The MDA concentration was calculated using a molar extinction coeffi cient of 1.56 x 105 M-1 cm-1 and expressed in nmol/g wet tissue.

3.1.2. Preparation of liver homogenate for GSH assessment. GSH was assessed by measuring non-protein sulfhydryls after precipitation of proteins

with TCA [12]. Tissues were homogenized in 5% TCA and centrifuged for 20min at 4000 x g. The reaction mixture contained 0.05 mL supernatant, 3 mL 0.05 M phosphate buffer (pH = 8) and 0.02 mL 5,5’-dithio-bis-(2-nitrobenzoic acid) reagent. The absorbance was determined at 412 nm, and the results expressed as nmol/g wet tissue.

Page 273: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

271

3.1.3. Preparation of liver microsomes for biochemical assay. The excised, perfused and minced livers were homogenized with 3 mL of 1.17%

KCl and then centrifuged at 10000 x g for 30 min. The supernatant fractions were further centrifuged at 105 000 x g for 60 min. The resulting microsomal pellets were stored at -20°C until assayed. At the day of assay, the microsomal pellets were re-suspended and diluted with phosphate buffer-ethylenediaminetetraacetic acid (pH= 7.4) [13]. Liver protein concentration was measured, using the method of Lowry (1951) [14] and was adjusted to 4 mg/mL.

3.1.3.1. Assessment of cytochrome P450 quantityCytochrome P450 was quantifi ed spectrophotometrically as a complex with CO at 450

nm and expressed as nmol/mg-1 protein [15].3.1.3.2. Assay of aniline 4-hydroxylase activity (AH)The enzyme activity was evaluated by 4-hydroxylation of aniline to 4-aminophenol that

was chemically converted to a phenol–indophenol complex with an absorption maximum at 630 nm. Enzyme activity is expressed as nmol/min/mg protein [16].

3.1.3.3. Assay of ethylmorphine-N-demethylase (EMNDactivity) The enzyme activity was evaluated by the formation of formaldehyde, trapped in the

solution as semicarbazone and measured by the colorimetric procedure of Nash, at 415 nm. Enzyme activity is expressed as nmol/min/mg protein [16].

3.1.4. Preparation of liver homogenates for antioxidant enzyme activity measurementThe livers were rinsed in ice-cold physiological saline and minced with scissors.

10% homogenates were prepared in 0.05 M phosphate buffer (pH = 7.4), then centrifuged at 7000 x g and the supernatants then used for antioxidant enzymes assay. The protein content of liver homogenate was measured by the method of Lowry [14] with bovine serum albumin as a standard.

3.1.4.1. Catalase activity (CAT)Catalase activity was assessed following the method of Aebi et al. [17]. Briefl y, 10 μL

of homogenate was added to 1 990 μL of H2O2 solution (containing 6.8 μL of 30 % H2O2 + 1 983.2 μL 0.05 M phosphate buffer, pH=7.4). CAT activity was determined by monitoring the H2O2 decomposition which was measured by the decrease in absorbance at 240 nm for 1 min. Enzyme activity was calculated using a molar extinction coeffi cient of 0.043 mM -1 cm-1 and expressed as μM/min/mg.

3.1.4.2. Superoxide dismutase activity (SOD)Superoxide dismutase activity was measured according to the method of Misura

and Fridovich [18], following spectrophotometrically the autoxidation of epinephrine at pH=10.4, 30° C, using the molar extinction coeffi cient of 4.02 mM-1 cm-1. The incubation mixture contained 50 mM glycine buffer, pH=10.4. The reaction is started by the addition of epinephrine. SOD activity is expressed as nmol of epinephrine that are prevented from autoxidation after addition of the sample.

3.1.5. Evaluation of aspartate aminotransferase, alanine aminotransferase and lactate dehydrogenase activities in serum.

The blood was taken into a tube containing ethylene glycol tetraacetic acid. Serum was separated by centrifugation in bench centrifuge (Eppendorf MiniPlus) at 10 000 rpm for 10

Page 274: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

272

min, 4°C. Serum activity of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) was measured, using automated optimized spectrophotometrical method (COBOS integra 400 plus Roche Diagnostics). Serum activity of lactate dehydrogenase (LDH) was measured using commercial available kit (LDH opt., Randox).

3.2. Biochemical parameters in brain tissue3.2.1. Preparation of brain tissue extracts and assessment of nNOS activityRats were decapitated and the brains were minced and homogenized in 10 volumes of

buffer, containing 320 mmol L-1 sucrose, 50 mmol L-1 Tris, 1 mmol L-1 DL-dithiotreitol, 100 μg/Lphenylmethylsulfonylfl uoride (pH=7.2) [19].The homogenates were then centrifugated at 17 000 x g for 60 min. The protein content was measured by the method of Lowry [14] with bovine serum albumin as a standard.

Neuronal NOS activity was measured spectrophotometrically using the oxidation of oxyhemoglobin to methemoglobin by NO [16]. The incubation medium contained 40 mM potassium phosphate buffer, pH=7.2, 200 μmol L-1 CaCl2, 1 mmol L-1MgCl2, 100 μmol L-1 L-arginine, 50 mmol L-1L-valine, 2.6 μmol L-1 oxyhemoglobin, 100 μmol L-1 NADPH and brain extract.The change in the difference in absorbance at 401 nm and 421 nm was monitored with a double split beam spectrophotometer (Spectro UV-VIS Split), at 37 °С. The activity of the enzyme was expressed in nmol/min/mg, using the milimolar extinction coeffi cient of methemoglobin 77.2 М-1cm-1.

3.2.2. Lipid peroxidation in brain homogenateLipid peroxidation was determined by measuring the rate of production of thiobarbituric

acid reactive substances (TBARS) (expressed as malondialdehyde equivalents (MDA) [11]. One volume of brain homogenate was mixed with one volume 25 % trichloracetic acid (TCA) and one volume 0.67 % thiobarbituric acid (TBA). Samples were then mixed thoroughly, heated for 20 min in a boiling water bath, cooled and centrifuged at 4000 rpm for 20 min. The absorbance of supernatant was measured at 535 nm against a blank that contained all the reagents except the tissue homogenate. Malondialdehyde concentration was calculated using a molar extinction coeffi cient of 1.56 x 105 M-1 cm-1 and expressed in nmol/g wet tissue.

3.2.3. GSH assessment in brain homogenate GSH was assessed by measuring non-protein sulfhydryls after precipitation of proteins

with TCA [12]. Brain tissues were homogenized in 5% trichloracetic acid (TCA) and centrifugated for 20 min at 4 000 x g. The reaction mixture contained 0.05 mL supernatant, 3 mL 0.05 M phosphate buffer (pH = 8) and 0.02 mL DTNB reagent. The absorbance was determined at 412 nm and the results expressed as nmol/g wet tissue.

3.3. Statistical analysisStatistical analysis was performed using MedCalc (MedCalc Software bvba,

Mariakerke, Belgium). Results are expressed as mean SEM for six rats in each group. The signifi cance of the data was assessed using the non-parametric Mann–Whitney U test. Values of p≤0.05 were considered statistically signifi cant.

4. Results4.1. Effects of ethanol on liver biochemical parametersThe effects of ethanol on phase I biotransformation enzymes are shown in Table 1.

Page 275: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

273

After repeated ethanol administration the following changes in the drug metabolizing enzyme activities were observed:

Total amount of cytochrome P450 was statistically signifi cant (p < 0.05) increased in both strains and in both sexes as follows: in WKY rats - by 56 % in male and by 25 % in female; in SHR – by 39 % in male and by 68 % in female.

AH activity was increased statistically signifi cant (p < 0.05) by 47 % and by 97 % in male and female WKY rats, respectively. In SHR strain the enzyme activity was elevated by 37 % in males and by 164 % in females.

EMND activity was increased statistically signifi cant (p < 0.05) only in female SHR by 44 %. In the other groups of animals, the activity of this enzyme was not altered after multiple administration of ethanol.

Table 1. Effects of repeated ethanol administration, on the amount and activity of certain enzymes from the fi rst phase of biotransformation, in male and female SHR and WKY

Parameters Groups Male WKY Female WKY MaleSHR FemaleSHR

Cyt. Р450 nmol/mg

Control 0,336±0,001 0,246±0,006* 0,328±0,001 0,212±0,004+

Ethanol 0,523±0,004* 0,307±0,004# 0,456±0,003+ 0,356±0,011&

AHnmol/mg/min

Control 0,043 ± 0,002 0,032 ± 0,003* 0,041 ± 0,004 0,025 ± 0,004+

Ethanol 0,063±0,002* 0,063±0,004# 0,056±0,002+ 0,066±0,003&

EMNDnmol/mg/min

Control 0,332 ± 0,007 0,223 ± 0,003* 0,472 ± 0,004* 0,143 ± 0,011#+

Ethanol 0,342 ± 0,012 0,236±0,006 0,468±0,008 0,206±0,009&

*p<0.05 vs male WKY; +p<0.05 vs male SHR; #p<0.05 vs female WKY; &p<0.05 vs female SHR

Effects of ethanol on hepatic GSH and MDA are shown in table 2. Multiple administration of ethanol reduced the amount of hepatic GSH only in normotensive animals by 35% (p<0.05) in male WKY and by 28% (p<0.05) in female WKY (table 2). In both genders hypertensive rats, GSH did not change signifi cantly. The level of hepatic MDA increased statistically signifi cant in male WKY by 53% (p<0.05), in female WKY by 36% (p<0.05), in male SHR by 24% (p<0.05) and in female SHR by 22% (p <0.05).

Table 2. Effects of ethanol on the level of hepatic GSH and MDA in male and female SHR, compared with male and female WKY.

Parameters Groups MaleWKY FemaleWKY MaleSHR FemaleSHR

GSH nmol/g Control 6,34 ± 0,16 4,82 ± 0,09* 4,53 ± 0,14* 5,38 ± 0,18#

Ethanol 4,12 ± 0,13* 3,46 ± 0,34# 4,02 ± 0,13 5,02 ± 0,34

Page 276: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

274

MDAnmol/g Control 1,05 ± 0,03 1,46 ± 0,06* 1,65 ± 0,04* 1,68 ± 0,03

Ethanol 1,61±0,05* 1,98±0,06# 2,05±0,07+ 2,05±0,03&

*p<0.05 vs male WKY; +p<0.05 vs male SHR; #p<0.05 vs female WKY; &p<0.05 vs female SHR

Table 3. Effects of ethanol after multiple administration on CAT and SOD in the liver of male SHR, compared to male WKY

Parameters Groups Male WKY Male SHR

САТ nmol/mg/min Control 33,38 ± 3,73 25,12 ± 2,81*

Ethanol 21,69 ± 4,98* 44,02 ± 3,12+

SODnmol/mg/min Control 152,5 ± 12,27 94,43 ± 9,11*

Ethanol 76,25 ± 10,21 145,8 ± 23,12+

*p<0,05 vsWKY; +p<0,05 vsSHR

Ethanol reduced the activity of liver CAT in WKY by 35% (p<0.05), whereas in SHR it increased enzyme activity by 75% (p<0.05) (Table 3). The activity of SOD decreased by 50% (p<0.05) in WKY, after administration of ethanol, while in SHR enzyme activity was increased by 54% (p<0.05), compared to the respective control group.

Table 4. Effect of ethanol on the activity of serum ALAT and ASAT, in male and female SHR and WKY

Parameters Groups Male WKY Female WKY Male SHR Female SHR

ALAT U/L Control 66,2 ± 6,2 76,2 ± 12,1 82,6 ± 4,2 87,2 ± 11,8

Ethanol 90,3±9,2* 80,5±8,1 112,1±9,1+ 72,2±18,3

ASAT U/L Control 134,2 ± 12,5 112,4 ± 4,5 129,3 ± 9,8 170,3 ± 11,3

Ethanol 188,2±19,1* 194,8±8,3# 190,2±12,2+ 132,2±12,1&

*p < 0.05 vs male WKY; +p < 0.05 vs male SHR ; #p < 0.05 vs female WKY; &p < 0.05 vs female SHR

Repeated administration of ethanol increased statistically signifi cant the activity of ALAT in both strains male rats by 36% and does not alter enzyme activity in females. Ethanol increased signifi cantly the activity of ASAT by 40% (p<0.05) in male WKY and by 74% (p<0.05) in female WKY. In male SHR , ASAT activity increased by 47% (p<0.05), while in female SHR , ASAT decreased by 22% (p<0.05) (Table 4).

4.2. Effects of ethanol chronic administration on brain biochemistry

The results obtained after repeated administration of ethanol on some brain biochemical parameters are given in Table 5. The level of GSH in the brain was reduced by 41%

Page 277: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

275

(p<0.05) in male WKY, by 42% (p<0.05) in female WKY, by 37% (p<0.05) in male SHR and by 20% (p<0.05) in female SHR . The amount of MDA was increased by 54% (p<0.05) in male WKY, by 37% (p<0.05) in female WKY, by 42% (p<0.05) in male SHR and by 54 % (p<0.05) in female SHR , compared to controls. The activity of nNOS was increased in male WKY by 84% (p<0.05), in female WKY by 79% (p <0.05) and by 34% (p<0.05) in male SHR , compared to controls. No change was observed in the enzyme activity in female SHR .

Table 5. Effect of ethanol on the levels of GSH and MDA and activity of nNOS in male and female SHR and WKY.

Parameters Groups Male WKY Female WKY Male SHR Female SHR

GSH nmol/g Control 1,28±0,03 1,21±0,07 0,83±0,05* 1,27±0,02+

Ethanol 0,75±0,07* 0,70±0,05# 0,52±0,12+ 1,02±0,06&

MDAnmol/g Control 3,82 ±0,05 5,14 ±0,07* 4,86 ±0,04* 5,51 ±0,09

Ethanol 5,89±0,12* 7,06±0,14# 6,88±0,16+ 8,48±0,13&

NOS nmol/mg/min

Control 0,583 ± 0,09 0,656 ± 0,12 0,762 ±0,11* 0,861 ± 0,08+

Ethanol 1,073± 0,18 1,171± 0,13# 1,021± 0,15+ 0,940± 0,09

*p < 0.05 vs male WKY; +p < 0.05 vs male SHR; #p < 0.05 vs female WKY; &p < 0.05 vs female SHR

5. DiscussionEthanol is a substance which induces drug metabolizing enzymes in humans and

rodents and in the same time is toxic for those species. A suitable model for studying the effect of alcohol on the metabolic capacity of the liver and its toxicity both in the liver and brain, in normotensive and hypertensive rats from both sex, is a 14-day oral treatment with 20% solution of ethanol (3g/ kg) [10]

It was found that the amount of total cytochrome P450 increased signifi cantly in all groups of animals (Table 1). In WKY rats ethanol induction of cytochrome P450 was higher in male rats, while in hypertensive strain this induction was higher in female rats. Similar results were obtained by other researchers [20]. The activity of AH was higher in both strains female rats (by 97% for WKY, and by 164% for SHR), compared with males (by 47% for WKY and by 37% for SHR) (Table 1). Our results are in agreement with the results obtained by Kishimoto et al. [21]. They found that the rate of ethanol metabolism was higher in females than in males because CYP2E expression in this sex was more pronounced. The authors had shown that the enzymes related to ethanol metabolism were controlled by testosterone and β-estradiol. Rachamin et al. [22] found that testosterone possessed an inhibitory effect and estradiol - stimulatory effect on the activity of alcohol dehydrogenase (ALDH). The letter was more active in female than in male SHR, i.e. women metabolize alcohol more quickly through this alternative enzyme system. This high enzyme activity leads to a faster production of larger quantities of a toxic reactive metabolite - acetaldehyde. To a large extent it is associated with more pronounced hepatotoxicity of ethanol in women [23]. Jarvelainen et al. [24] observed over the 40-fold increase in expression of CYP2E1 in female Wistar rats after chronic administration of alcohol, which is a prominent cause

Page 278: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

276

of oxidative stress, increased production of ROS (superoxide radical - O2. - and hydrogen peroxide H2O2 ) and in the presence of iron catalyzes the formation of potent oxidants such as hydroxyl radicals. Extremely pronounced induction of CYP2E1-dependent oxidation of ethanol, explain a heavy alcoholic liver disease in women, established by many authors [25, 26].

In our study we observed an increase by 44% in the activity of EMND in female SHR ( Table 1). There are no published data on the impact of alcohol on the activity of this enzyme but given the pathological status of these animals and the proved involvement of this enzyme system in the pathogenesis of hypertension [27, 28] it was possible to assume such enzymatic alteration in the rate of demethylation.

Repeated administration of ethanol was accompanied by an alteration on the activity of hepatic transaminases ALAT and ASAT (Table 4), by a depletion of GSH, with CYP2E1-mediated oxidative stress and a reduction in the antioxidant defenses [29, 30]. In the performed tests it wasfound a reduction in the level of hepatic GSH in male andfemale WKYrats (Table 2). Ethanol exhausted GSH through the generation of free radicals as well as by inhibiting the mitochondrial GSH transporter [31].

In SHR alcohol did not affect the level of GSH, while the activity of antioxidant enzymes CAT and SOD was increased (Table 3). Similar results were established by Csonka et al. [32] who concluded that the additional exposure on prooxidants, (i.e. ethanol) increased the antioxidant defense activity in SHR. They explained this phenomenon by a rebound GSH synthesis triggered by oxidative stress. It is possible that ethanol induces the activity of enzymes involved in de novo synthesis of GSH in the liver. In normotensive rats the activity of antioxidant enzymes CAT and SOD was decreased (Table 3), as it was discovered by other researches [29, 33]

Induction of microsomal CYP2E1 activity is accompanied not only by the generation of ROS and reduced oxidation resistance, but also by lipid peroxidation (LP) and by an increased formation of MDA [34]. We found that the amount of hepatic MDA increased more pronounced in both genders WKY rats while the increase of MDA level in SHR was lower and nearly the same for both sexes, although the initial level of MDA was higher in hypertensive control rats, compared with normotensive control groups (Table 2).

The changes of this parameter after ethanol administration, in the brain of WKY rats, were similar as in the liver. MDA increased more pronounced in the brain than in the liver tissue of male SHR ( by 42%) and of female SHR ( by 54%) (Table 5). More prominent changes in the brain of hypertensive animals might be due to the higher sensitivity of the neuronal tissue to the effects of oxygen radicals [35]. Ethanol can cause processes of lipid peroxidation through several mechanisms. Altering the redox status of the cell, it causes the accumulation of fatty acids, which are substrates for LP. Das et al. [30] explain the process of induction of LP by increasing the amount of phospholipids in membranes, by altered membrane fl uidity and transmembrane potential resulting from the received alcohol. Acetaldehyde also plays a role because of the direct reduction of GSH by binding to its sulfhydryl groups. According to Bachmanov [36] spontaneously developing hypertension changes hemodynamic and induces hypoxia, which activates processes of LP in the brain. Becker et al. [37] found that brain atrophy occurs more rapidly in women addicted to alcohol than in men.

Page 279: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

277

Higher GSH depletion in the brain was observed in both sex WKY rats (Table 5). A slight decrease in the level of brain GSH in hypertensive rats is most likely associated with a pronounced stimulation of the activity of antioxidant enzymes in this pathology.

Based on the data about accelerated processes of lipid peroxidation and oxidative stress in the brain we investigated the changes in the activity of nNOS. Dizon et al. [38] showed that chronic alcohol exposure increased nNOS expression and NO production in neuronal cultures. Zima et al. [39] found increased formation of peroxynitrite (O2 * - + NO * → ONOO-), which was involved in ethanol-induced oxidative stress.

Nitric oxide is a key factor in vascular homeostasis, an important mediator in neuronal transduction and has cytotoxic effects. After repeated administration of ethanol, the activity of nNOS was increased statistically signifi cantly in male and in female WKY. In male SHR, enzyme activity was induced by 34%, while in female SHR no change was observed (Table 5).

Yoshimoto et al. [40] established that spontaneously hypertensive rats predisposed to stroke (SHRSP) voluntarily consumed more frequently and in greater amounts alcohol. At the same time the levels of dopamine and serotonin (5-HT) in these rats were lower. Hoskins et al. [41] found a lower activity of tyrosine-hydroxylase and an increased infl ux of Ca2 + ions, which was associated with increased activity of nNOS, as well as an increased levels of NO in untreated SHR, as we confi rmed in our study. However Yoshimoto [40] showed that hypertensive SHRSP condition was associated with dysfunction in the reward system of the brain, which explained the increased alcohol consumption in these animals, and especially in females [42]. We established a very low activation of nNOS in male SHR and no change in enzyme activity in female SHR after alcohol consumption, which showed an altered reactivity of females.

On the basis of this study we might suggest that the observed differences are probably due to the changed reactivity and pathophysiological characteristics of SHR and that this strain possess some compensatory mechanisms to protection.

References:

[1] C. S. Lieber. Alcohol and the liver: metabolism of alcohol and its role in hepatic and extrahepatic disease, Mount Sinai J Med67 (2000), 84–94.

[2] H. J. Zimmerman. Hepatotoxicity: the adverse effects of drug and other chemicals on the liver. Lippincott Williams and Wilkins, Philadelphia, (1999), 147–175.

[3] H. Jarvelainen. Infl ammatory Responses in Alcoholic Liver Disease. National Public Health Institute, Helsinki, (2000), 1–75.

[4] N. Enomoto et al., Gender differences in alcoholic liver injury. Nihon Arukoru Yakubutsu Igakki Zasshi39 (2004), 163–167.

[5] C. Muller. Liver, alcohol and gender, Wien Med. Wochenschr156 (2006), 523–526[6] Sharma et al., Increased severity of alcoholic liver injury in female verses male rats:A microarray analysis,

Exp Mol Pathol84 (2008) 46–58.[7] C. Lian. L’alcoholisme cause d’hypertension arterielle, Bulletin of Academy National Medicine, Paris, 7

(1915), 525-528[8] N. Nakanishi, et al. Alcohol consumption and risk for hypertension in middle-aged Japanese men, J

Hypertension19 (2001), 851-855

Page 280: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

278

[9] Concil of Europe (1991) European Convention for the Protection of Vertebrate Animals Used for Experimental and Other Scientifi c Purposes (ETS123) http://conventions.coe.int/treaty/en/treaties/html/123.htm

[10] P. Pramyothin et al. Hepatoprotective activity of Phyllanthus amarus Schum. et. Thonn. extract in ethanol treated rats: In vitro and in vivo studies Journal of Ethnopharmacology114 (2007), 169–173

[11] A. H. Polizio, C. Pena. Effects of angiotensin II type 1 receptor blockade on the oxidative stress in spontaneously hypertensive rat tissues, Regulatory Peptides128 (2005), 1-5.

[12] E. A. Bump, Y. C. Taylor, M. J. Brown. Role of glutathione in the hypoxic cell cytotoxicity of misonidazole, Cancer Res, 43 (1983), 997 – 1002

[13] F. P. Guengerich. Microsomal enzymes involved in toxicology. Analysis and separation. In:. Principals and Methods of Toxicology Heis AW (ed.) New York (NY); Raven Press; (1987), 609-634.

[14] O. H. Lowry. Protein measurement with the Folin phenol reagent. J Biol Chem193 (1951), 265–75.[15] T. Omura, R. Sato. The carbon-monoxide-binding pigment of liver microsomes, J Biol Chem239 (1964),

2370[16] L. H. Cohen, R. E. W. van Leeuwen, G. C. F. van Thiel, J. F. van Pelt, S. H. Yap.. Equally potent inhibitors

of cholesterol synthesis in human hepatocytes have distinguishable effects on different cytochrome P 450 enzymes, Biopharm Drug Dispos 21 (2000), 353-364

[17] H. Aebi. Catalase In: Methods of Enzymatic Analysis. 2nd ed. New York: H. V. Bergrenyer, editor. Academic Press, (1974), 673-684.

[18] H. P. Misura, I. Fridovich. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase, J Biol Chem247 (1972), 3170-3175.

[19] R. G. Knowles, S. Moncada. Nitric oxide synthases in mammals. Biochem J298 (1994), 249–258.[20] P. Sessink et al. Infl uence of Aroclor 1254, phenobarbital, β-naphthofl avone, and ethanol pretreatment on

the biotransformation of cyclophosphamide in male and female rats, Toxicology112 (1996), 141-150[21] R. Kishimoto et al. Gender-related differences in mouse hepatic ethanol metabolism J Nutr Sci

Vitamin, 48, (2002), 216-224.[22] G. Rachamin et al. Modulation of alcohol dehydrogenase and ethanol metabolism by sex hormones in the

spontaneously hypertensive rat. Effect of chronic ethanol administration, Biochem J,2 (1980), 483-90[23] S. Kunitoh et al. Acetaldehyde as well as ethanol is metabolized by human CYP2E1,J Pharmacol Exp

Ther280 (1997), 527-32.[24] Jarvelainen et al. The antiestrogen toremifene protects against alcoholic liver injury in female rats, 35

(2001), 46-52[25] A. Nanji et al. Increased severity of alcoholic liver injury in female rats: role of oxidative stress, endotoxin,

and chemokines,Am J Physiol Gastrointest Liver Physiol281 (2002), 1348-1356.[26] Sharma et al. Increased severity of alcoholic liver injury in female verses male rats: A microarray analysis,

Exp Mol Pathol84 (2008), 46–58 [27] A.K. Basu etal. Augmented arterial pressure responses to cyclosporine in spontaneously hypertensive

rats. Role of cytochrome P-450 3A,Hypertension4(1994), 480−485.[28] S. S. Ghosh etal. Renal and hepatic family 3A cytochromes P450 (CYP3A) in spontaneously hypertensive

rats,Biochem Pharmacol50(1995), 49-54.[29] N. Agnihotri etal.Role of oxidative stress in lansoprazole-mediated gastric and hepatic protection in

Wistar rats,Indian J Gastroenterol3 (2007), 118-21.[30] S. K. Das, D. M. Vasuden. Alcohol-induced oxidative stress, Life Sciences81 (2007)177-187 [31] D. Wu and A. I. Cederbaum. Oxidative stress mediated toxicity exerted by ethanol-inducible CYP2E1,

Toxicol Appl Pharmacol 2 (2005), 70-76[32] C. Csonka et al. Effects of oxidative stress on the expression of antioxidative defense enzymes in

spontaneously hypertensive rat hearts,Free Rad Biol Med29 (2000), 612-619.[33] E. Lamy et al. Ethanol enhanced the genotoxicity of acrylamide in human, metabolically competent

HepG2 cells by CYP2E1 induction and glutathione depletion, Int J Hyg Environ Health211 (2008),74-81.

[34] K. Abir et al. Dietary virgin olive oil protects against lipid peroxidation and improves antioxidant status in the liver of rats chronically exposed to ethanol Nutrition Research28 (2008), 472–479

Page 281: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

279

[35] В. Ю.Куликов и съавт Прекисное окисление липидов и холодовой фактор Клин Мед, (1988) 29-51[36] А. А. Бачманов. Поведенческие и метаболические особености спонтанно-гипертензивных крыс

Физиологический журнал, СССР, 11 (1988)[37] J. B.Becker etal. Sex differences in drug abuse Front Neuroendocr29(2008), 36–47[38] M. Dizonetal. Brain nitric oxide synthase levels increase in response to antenatal ethanol exposure

Alcohol & Alcoholism39 (2004), 101–105[39] Zimaetal.Oxidative Stress, Metabolism of Ethanoland Alcohol-Related DiseasesJ Biomed Sci8 (2001),

59-70[40] K. Yoshimoto et al. Pharmacological studies of alcohol susceptibility and brain monoamine function in

stroke-prone spontaneously hypertensive rats (SHRSP) and stroke-resistant spontaneously hypertensive rats (SHRSR). Tohoku J Exp Med201 (2003), 11-22.

[41] B. Hoskins et al. Effects of cocaine on tyrosine hydroxylase activity in brain areas from SHR and WKY, Brain Res Bull25(1990), 639-41.

[42] Da Silva et al. Effects of ethanol on locomotor and anxiety-like behaviors and the acquisition of ethanol intake in Lewis and spontaneously hypertensive rats, Life Sciences 77 (2005), 693-706

Page 282: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

280

Chapter 36

Gold Nanoparticles Cytotoxicity EvaluationIn HEP G2 Cells

Virginia TZANKOVAa,1,Maria KACHAMAKOVAa, Massimo VALOTIb,aMedical University of Sofi a, Faculty of Pharmacy, Dunav 2 Str. 1000 Sofi a, Bulgaria,

bDipartimento di Neuroscienze, Sezione di Farmacologia, Universitа di Siena, viale A. Moro 2, 53100 Siena, Italy.

AbstractGold nanoparticles are widely used in medicine, biomedical imaging and diagnostictests. Anti-infl ammatorytherapy, chrysotherapy,often relies on goldcomplexes. Gold(III) saltsattenuate antigen presentation and thus reduce autoimmunereactions in rheumatoid arthritis. Recent publications contain contradictory data regardingthe safety of gold nanoparticles. We conducted the in vitro cytotoxicitytesting of gold nanoparticles (GNPs) or gold nanoparticles coated with polyvinylpyrrolidone (PVP GNPs) of varying size, ranging from 5 to 40 nm. The cytotoxicity was assessed through lactate dehydrogenase releaseand glutathione levels. HEP G2 cell line was used to assess the impact of gold nanoparticles on the mammalian liver cells, following exposure to different concentrations and for duration from 24 h up to 72 h.The results from our experiments showed that naked GNPs of different sizes (5–40 nm) were toxic in a size-dependent manner on HepG2 cells incubated for up to 72 h. 5-nm naked GNPs promoted a signifi cant LDH release, while 15 nm and 40 nm gold nanoparticles does not affect LDH leakage. Coating with PVP decrease the cytotoxicity of 5-nm GNPs, as no statistically signifi cant effect on cell viability was observed.

Key words: gold nanoparticles, HEP G2, Nano toxicity

1. Introduction

Toxicity of nanoparticles (NPs) depends on many different factors: physical-chemical characteristics, the routes of administration or entry into the human body, the biological activity of NPs, and the target organs. According to several observations, liver is one of the main site where NPs can enter and accumulate, depending of their physical -chemical structure (i.e., gold, silver, silica NPs, quantum dots, and nanotubes) [1,2,3,4].

The ability of liver to take up NPs is well known. Kupffer cells are primarily involved in uptake; they are mainly capable to internalize NPs of various diameters and composition, depending of their coating [5,6,7]. However, the ability of hepatocytes to internalize NPs and, more specifi cally, gold nanoparticles (GNPs) is still debated. In spite of numerous

Page 283: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

281

toxicological studies carried out on the hepatoma cell line HepG2 [8], only a few in vitro studies using cell cultures have addressed attention to the ability of hepatocytes to internalize GNPs [9,10,11]. Athough NP preparations were consistently different in terms of functionalization, size, and chemical composition, NPs were taken up inside the cells in all cases. In addition, some GNPs were reported to get into the nuclei of HepG2 cells [11], but direct evidence was not given. It should be noted that all of these studies were conducted on 2D cell cultures [12].

Gold nanoparticles were variously described as nontoxic or toxic. Gold nanoparticles are used as tracers and the cellular trajectories change according to the biological signals added to the bulk material, again suggesting that gold particles by themselves are nontoxic [11]. Furthermore, oligonucleotide-modifi ed 13-nm gold particles were applied to intracellular gene regulation [13]. An entire anti-infl ammatory type of therapy, chrysotherapy, actually relies on gold complexes. A recent report elaborated that gold (III) salts attenuate antigen presentation and thus reduce autoimmune reactions in rheumatoid arthritis [14] which suggests a molecular mechanism for the anti-infl ammatory/anti-rheumatoid arthritis activity of gold drugs. Successful application of gold nanoparticles in biomedicine requires extensive safety assessment.

There are confl icting data in the literature suggesting that toxicity of GNPs appears to be related to the choice of the coating materials and/or to their size [15, 16]. In particular, Pan et al. [15]found that 1.4-nm GNPs capped with triphenylphosphine monosulfonate were much more cytotoxic than 15-nm GNPs capped with the same material. However, because 1.4-nm GNPs capped with GSH resulted markedly less cytotoxic, they concluded that the toxicity of small GNPs depends on their ability to trigger the intracellular formation of ROS from dioxygen [16].

The toxicological profi le of gold nanoparticles remains controversial. Signifi cant efforts to develop surface coatings to improve biocompatibility have been carried out. In vivo biodistribution studies have shown that the liver is a target for GNPs accumulation.Therefore, we investigated the effects on cell viability and oxidative stress, induced by non coated gold nanoparticles GNPs andGNPs coated withpolyvinilpyrolidone (PVP) in human HepG2 cells.

2. Materials and methods

2.1. Cell culture and cytotoxicity assay

Human hepatocellular carcinoma cell line, HEP G2, was purchased from American Type Culture Collection. It was cultured in RPMI medium (Gibco BRL, USA) supplemented with 10% fetal bovine serum (Gibco BRL, USA) and maintained at 37 ºC in a humidifi ed atmosphere of 5% carbon dioxide/95% air.

2.2. Measurement of GSH levelCellular GSH levels were analyzed using 5- chloromethylfl uorescein diacetate

(CMFDA, Molecular Probes) or the Glutathione Assay Kit (Sigma, St. Louis, MO). CMFDA is a membrane-permeable dye for determining levels of intracellular glutathione

Page 284: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

282

[17]. In brief, cells were incubated with GNPs and PVP-GNPs in different concentrations for 24 h. Cells were then washed in PBS and incubated with 5μM CMFDA at 37◦C for 30 min according to the instructions of the manufacturer. Cytoplasmic esterases convert nonfl uorescent CMFDA to fl uorescent 5-chloromethylfl uorescein, which can then react with the glutathione (Ex/Em = 522 nm/595 nm).

2.3. Measurement of LDH level

In order to evaluate cell viability, lactate dehydrogenase (LDH) leakage was determined after 24 to 72 h of incubation. LDH activity was assessed as previously described [18].

2.4. Statistical analysisAll experiments were performed at least in triplicate on three separate experiments.

Data are presented as means ± S.D. When a signifi cant effect was obtained with one-way ANOVA, Dunnett’s test was used to compare all values to the control.

3 Results and discussion

3.1. Gold nanoparticles characterization

Briefl y, TEM images indicated that the GNPs have an average diameter of 5 nm –40 nm [19]. TEM refl ection data and ICP-OES analysis confi rmed that the crystalline structure of GNPs was compatible with an Au lattice, and the percentage of Au present in GNPs resulted to be 99.1%. In some experiments GNPs, once had been coated with PVP in methanol, were dried and suspended, with sonication at nominal 0–500 μM concentration (as average number of Au atoms), corresponding to 0.98 and 98 μg gold Ч ml–1 water or culture medium. Dynamic light scattering showed that the average diameter of GNPs differed according to the suspension vehicle used, resulting 15.7 nm in methanol, 23.7 nm in water, and 10.9 nm in the culture medium, respectively. GNPs in these suspensions exhibited a great variance in the polydispersion index (PI) ranging from 0.02 in methanol to 0.298 in RPMI. Furthermore, 24-h incubation of NPs in RPMI did not affect their average diameter, whereas PI reached 0.330 value. Z-potential assay showed that GNPs were more dispersed in the cell culture medium (–9.74 mV), than in a water (–20.03 mV), probably owing to the solvation effect of proteins, as already reported [10].

3.2. Toxicity studies of gold nanoparticles3.2.1. Cell viability assayLDH is a sensitive and accurate marker for cellular toxicity. To investigate the

possible cytotoxicity induced by gold nanoparticles on human HEP G2, the amount of LDH released was evaluated. Figure 1 presents a comparative study of cytotoxicity of non-coated GNPs and GNPs, coated with polyvinylpyrrolidone (PVP- GNPs) [0.1 – 10 μM]. The cell viability was assessed after incubation from 24 to 72h. The results clearly show that after 24h exposure (Fig. 1A) gold nanoparticles induced statistically signifi cant LDH release in the human HEP G2 cells. Interestingly, the LDH released was signifi cantly high and comparable after short (24h) and long (48–72 h) exposure (Figure 1 A, B, C).

These data were in accordance with those reported by several authors demonstrating that naked GNPs are harmful to liver both in in vivo and in vitro conditions [20].

Page 285: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

283

Coradeghini et al. showed cytotoxic effects for GNPs 5 nm at concentration ≥ 50 μM in Balb/3T3 mouse fi broblasts exposed to the nanoparticles for 72 h [21]. Furthermore, gold nanoparticles were internalized by Balb/3T3 cells and located within intracellular endosomal compartments, with consequent disruption of the actin cytoskeleton.

In contrary, cell viability was not statistically signifi cant changed when coated with PVP GNPs were incubated with HEP G2 cells for up to 72h. Thus, coating with PVP decreased the cytotoxicity of GNPs, even in a concentration of 10 μМ for 72h incubation time (Fig. 1C). The results indicate that coated PVP-GNPs did not alter statistically signifi cant LDH-release in HEP G2 cell. Thus, our data supports the importance of the surface properties to increase the biocompatibility and safety of GNPs [22, 23].

3.2.2. GSH depletion

There are reports in the literature showing that the metal-containing nanoparticles may increase the production of reactive oxygen species (ROS) and consequently may lead to oxidative stress [24,25]. GNPs may possibly transport into the cell due to their small size (most range in size below 50 nm). Consequently, they can react with some components associated with regulation of cellular redox status and cell death. Cellular redox potential is largely determined by GSH, which accounts for more than 90% of cellular non-protein thiols [26]. One well-known reaction is GNPs possess strong Au–S bonding interaction with GSH [27]. Therefore, we reasonably presume that the GNPs may induce GSH depletion which results from the formation of thiolate occurring in the reaction between GNPs and GSH.

The effect of different concentrations (0.1-10 μM) of gold nanoparticles (5, 15 and 40 nm) on GSH content was studied (Fig. 2). GSH contents of the cells incubated with 40 nm GNPs (0.1-10 μM) were comparable to the controls (Fig. 2 C). The smaller sizes of GNPs of 5 nm (Fig. 2A) and 15 nm (Fig. 2B) caused dose-dependent decrease in GSH content (by 25 and 23 %, respectively, in a concentration of 10 μM). Thus our results confi rm that the size of GNPs is one of the most important factors determining their toxicity, and suggest that, at size of ≤ 15 nm, GNPs are slightly toxic and increasing GSH depletion in HEP G2 cells.

Conclusion

Gold has been used on numerous occasions to assess the biodistribution and cellular uptake of NPs following exposure. The primary site of gold particulate accumulation has been consistently demonstrated to be the liver, and it is therefore relevant that a number of in vitro investigations have focused on this potential target organ. However, in general there is a lack of in vivo and in vitro toxicity information that allows correlations between the fi ndings to be made [28].

Our results show that GNPs of different sizes (5–40 nm) were toxic in on HepG2 cells incubated for 24 h-72h. In particular, 5 nm GNPs caused a signifi cant LDH-release in HEP G2 cells. This observation confi rms the reports from the literature stating that gold and silver particle sizes are infl uential in dictating the observed toxicity, with smaller particles exhibiting a greater response than their larger counterparts, and this is likely to be driven by differences in particle surface area, when administered at an equal-mass dose. Our data

Page 286: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

284

showed the importance of the NPs coating on cell viability of human liver HEP G2 cells. Uncoated (naked) GNPs are more toxic than PVP-coated GNPs. While uncoated GNPs appear to increase LDH-leakage and to enhance oxidative stress in HEP G2 cells, PVP-GNPs showed no effects even in a higher concentration of 10 μM. Our fi ndings support the toxicity of GNPs as being size- and coating- dependent and provide additional insight on the health impact of GNPs.

Figure 1. Cell viability of GNPs treated cell cultures relative to the controls. HEP G2 cells were exposed to uncoated GNPs or coated with PVP GNPs in concentrations from 0.1 - 10 μМ for incubation periods 24, 48 and 72 h. LDH release was assessed in the culture medium as reported in Materials and Methods. Results are expressed as mean ± SEM of values obtained from 3 independent experiments. (*) indicate statistically signifi cantdifferences compared to the controls (p<0.05).

Page 287: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

285

Figure 2. GSH levels in HEP G2 cells in the presence of different sizes of GNPs. HEP G2 cells were incubated for 72 h in the presence of different concentrations of GNPs (0.1; 1.0; 10 μM) (panel A, B, and C). GSH content was measured as reported in the Materials and Methods. Results are expressed as mean ± SEM of values obtained in triplicate determinations. (*) indicate statistically signifi cant differences compared to the controls (p<0.05).

References

[1] S. K. Balasubramanian, Jittiwat, J., Manikandan, J., Ong, C., Yu, L. E., andOng, W. (2010).Biodistribution of gold nanoparticles and gene expressionchanges in the liver and spleen after intravenous administration in rats. Biomaterials31, 2034–2042.

[2] H. C. Fischer, Liu, L., Pang, K. S., and Chan, W. C. W. (2006). Pharmacokineticsof nanoscale quantum dots: In vivo distribution, sequestration, andclearance in the rat. Adv. Funct. Mater. 16, 1299–1305.

[3] R. Kumar, Roy, I., Ohulchanskky, T. Y., Vathy, L. A., Bergey, E. J., Sajjad,M., and Prasad, P. M. (2010). In vivo biodistribution and clearance studiesusing multimodal organically modifi ed silica nanoparticles. ACS Nano. 4,699–708.

[4] M. L. Schipper, Nakayama-Ratchford, N., Davis, C. R., Kam, N. W. S., Chu,P., Liu, Z., Sun, X., Dai, H., and Gambhir, S. S. (2008). A pilot toxicologystudy of single-walled carbon nanotubes in a small sample of mice. Nat.Nanotechnol. 3, 216–221.

[5] W. S. Cho, Cho, M., Jeong, J., Choi, M., Cho, H-Y., Han, B. S., Kim, S. H.,Kim, H. O., Lim, Y. T., Chung, B. H., et al. (2009). Acute toxicity and pharmacokineticsof 13 nm-sized PEG-coatedgold nanoparticles. Toxicol. Appl.Pharmacol. 236, 16–24.

[6] C. H. J. Choi, Alabi, C. A., Webster, P., and Davis, M. E. (2010). Mechanismof active targeting in solid tumors with transferrin-containing gold nanoparticles.Proc. Natl. Acad. Sci. U.S.A. 107, 1235–1240.

[7] E. Sadauskas, Danscher, G., Stoltenberg, M., Vogel, U., Larsen, A., and Wallin,H. (2009). Protracted elimination of gold nanoparticles from mouse liver.Nanomed: Nanotechnol. Biol. Med. 5, 162–169.

[8] N. Khlebtsov, and Dykman, L. (2011). Biodistribution and toxicity of engineeredgold nanoparticles: A review of in vitro and in vivo studies. Chem.Soc. Rev. 40, 1647–1671.

[9] H. J. Johnstone, Semmler-Behnke, M., Brown, D. M., Kreyling, W., Tran, L.,and Stone, V. (2010). Evaluating the uptake and intracellular fate of polystyrenenanoparticles by primary and hepatocytes cell lines in vitro. Toxicol.Appl. Pharmacol. 242, 66–78.

[10] J. Ponti, Colognato, R., Franchini, F., Gioria, S., Simonelli, F., Abbas, K.,Uboldi, C., Kirkpatrick, J., Holzwarth, U., and Rossi, F. (2009). A quantitativein vitro approach to study the intercellular fate of gold nanoparticles:From synthesis to cytotoxicity. Nanotoxicology 3, 296–306.

[11] A. G. Tkachenko, Xie, H., Liu, Y., Coleman, D., Ryan, J., Glomm, W. R.,Shipton, M. K., Franzen, S., and Feldheim, D. L. (2004). Cellular trajectoriesof peptide-modifi ed gold particle complexes: Comparison of nuclearlocalization signals and peptide transduction domains. Bioconjug. Chem. 15,482–490.

Page 288: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

286

[12] J. Lee, Lilly, G. D., Doty, R. C., Podsiadlo, P., and Kotov, N. A. (2009). In vitrotoxicity testing of nanoparticles in 3D cell culture. Small 10, 1213–1221.

[13] N. L.Rosi, D. A. Giljohann, C. S. Thaxton, A. K. Lytton-Jean, M. S. Han, C. A. Mirkin, Science 2006, 312, 1027.

[14] C.Wall, Painter, J. D. Stone, R. Bandaranayake, D. C. Wiley, T. J. Mitchison, L. J. Stern, B. S. DeDecker, Nat. Chem.Biol. 2006, 2, 197.

[15] Y. Pan, Leifert, A., Ruau, D., Neuss, S., Bornemann, J., Schmid, G., Brandau,W., Simon, U., and Jahnen-Dechent, W. (2009). Gold nanoparticles of diameter1.4 nm trigger necrosis by oxidative stress and mitochondrial damage.Small 5, 2067–2076.

[16] Y. Pan, Neuss, S., Leifert, A., Fischler, M., Wen, F., Simon, U., Schmid, G.,Brandau, W., and Jahnen-Dechent, W. (2007). Size-dependent cytotoxicityof gold nanoparticles. Small 3, 1941–1949.

[17] A.Macho, T. Hirsch, I. Marzo, P. Marchetti, B. Dallaporta, S.A. Susin, N. Zamzami, G. Kroemer, 1997. Glutathione depletion is an early and calcium elevation is a late event of thymocyteapoptosis, J. Immunol. 158 (10): 4612–4619.

[18] R. J. Gay, McComb, R. B., and Bowers, G. N. Jr. (1968). Optimum reactionconditions for human lactate dehydrogenase isoenzymes as they affect totallactate dehydrogenase activity. Clin. Chem. 14, 740–753.

[19] S. Dragoni,Franco, G., Regoli M., Bracciali, M., Morandi,V., Sgaragli,GP.,Bertelli,E., Valoti, M., Gold Nanoparticles Uptake and Cytotoxicity Assessed on Rat Liver Precision-Cut Slices 2012, Toxicol. Sci. 128(1), 186–197.

[20] S. Sabella, Galeone, A., Vecchio, G., Cingolani, R., and Pompa, P. P. (2011).AuNPs are toxic in vivo and in vitro: A review. J. Nanosci. Lett. 1, 145–165.

[21] R.Coradeghini ,Gioria S, Garcнa CP, Nativo P, Franchini F, Gilliland D, Ponti J, Rossi F. Size-dependent toxicity and cell interaction mechanisms of gold nanoparticles on mouse fi broblasts, 2013. Toxicol Lett. 13;217(3):205-16. doi: 10.1016/j.toxlet.2012.11.022. Epub 2012 Dec 13

[22] C.Uboldi , Bonacchi D, Lorenzi G, Hermanns MI, Pohl C, Baldi G, Unger RE, Kirkpatrick CJ. 2009. Gold nanoparticles induce cytotoxicity in the alveolar type-II cell lines A549 and NCIH441.Part Fibre Toxicol. 22;6:18

[23] T.Morais , Soares ME, Duarte JA, Soares L, Maia S, Gomes P, Pereira E, Fraga S, Carmo H, Bastos Mde L.2012. Effect of surface coating on the biodistribution profi le of gold nanoparticles in the rat.Eur J Pharm Biopharm. Jan;80(1):185-9.

[24] H.Y. Jia, Y. Liu, X.J. Zhang, L. Han, L.B. Du, Q. Tian and Y.C. Xu, Potential oxidative stress of gold nanoparticles by induced-NO releasing in serum, J. Am. Chem. Soc. 131 (2009), pp. 40–41.

[25] H. Li, Q. Li, X. Wang, K. Xu, Z. Chen, X. Gong, X. Liu, L. Tong and B. Tang, Simultaneous determination of superoxide and hydrogen peroxide in macrophage RAW 264.7 cell extracts by microchip electrophoresis with laser-induced fl uorescence detection, Anal. Chem. 81 (2009), pp. 2193–2198.

[26] http://www.sciencedirect.com/science/article/pii/S0378427411012185 - bbib0105Y.H. Han, S.H. Kim, S.Z. Kim and W.H. Park, Intracellular GSH levels rather than ROS levels are tightly related to AMA-induced HeLa cell death, Chem. Biol. Interact. 171 (2008), pp. 67–78.

[27] S.J. Chen and H.T. Chang, Nile red-adsorbed gold nanoparticles for selective determination of thiols based on energy transfer and aggregation, Anal. Chem. 76 (2004), pp. 3727–3734.

[28] HJ.Johnston , Hutchison G, Christensen FM, Peters S, Hankin S, Stone V. A review of the in vivo and in vitro toxicity of silver and gold particulates: particle attributes and biological mechanisms responsible for the observed toxicity. Crit Rev Toxicol 2010 Apr;40(4):328-46.

Page 289: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

287

Chapter 37

Pilot studies of pharmacological and toxicological effects of newly synthesized neuropeptides

with short chains

Presenting author: Stoeva S1.e-mail: [email protected], R. Кlisurov2, L.Tancheva1, S.Dragomanova3, T.Pajpanova4, R.Kalfi n1, A. Georgieva1

1Institute of Neurobiology 3Bulgarian Academy of Sciences,Sofi a1113 , “Acad. G. Bonchev”str. , bl.23

2 Medical Faculty,2Мedical University-Sofi a, Address: Sofi a1431, №1“G. Sofi iski” bld.

3 Medical faculty,2Мedical University-Varna, Address:Varna 9000, №55”Marin Drinov”str.

4 Bulgarian Academy of Sciences, 4Institute of Molecular Biology, Address: Sofi a 1113, “Acad. G. Bonchev”str, bl. 21

Abstract. Two new short-chain neuropeptides, synthesized by T. Pajpanova, (2013), analogues of Tyr- MIF– 1 and nociceptine are object of this research. The aim of the study is to investigate the basic pharmacological and toxicological effects of two new neuropeptides (P1 and P2) on laboratory rodents. Methods: Some basic toxicological characteristics of neuropeptides applied on Albino male ICR mice in several doses (5, 10, 20 and 50 mg/kg b. wt. i.p.) were studied. Their activity on the CNS was studied as well as their infl uence on the hexobarbital sleeping time (HB– 100 mg/kg b. wt. i.p.). We studied the infl uence of the two compounds on the nociception in mice (test with acetic acid) when effective doses (4, 8 and 16 mg/kg b. wt. i.p.) were applied. Results: The newly synthesized neuropeptides demonstrated several suppressive effects on the CNS when a dose of 50 mg/kg b. wt. was applied, which disappeared within 48 hours. There was no mortality after the acute treatment – both on the 48th hour (acute toxicity) and on the 5th day (prolonged toxicity). Pathological changes in the internal organs of treated animals were not found. P1 and P2 have similar effects on HB narcosis (P1 shortens it by 40%, and P2 by over 50%), but the mechanism is unknown. It is possible that the substances accelerate the elimination of HB or have modulating CNS effect related to their neuropeptidic nature. On the other hand the compounds had different effects on the nociception– P1 increases it (by 262%) and P2 has signifi cant analgesic effect (by over 25%). The analgesic effect of P2 is dose-dependent. We suggest that it is related to its nociceptine structure and due to possible interaction with CNS receptors.

Key words: toxicity, neuropeptides, hexobarbital, nociception

Page 290: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

288

IntroductionSearching for biologically active peptidomimetics as a new direction in modern

pharmacology requires complex, interdisciplinary researches. The modern drug design creates medicaments on the basis of well-known active peptides with improved pharmacokinetic properties (Miersh et al. 2000, Mateeva et al. 2011, Pancheva et al, 2003).

Objectof this study is two new short-chain neuropeptides, synthesized by T. Pajpanova, (2013), analogues of Tyr- MIF (P1) and Nociceptine (P2).

Our previous data found activity of similar compounds on central nervous system (CNS) (Encheva et al. 2013, Novoselski et al. 2013).

Having in mind their molecular design - very similar to some neurotransmitters in the central nervous system wedecidedtostudytheireffectsoncentral nervous system (CNS).

The aim of the study is to establish some basic pharmacological and toxicological effects of the new neuropeptides (P1 and P2) on laboratory mice.

1. Materials and Methods1.1. ChemicalsThe new compounds were synthesized by Pajpanova et al. (2013) in the Institute

of Molecular Biology at Bulgarian Academy of Sciences. Acetic acid and Hexobarbital sodium salt were provided by SIGMA-ALDRICH.

1.2. Amimals Albino male ICR mice (body weight 18–20 g) were supplied by ExperimentalBreeding

Base-Slivnitza at the Institute of Neurobiology (Bulgarian Academy of Sciences). Animals were housed in plexiglass cages (6 per cage), under standard laboratory conditions (ambient temperature 20°C ± 2°C and humidity 72 % ± 4 %), water and standard pelleted food ad libitum. All performed procedures were approved by the Institutional Animal Care Committee and the principles stated in the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientifi c Purposes (ETS123) were strictly followed throughout the experiment.

1.3. Methods1.3.1. Toxicological studiesSome basic toxicological characteristics of neuropeptides applied on Albino male ICR

mice (18-20 g) in several doses (5, 10, 20 and 50 mg/kg b. wt intraperitoneally- i.p.) were studied.

The observation on their effects and toxicity was till the 48thhour. The studies for prolonged toxicity continued till 5thday following changes in body growth, appetite and behavior. Dissection of the bodies was performed on the 48th hour and on the 5th day after compounds application.

1.3.2. Effects of the new compounds on CNSThe activity of the new compounds on the CNS was studied evaluating their infl uence

on the hexobarbital sleeping time (HB – 100 mg/kg b. wt.- i.p.). Hexobartal was used as

Page 291: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

289

central nervous active agent, but also as known modelsubstrate of hepatic cytochrome P-450 monooxygenases.

An effective dose 5 mg/ kg i.p. of new compounds was applied 20 minutes before Hexobarbital administration. The changes in duration of HB sleeping time (in minutes) were estimated in the groups according to the refl ex of reversal.

1.3.3. Studies for analgesic effectThe two compounds applied in an effective dose of 5 mg/kg were studied for analgesic

activity using Acetic acid test (HendershotandForsaith, 1959). The number of abdominal cramps for 20 minites after acetic acid application was measured. Dose-effect analgesic activity of compound P2 was studied in doses 4, 8 and 16 mg/kg b. wt. i.p. according to the same method.

1.4. Statistical analysisResults were performed using t-test of Student Fisher.

2. Result and discussionAcute ToxicityWe established low acute toxicity of the both compounds. On the 48th hour after the

application of the studied doses (over 50 mg/kg b. wt.) mortality was not observed. With an acute dose of 50 mg/kg b. wt. the compounds produced transit ataxia, respiration changes and sedation.We did not fi nd any changes in the body growth, appetite and behavior of animals after treatment with P1 and P2 (in doses 5, 10 and 20 mg/kg i.p.) compare to the controls.

Compounds are pharmacologically active when a dose of 5 mg/kg i.p. is applied.

Studies for prolonged toxicity- on 5th day after administration Prolonged toxicity was not established on the 5th day. Pathological changes in the

internal organs of the treated animals were not observed either.

Infl uence of compounds on the HB-sleeping timeSurprisingly we established in our experiments that P1 and P2 decreased duration

of HB narcosis (P1shortens it by 40%, and P2 by over 50%), but the mechanism is still unknown (Fig. 1).

It is possible that the new substances accelerate the elimination of HB via its hepatic metabolism. But we suggest that established drug interaction can be due predominately to functional antagonism between HB and new neuropeptides on the level of central nervous system and/or to receptor interactions. Only further experiments can clarify whether the mechanism of this interaction is on the metabolic level or on central nervous system level.

Analgesic effectWe established in our experiments that the new compounds had different effects on the

nociception– P1 increases it (by 262%) and P2 decreased it ( by over 25%).Established nociceptive effect of compound P1 is probably due to increased local

irritation in animals after combine administration of the compound with acetic acid.

Page 292: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

290

Figure 1. Effect of P1 and P2 on Hexobarbital sleeping time

Our experiments also demonstrated that compound P2 has a signifi cant analgesic effect- and this effect is dose-related (Fig. 2 and 3).

Figure 2. Dose dependent analgesic effect of P2 according

Acetic acid test (p<0.05)

The analgesic effect (% vs controls) of the 3 doses of compound P2- (4, 8 and 16 mg/kg b. wt. i.p.) was signifi cant and dose dependent in comparison to the control group (accepted for 100%)- Fig. 3.

The mechanism of the analgesic effect of P2 is not clear. We suggest that it may be related to the chemical structure of P2 wich is close to this of mediator nociceptine. Pain modulation probably is a result of possible interactions with some CNS receptors and deserves further experimental studies (Mateeva et al. 2011, Pancheva et al, 2003).

Page 293: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

291

Figure 3. Dose-dependent analgesic effect of P2 (% v/s controls) p<0.05

3. Conclusion

Newly synthesized neuropeptides demonstrated low toxicity and signifi cantly antagonized HB sleeping time in mice. Compound P2 also has a dose-dependent analgesic effect and as a derivative of nociceptine deserve further studies.

References

[1] Encheva, E., L. Tancheva, R. Klisurov, M. Novoselski, D. Tsekova, N. Belova, S. Stoeva, Pharmacological modulation of aggressive behavior in rats with new peptidomimetics European Conference Psychopharmacological Researches, 8-10 Octob., 2013, Barcelona, Spain

[2] Hendershot LC and J. Forsaith. Antagonism of frequency of phenylquinone- inducing writhing in the mouse by weak analgesics and nonanalgesics. Pharmacol. Exp. Ther., 1959, 125: 237-240

[3] Miersh, J., Grancharov, K., Pajpanova, T., Neumann, D., Tabakova, S., Stoev, S., Krauss, G-J. and Golovinsky, E., (2000), Amino Acids, 18 (1) 41-59.

[4] Mateeva, P., Zamfl rova, R., Pajpanova, T., Naydenova, E., Vezenkov, L. (2011) Changes in the biological activity of N/OFQ(1-13)NH2 after substitution of Lys9 by canavanine, Dab and Dap. C. rend. acad. bulg. Sci. 64 (11), 1645-1650..

[5] Novoselski, M , E. Encheva, R. Klisurov, Tancheva L, D. Tsekova, N. Belova, Sv. Stoeva Modulating effect of newly synthesized peptidomymetics over congnitive functions and their biochemical correlatons in Wistar rats, ICMS, May 2013,

[6] Pancheva, Svetlana, Efrossina Popgeorgieva, Rositsa Hristova, Tamara Pajpanova. (2003). Synthesis of several substituted at position 3 analogues of the naturally occurring peptide Tyr-MIF-1 (Tyr-Pro-Leu-Gly-NH2)”, In: Peptides 2002, Ettore Benedetti and Carlo Pedone (Eds.), Edizioni Ziino (Italy), pp 248-249.

Page 294: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

292

Chapter 38

The Procsses of Bioactivation, Toxicity and Detoxication – Experimental Models for Evaluation

of the Toxicity

Mitka MITCHEVAa1 ,Magdalena KONDEVA-BURDINAa

a Laboratory “Drug metabolism and drug toxicity”, Department of Pharmacology, Pharmacotherapy and Toxicology, Medical University-Sofi a,

Faculty of Pharmacy, Sofi a, BULGARIA

Abstract. Liver is the organ, where the main processes of biotransformation – deactivation, bioactivation and detoxication of xenobiotics – take place. The main experimental models for evaluation of these processes are: carbon tetrachloride (CCl4)-induced cytotoxicity (model of metabolic bioactivation) and tert-butyl hydroperoxide (t-BuOOH)-induced oxidative stress. Our studies are connected with evaluations of in vitro/in vivo harm effects of reactive metabolites, formed at metabolic level. We have investigated the possibilities and the conditions of detoxication and protection by different drugs and new perspective compounds from synthetic and natural origin. The results show that the used experimental models are suitable for evaluation of the bioactivation and detoxication processes.

Key words. Bioactivation, detoxication, protective and antioxidant effect

IntroductionThe biotransformation of drugs and other foreign chemicals may not always be an

innocuous biochemical event leading to detoxifi cation and elimination of the compound. Several compounds have been shown to be metabolically transformed to highly reactive metabolites, which can interact with vital intracellular macromolecules, resulting in toxicity –a process that is commonly referred to as metabolic activation or bioactivation.

The present study was conducted to confi rm this toxicological process of metabolic bioactivation and induced oxidative stress in experimental research and also to investigate the possible mechanism to detoxifi cation and protection by different drugs and new perspective compounds from synthetic and natural origin.

Carbon tetrachloride is widely used as a model of metabolic biotransformation in rats. Its toxic effects that led to liver injury are mainly due to a cytochrome P450-dependent biotransformation of CCl4 to free radicals. They initiate the process of lipid peroxidation, which is often the cause of inhibition of enzyme activity [1].

Page 295: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

293

The cellular system of energy supply localized in mitochondria is another target of many hepatotoxic substances causing oxidative stress and is one of the most important mechanisms through which hepatotoxic factors induced apoptotic and necrotic processes [2]. Tert-butyl hydroperoxide is a chemical with pro-oxidant activity. It has been used as a model of toxicity, on which metabolism of different compounds have been elucidated.

Based on the information available, the objective of the following study was to investigate the possible protective effect of Diosgenin, isolated from Astragalus offi cinalis, on a model of metabolic bioactivation (with CCl4) and Propofol on a model of oxidative stress (with tert-BuOOH) and the effects of Chlorpromazine and Metoprolol, drugs with cytochrome P450 – mediated metabolism, incubated alone, and in combination with Quinidine (specifi c CYP 2D6 inhibitor) and also with known protectors – Silymarin and Diosgenin [3].

1. Materials and methods1.1.AnimalsMale Wistar rats (body weight 200–250 g) were housed in plexiglass cages (3 per

cage), under standard laboratory conditions (ambient temperature 20°C ± 2°C and humidity 72 % ± 4 %) with free access to water and standard pelleted rat food. Animals were purchased from the NationalBreedingCenter, Sofi a, Bulgaria. All performed procedures were approved by the Institutional Animal Care Committee and the principles stated in the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientifi c Purposes (ETS123) were strictly followed throughout the experiment.

1.2. Isolation and incubation of hepatocytes

Rats were anesthetized with sodium pentobarbital (0.2 ml/100 g). In situ liver perfusion and cell isolation were performed as described by Fau, with modifi cations [4,5].

1.2.1. Lactate dehydrogenase release

Lactate dehydrogenase (LDH) release in isolated rat hepatocytes was measured as described by Bergmeyer [6].

1.2.2.Cell viability, GSH depletion and MDA assay

Cell viability and the levels of reduced glutathione (GSH) and malondialdehyde (MDA) were measured as described by Fau [4].

1.3. GSH depletion and MDA assay in liver homogenate

The levels of GSH depletion and MDA assay in liver homogenate were measured as described by Bai [7].

1.4. In vivo models of toxicity and protection

Carbon tetrachloride – 33 % solution in Ol. Helianthi p.o./ 1 time/ 90 min after the last application of the protector. 24 hours later the animal was killed [8].

Tert-butyl hydroperoxide – 1 mg/kg i.p./ on the 6 day (24 hours after the last application of the protector). 24 hours later the animal was killed [9].

Page 296: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

294

Silymarin ( antioxidant and hepatoprotector) – 20 mg/kg i.p./ once a day/ 3 days [10].Diosgenin, isolated from Asparagus offi cinalis – 50 mg/kg (121 μmol/kg) i.p./ 6

days [11].Propofol (local anaesthetic) – 11mg/kg i.v./ 5 days [12].

1.5. Ex vivo/ in vitro models of toxicity and application of inhibitor of CYP2D6

Carbon tetrachloride (86 μM) – model of bioactivation (solution with DMSO) [13].Tert-butyl hydroperoxide (75 μM) – model of oxidative stress (water solution) [9].Chlorpromazine (known antipsychotic) – 100 μM water solution [14].Metoprolol (selective β-blocker) – 100 μM water solution [15].Isolated rat hepatocytes were incubated with 25 μM Quinidine (as inhibitor) 15 min

before addition of the CYP2D6 substrate [16].

1.6. Statistical analysis

Statistical analysis was performed using statistical programme ‘MEDCALC’. Results are expressed as mean ± SEM for 6 experiments. The signifi cance of the data was assessed using the nonparametric Mann–Whitney test.

2. Results and DiscussionStrategies that have been used to minimize this risk are: blocking their metabolism and

also using of antioxidants.Diosgenin is known to have antioxidative effect in older patients, anti-

hypercholesterolememic and anti-infl ammatory effects [17]. According to these data it’s interesting to investigate the protective and antioxidant effect of Diosgenin, isolated from Asparagus offi cinalis, in conditions of metabolic-mediated toxicity, induced by carbon tetrachloride in vitro/in vivo.

In our experiments in isolated rat hepatocytes, CCl4 (86μM) had statistically signifi cant cytotoxic effects on some markers of toxicity. CCl4 decreased cell viability and GSH level by 64 % and 55 %, respectively; increased LDH leakage with 394 % and MDA level by 108 %, respectively, compared to the control.

In conditions of CCl4-induced toxicity, Diosgenin had statistically signifi cant cytoprotective effect. It preserved cell viability and GSH level by 91 % and 59 %, respectively; decreased LDH leakage and production of MDA by 54 % and 44 %, respectively. The effects were similar to those of Silymarin (Table 3).

Table 3. Effect of Diosgenin (Dg) and Silymarin (S) in CCl4-induced toxicity on cell viability, LDH leakage, GSH and MDA level in isolated rat hepatocytes

Group Cell viability (%)

LDH (μmol/min/mill cells)

GSH (nmol/mill cells)

MDA (nmol/mill cells)

Control 91 ± 5,4 0,121 ± 0,02 28 ± 4,3 0,102 ± 0,03

86 μM CCl4 33 ± 5,5 *** 0,598 ± 0,07 *** 12 ± 2,6 *** 0,395 ± 0,05 ***

Page 297: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

295

100 μM Dg + CCl4

63 ± 3,2 *** # # # 0,274 ± 0,02 ** # # 18 ± 3,2 ** # # 0,220 ± 0,04 * #

100 μM S + CCl4

60 ± 3,2 *** # # # 0,134 ± 0,02 # # # 15 ± 4,4 *** # 0,280 ± 0,04 * #

* p< 0,05; ** p < 0,01; *** p < 0,001 vs control#p< 0,05; # # p < 0,01; # # # p < 0,001 vs CCl4

According in vitro cytoprotective and antioxidant effects of Diosgenin, isolated from Asparagus offi cinalis, we investigate its’ in vivo effects.

In vivo administered alone, CCl4 decreased GSH with 56 % and increased MDA with 203 %, compared to the control. The combination between Diosgenin and CCl4, lead to statistically signifi cant decreased toxicity of the toxic agent – the level of GSH was preserved with 100 % and the level of MDA was decreased by 71 %. The effects were comparable to those of Silymarin (Table 4).

Table 4. In vivo effect of Diosgenin (Dg) and Silymarin (S) in CCl4-induced toxicity on GSH and MDA level in rat liver homogenate

Group GSH (nmol/mill cells) MDA (nmol/mill cells)

Control 18 ± 2,4 0,791 ± 0,8

33 % CCl4 8 ± 2,8 *** 2,4 ± 1,2 ***

50 mg/kg Dg + CCl4 16 ± 3,7 # # # 0,795 ± 2,5

20 mg/kg S + CCl4 16 ± 4,8 # # # 0,846 ± 3,7* #

* p< 0,05; *** p < 0,001 vs control#p< 0,05; # # # p < 0,001 vs CCl4

It is known that the toxicity of CCl4 is a result of metabolic bioactivation by the participation of CYP2E1, as well as CYP2B1 and possibly CYP3A isoforms [18].

The effects of Diosgenin were compared with those of Silymarin – a classical hepatoprotector and antioxidant. There are literature data that Silibin (the main compound in Silymarin) inactivate the human CYP3A4 and CYP2C9 [19]. The in vitro/in vivo effects of Silymarin in conditions of CCl4-induced hepatotoxicity, might be due to decrease of the activity of some isoforms of CYP450, which metabolized carbon tetrachloride.

In our previous experiments we found that Diosgenin is a substrate of CYP3A and in the same time increased the expression of this isoform, analyzed by Western blot analysis. These results correlate with the study of Kosters et al. (2005), who suggested that CYP3A play role in the metabolism of Diosgenin [20]. The protective effects of Diosgenin in vitro/in vivo on CCl4-mediated toxicity might be due to interference the activity of CYP3A, who play role in the carbon tetrachloride metabolism. The reduction of the CCl4 toxicity by Diosgenin may be also connected with the concurrent mechanism on metabolic level between both substrates.

Page 298: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

296

Base on these results, we investigate the protective effects of Diosgenin on the other examples of metabolic bioactivation, connected with other isoforms of cytochrome P450, like some substrates of CYP2D6 – as Chlorpromazine and Metoprolol metabolized to reactive metabolites, which lead to toxic processes in the cell [21].

CYP2D6 is one of the important human isoforms, which is not inducible and has genetic polymorphism, connected with good (E-extensive) and poor (P-poor) metabolizers.

Wistar rats are model of phenotype - extensive metabolizers in humans [22].Administered alone, Chlorpromazine and Metoprolol revealed statistically signifi cant

cytotoxic effects – decreased cell viability with 56 % and 59 %, respectively; reduced GSH level with 86 % both of the toxic agents; increased LDH leakage with 194 % and 199 %; MDA level – with 237 % and 286 %, respectively.

Quinidine is a selective inhibitor of CYP2D6. In combination with this inhibitor there was statistically signifi cant decrease of the cytotoxicity of Chlorpromazine and Metoprolol. The cell viability and GSH level were preserved with 62 % and 275 % (for Chlorpromazine) and with 66 % and 300 % (for Metoprolol); the LDH leakage and MDA level were decreased by 9 % and 57 % (for Chlorpromazine) and with 6 % and 61 % (for Metoprolol).

The studied drugs in combination with some protectors – as Diosgenin and Silymarin, statistically signifi cant decreased their toxic effects on isolated rat hepatocytes.

Diosgenin preserved cell viability and GSH level with 84 % and 250 % (for Chlorpromazine) and with 89 % and 250 % (for Metoprolol); the LDH leakage and MDA level were decreased by 16 % and 68 % (for Chlorpromazine) and with 27 % and 67 % (for Metoprolol).

Silymarin preserved cell viability and GSH level with 84 % and 250 % (for Chlorpromazine) and with 77 % and 200 % (for Metoprolol); the LDH leakage and MDA level were decreased by 26 % and 69 % (for Chlorpromazine) and with 16 % and 68 % (for Metoprolol) (Table 5).

Table 5. Effect of Chlorpromazine (Chl) and Metoprolol (M), administered alone, in combination with Quinidine (Q), Diosgenin (Dg), Silymarin (S) on cell viability, LDH leakage, GSH and MDA

level in isolated rat hepatocytes

Group Cell viability (%)

LDH (μmol/min/mill cells)

GSH (nmol/mill cells)

MDA (nmol/mill cells)

Control 85 ± 3,2 0,087 ± 0,02 28 ± 4,0 0,078 ± 0,03100 μM Chl 37 ± 6,3 *** 0,229 ± 0,03 *** 4 ± 2,1 *** 0,263 ± 0,05 ***

25 μM Q + Chl 60 ± 1,1 *** # 0,208 ± 0,02 *** # 15 ± 2,2 *** # # # 0,112 ± 0,004 * # # #

100 μM Dg + Chl 68 ± 3,1 ** # # 0,193 ± 0,02 *** # 14 ± 2,3 *** # # # 0,085 ± 0,02 # # #

100 μM S + Chl 68 ± 2,7 ** # # 0,169 ± 0,02 *** # # 14 ± 4,4 *** # # # 0,082 ± 0,04 # # #

100 μM M 35 ± 6,8 *** 0,233 ± 0,01 *** 4 ± 1,1 *** 0,301 ± 0,02 ***

25 μM Q + M 58 ± 1,1 *** # 0,218 ± 0,01 *** # 16 ± 3,3 *** # # # 0,116 ± 0,001 * #

100 μM Dg + M 66 ± 3,3 ** # # 0,169 ± 0,02 *** # 14 ± 2,2 *** # # # 0,100 ± 0,01 # # #

100 μM S + M 62 ± 2,7 ** # # 0,196 ± 0,03 ** # # 12 ± 2,2 ** 0,095 ± 0,03 # # #

* p< 0,05; ** p < 0,01; *** p < 0,001 vs control#p< 0,05; # # p < 0,01; # # # p < 0,001 vs chlorpromazine and metoprolol

Page 299: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

297

It’s known that hepatotoxicity of Chlorpromazine and Metoprolol is connected to the formation of reactive metabolites, which damage the membranes and cell structure [23]. In rat liver microsomes was found the role of different isoforms of CYP450 in Chlorpromazine metabolism, including CYP2D6 [24]. Later was found that Metoprolol was a substrate of CYP2D6 [25].

In combination with Quinidine (specifi c inhibitor of CYP2D6) the toxic effects of Chlorpromazine and Metoprolol on isolated rat hepatocytes decreased. These effects were connected with the inhibiting effect of Quinidine on the activity of CYP2D6, which play essential role in the metabolism of Chlorpromazine and Metoprolol.

The cytoprotective effects of Diosgenin on Chlorpromazine- and Metoprolol-nduced cytotoxicity might be due also to infl uence of their CYP-mediated metabolism and stabilization of hepatocytes membrane. In our previous experiments, we found that Diosgenin had antioxidant effects in conditions of enzyme- and non-enzyme-induced lipid peroxidation, comparable with known scavenger of free radicals – Promethazine [26]. We suppose that Diosgenin could lead to stabilization of the hepatocytes membrane by increasing the membrane fl uidity. Yamaguchi A et al. (2003) found that Diosgenin increased the fl uidity of the bile duct in conditions of toxicity [27].

Another model of toxicity, which mechanism is mitochondrial and microsomal connected, is tert-butyl hydroperoxide-induced oxidative stress [16].

Administered alone, tert-butyl hydroperoxide (75μM) had statistically signifi cant cytotoxic effect on isolated rat hepatocytes – decreased cell viability and GSH level by 68 % and 84 %, respectively; increased LDH leakage ten times and MDA level by 162 %, respectively, compared to the control.

Propofol is preferable anesthetic in cardio and neuro-surgery and in patients with hormonal problems, liver and kidney failure, chronic alcoholics [28]. There are literature data that in vitro Propofol had antioxidant and scavenging activity [29]. De la Cruz et al. (1998) found that Propofol have antioxidant activity in liver microsomes and liver mitochondria [30].

In our experiments we found that administered alone Propofol didn’t change cell viability and the level of GSH and MDA; it increased statistically signifi cant the LDH leakage with 83 %, compared to the control.

In conditions of tert-BuOOH-induced oxidative stress, Propofol revealed statistically signifi cant cytoprotective effect. It preserved cell viability and GSH level by 192 % and 424 %, respectively; decreased LDH leakage and production of MDA by 75 % and 56 %, respectively (Table 1).

Table 1. Effect of Propofol (P), administered alone and in combination with tert-BuOOH on cell viability, LDH leakage, GSH and MDA level in isolated rat hepatocytes

Group Cell viability (%)

LDH (μmol/min/mill cells)

GSH (nmol/mill cells)

MDA (nmol/mill cells)

Control 79 ± 2,3 0,105 ± 0,01 27 ± 3,2 0,117 ± 0,0375 μM tert-BuOOH 25 ± 3,1 *** 1,06 ± 0,4 *** 9 ± 5,5 *** 0,412 ± 0,06 ***

100 μM P 74 ± 6,0 0,192 ± 0,1** 22 ± 3,3 0,120 ± 0,05100 μM P + tert-BuOOH 73 ± 4,5 * # # # 0,267 ± 0,1 ** # # # 19 ± 4,5 * # # # 0,183 ± 0,06 * # # #

* p < 0,05; ** p < 0,01 ; *** p < 0,001 vs control# # # p < 0,001 vs tert-BuOOH

Page 300: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

298

In vivo administered alone, Propofol didn’t change statistically signifi cant the level of GSH and MDA, while tert-BuOOH decreased GSH with 26 % and increased MDA with 56 %, compared to the control. The combination between Propofol and tert-BuOOH, lead to statistically signifi cant decreased toxicity of the toxic agent – the level of GSH was preserved with 23 % and the level of MDA was decreased by 56 % (Table 2).

Table 2. In vivo effect of Propofol (P), administered alone and in combination with tert - BuOOH on GSH and MDA level in rat liver homogenate

Group GSH (nmol/mill cells) MDA (nmol/mill cells)Control 19 ± 3,7 0,666 ± 0,91 mg/kg tert-BuOOH 14 ± 7,5 * 2,7 ± 2,3 *

11 mg/kg P 18 ± 8,3 0,685 ± 3,4P + tert-BuOOH 17 ± 9,5 * # 0,764 ± 1,2 # # #

* p < 0,05 vs control# p < 0,05; # # # p < 0,001 vs tert-BuOOH

The metabolism of tert-BuOOH to free radicals undergoes through several steps. In microsomal suspension, in the absence of NADPH, it has been shown to undergo one-electron oxidation to a peroxyl radical (reaction 1), whereas in the presence of NADPH it has been shown to undergo one-electron reduction to an alkoxyl radical (reaction 2). In isolated mitochondria and intact cells, the t-BuOOH has been shown to undergo β-scission to the methyl radical (reaction 3). All these radicals cause lipid peroxidation process [31, 32].

(CH3)3COOH → (CH3)3COO• + e- + H+ (reaction 1)(CH3)3COOH + e- → (CH3)3CO• + OH- (reaction 2)(CH3)3CO• → (CH3)2CO + •CH3 (reaction 3)

The protective effects of Propofol in vitro/in vivo in conditions of oxidative stress, might be due to its structure, which is similar to those of vitamin E – known antioxidant and membrane stabilizator [12]. Propofol inhibit lipid peroxidation by forming a non-radical molecule that disrupts the chain of formation of other free radicals potentially able to cause greater damage to cell membranes [30]. Our results correlate with literature data about antioxidant effects of Propofol.

3. ConclusionsIsolated hepatocytes are convenient cell system, which allow the reading of toxicity

markers as result of biotransformation. In isolated rat hepatocytes, administered alone, the toxic agents: carbon tetrachloride and tert-butyl hydroperoxide, lead to cytotoxic effects, which are results of their metabolism and induced oxidative stress. The toxicity was proved by the experiments in vivo. Chlorpromazine and Metoprolol, drugs – substrates of CYP2D6, in experiments in vitro, revealed also metabolites-mediated cytotoxicity, which decreased in combination with selective inhibitor of CYP2D6. Our results proved metabolite-mediated toxicity.

The using of different models of intoxication in vitro/in vivo gives the opportunity to be revealed the possible mechanisms of cytoprotective and antioxidant activity of Diosgenin,

Page 301: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

299

isolated from Asparagus offi cinalis and of the drug Propofol. The effects were similar to those of the fl avoniod Silymarin – a classical hepatoprotector and antioxidant, which had also CYP450 – mediated metabolism.

4. AcknowledgementsThe authors would like to thank the Medical Science Committee for funding this

research (Project 32/2005) through Concurs Grant 2005.

References

[1] M. Ferreira, O. Nunes, L. Leal, O. Pessoa, T. Lemos, G. Viana, Antioxidant effects in the quinine fraction from Auxemma oncocalyx Taub., Biol Pharm Bull26 (2003), 595-599.

[2] G. Kroemer, B. Dallaporta, M. Resche-Rigon, The mitochondrial death/life regulation in apoptosis and necrosis, Annu Rev Physiol60 (1998), 619-642.

[3] M. Mitcheva, M. Kondeva-Burdina, I. Krasteva, S. Nikolov, In vitro assessment of effect of Diosgenin, isolated from Asparagus offi cinalis, on spontaneously hypertensive ( SHR) and old rats, Arch Bal Med Union44(2) (2009), 137-141.

[4] D. Fau, A. Berson, D. Eugene, B. Fromenty, C. Fisch, D. Pessayre, Mechanism for the hepatotoxicity of the antiandrogen, nilutamide. Evidence suggesting that redox cycling of this nitroaromatic drug leads to oxidative stress in isolated hepatocytes, J Pharmacol Exp Ther263(1) (1992), 69-77.

[5] M. Mitcheva, M. Kondeva, V. Vitcheva., P. Nedialkov, G. Kitanov, Effect of benzophenones from Hypericum annulatum on carbon tetrachloride-induced toxicity in freshly isolated rat hepatocytes, Redox Report11(1) (2006), 1-8.

[6] H. U. Bergmeyer, K. Gawehn, M. Grassl, Methods of Enzymatic Analysis. Ed. By H. U. Bergmeyer , Verlag Chemie, Weinheim, vol.1 (1974), 481-482.

[7] X. Bai, A. Qiu, J. Guan, Z. Shi, Antioxidant and protective effect of an oleanolic acid-enriched extract of A. deliciosa root on carbon tetrachloride induced rat liver injury, Asia Pac J Clin Nutr16 (2001), 169-173.

[8] K. Raju, G. Anbuganapathi, V. Gokulakrishnan, B. Rajkapoor, B. Jayakar, S. Manian, Effect of dried fruits of Solanum nigrum Linn against CCl4-induced hepatic damage in rats, Biol Pharm Bull26 (2003), 1618-1619.

[9] F. Takayama, T. Egashira, Y. Yamanaka, Protective effect of Ninjin-yoei-to on damage to isolated hepatocytes following transient exposure to tert-butyl hydroperoxide, Jpn J Pharmacol85 (2001), 227-233.

[10] D. Radovanovic, D. Jovanovic, D. Mihailovic, G. Rankovic, N. Stojiljkovic, V. Dimitrov, Hepatoprotective effects of Silymarin in androgenic-anabolic steroid-induced liver damage, Med Pregl56 (2003), 79-83.

[11] M. Aragno, E. Brignardello, E. Tamagno, V. Gatto, O. Danni, G. Boccuzzi, Dehydroepiandrosterone administration prevents the oxidative damage induced by acute hyperglycemia in rats, J Endocrinology155 (1997), 233-240.

[12] L. Aarts, R. Van der Hee, I. Dekker et al., The widely used anesthetic agent Propofol can replace alpha-tocopherol as antioxidant, FEBS Lett357(1) (1995), 83-85.

[13] M.U. Dianzani, G. Poli, E. Gravela, E. Chiarpotto, E. Albano, Infl uence of lipid peroxidation on lipoprotein secretion by isolated hepatocytes, Lipids16 (1981), 823-829.

[14] W. A. Daniel, A. Haduch, J. Wojcikowski, Inhibition of rat liver CYP2D in vitro and after 1-day and long-term exposure to neuroleptics in vivo-possible involvement of different mechanisms, Eur Neuropsychopharmacol15(1) (2005), 103-110.

[15] A. Kovacs, J. Magyar, T. Banyasz, P. P. Nanasi, G. Szenasi, Beta-adrenoreceptor activation plays a role in the reverse rate-dependancy of effective refractory period lengthening by dofetilide in the guinea-pig atrium in vitro,Br J Pharmacol139(8) (2003), 1555-1563.

[16] U. Fuhr, S. Kober, M. Zaigler, E. Mutschler, H. Spahn-Langguth, Rate-limiting biotransformation of tramterene is mediated by CYP1A2, Int J Clin Ther43(7) (2005), 327-334.

Page 302: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

300

[17] A. Mohsen, M. E. Omar, O. M. Aboulwafa, I. M. Labouta, A. A. El-tombary, A. I. Mallah, 4’, 17-dioxo-5’H-estra-1(10), 4-dieno[3, 2-b] furan: sнntesis, binding affi nity to the estrogen receptor, uterotrophic and antiimplantation activities, Arch Pharm329 (1996), 61-65.

[18] L. W. Weber, M. Boll, A. Stampfl , Hepatotoxicity and mechanism of action of haloalkanes: Carbon tetrachloride as a toxicological model, Crit Rev Toxicol33 (2003), 105-136.

[19] C. Sridar, T. C. Goosen, U. M. Kent, J. A. Williams, P. F. Hollenberg. Silybin inactivates cytochromes P450 3A4 and 2C9 and inhibits major hepatic glucuronosyltransferases, Drug Met Dispos,32 (2004), 587-594.

[20] A. Kosters, R. J. J. Frijters, C. Kunne, E. Vink, M. S. Schneiders, F. G. Schaap, C. P.Nibbering, S. B. Patel, A. K. Groen, Diosgenin-induce biliary cholesterol secretion in mice requires Abcg8, Hepatology41 (2005), 141-150.

[21] D. Zhang, J. P. Freeman, J. B. Sutherland, A. E. Walker, Y. Yang, C. E. Cerniglia, Biotransformation of Chlorpromazine and Methdilazine by Cunninghamella elegans,Apple Environ Microbiol62(3) (1996), 798-803.

[22] T. Kraemer, T. Pfl ugmann, M. Bossmann, N. M. Kneller, F. T. Peters, L. D. Paul, D. Springer, R. F. Staack, H. H. Maurer. Fenproporex N-dealkylation to amphetamine-enantioselective in vitro studies in human liver microsomes as well as enantioselective in vivo studies in Wistar and Dark Dark Agouti rats, Biochem Pharmacol68(5) (2004), 947-957.

[23] M. D. Johnson, G. Newkirk, J. R. White Jr., Clinically signifi cant drug interactions, Postgrad Med105(2) (1999), 193-195, 200, 205-6 passim.

[24] H. B. Niznik, R. F. Tyndale, F. R. Sallee, F. J. Gonzalez, J. P. Hardwick, T. Inaba, W. Kalow, The dopamine transporter and cytochrome IID1 (debrisoquine 4-hydroxylase) in brain: resolution and identifi cation of two distinct [3H]GBR-12935 binding proteins, Arch Biochem Biophys276(5) (1990) 424-432.

[25] T. Hiroi, S. Imaoka, D. Funae, Dopamine formation from tyramine by CYP2D6, Biochem Biophys Res Commun249(3) (1998), 838-843.

[26] M. Mitcheva, Mechanisms of toxicity – possibilities of infl uence, ECO-RING, Sofi a (2008).[27] A. Yamaguchi, S. Tazuma, H. Ochi, K. Chayama, Choleretic action of Diosgenin is based upon the

increases in canicular membrane fl uidity and transporter activity mediating bile acid independent bile fl ow, Hep Res25 (2003), 287-295.

[28] I. Gulcin, H. A. Alici, M. Cesur, Determination of in vitro antioxidante and radical scavenging activities of Propofol, Chem Pharm Bull53(3) (2005), 281-285.

[29] J. P. De la Cruz, G. Senedo, J. A. Carmona, F. Sanchez de la Cuesta, The in vitro effects of Propofol on tisular oxidative stress in the rat, Anesth Analg87 (1998), 1141-1146.

[30] K. Ollinger, U. T. Brunk, Cellular injury induced by oxidative stress is mediated through lysosome damage, Free Radic Biol Med 19 (1995), 565-574.

[31] H. A. Bae Jin, J. Y. Lee, The effect of chondroitin sulfate against CCl4-induced hepatotoxicity, Biol Pharm Bull26 (2003), 622-626.

[32] V. O’Donnell, M. J. Burkit, Mitochondrial metabolism of a hydroperoxide to free radicals in human endothelial cells: An electron spin resonance spin-trapping investigation, Biochem J304 (1994), 707-713.

Page 303: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

301

Chapter 39

OPRD1 Polymorphism is not Associated with Chronic Cocaine Use

Elena VAKONAKIa, Olympia MANOLIa, Leda KOVATSIb, Mary MANTSIc, Eirini DIAMANTARAa, Stamatios BELIVANISa, Manolis TZATZARAKISa

and Aristidis M. TSATSAKISa,1

aCenter of Toxicology Science and Research, School of Medicine, University of Crete, Voutes 71003, Heraklion, Greece,

bLaboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124

cGreek National Organization Against Drugs, Athens, Greece

Abstract. Drug dependence is infl uenced to a great extent by genetic factors. We have currently focused on the highly polymorphic OPRD1 gene and the rs678849 polymorphism in chronic cocaine users. Urine and nail samples were collected from 73 individuals (33 chronic cocaine users and 40 non cocaine-users), following written informed consent. The drug users were in the process of detoxifi cation, following either a buprenorphine substitution program, or a “dry” rehabilitation program. All samples were analyzed by LC-MS for cocaine and benzoylecgonine. DNA was extracted from the nails and specifi c primers were designed for the recognition of the OPRD1 selected polymorphism, which was detected by PCR-RFLP.Our results showed that there is no signifi cant relationship between the rs678849 SNP alleles and cocaine use. For the rs678849 T allele the p- value was 0.580 and the odds ratio (Cl) 1.346(0.468-3.871), while for the C allele the p-value was 0.761 and the odds ratio (Cl) 0.870(0.353-2.142). Previous studies have shown that there is a statistically signifi cant difference in the American populations for the T allele of the rs678849 SNP between cocaine users and non-users, while in the European population there is no statistically signifi cant difference in the frequency of this allele between users and non-users. In conclusion, studies so far revealed that the rs678849 T allele is associated with cocaine dependence only in American populations and not in Europeans.

Key words. OPRD1, cocaine, dependence, drug users

1. IntroductionCocaine (benzoylmethylecgonine) is an alkaloid that consists of a lipophilic, a

hydrophilic and an aliphatic group. Cocaine is rapidly metabolized by cholinesterases to the ecgonine methyl ester, an inactive metabolite. Benzoylecgonine, the other major inactive

1 Corresponding Author:Prof. Tsatsakis Aristidis, Laboratory of Toxicology, University of Crete, Voutes 71003, Heraklion, Greece, e-mail: [email protected], tel: ++30 2810 394679

Page 304: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

302

metabolite of cocaine, is formed from the non-enzymatic hydrolysis which occurs in the blood. Norcocaine, is the active metabolite of cocaine. It is formed by an N-demethylation reaction and it represents less than 5% of the total cocaine metabolites [1].

Cocaine is a central nervous system stimulant with two properties at the cellular level. By interfering with the sodium channel, it acts as a local anesthetic. Furthermore, it has an effect on neuron receptors. Cocaine blocks norepinephrine, serotonin, dopamine and other neurotransmitters from being reabsorbed [2]. Cocaine abuse increases excitatory neurotransmitter levels in the brain. Therefore, it induces euphoria in the short term and addiction in the long term [3].

Activation of δ-opioid receptors (DOR) is known to regulate drug reward, inhibitory controls and learning processes and their dysfunction contributes to the development of addiction. Moreover, the δ-opioid receptor of the amygdala- cortico- hippocampal circuitry controls emotional responses. DOR plays a regulatory role in drug intake as well as in drug seeking and dependence issues, which vary according to the drug [4]. OPRD1 ( human δ-opioid receptor) is localized on chromosome 1p34 [5] and is known to encode the δ-opioid receptor, a group of G protein-coupled receptors, that are natural targets for a family of endogenous opioid peptides such as dynorphins, enkephalins, endorphins and nociseptin [6].

These G protein-coupled receptors are activated by ligands that bind to a receptor’s domain outside the cell. There are three major subtypes of opioid receptors, based on the fi rst ligand that was found to bind to them; δ from mouse vas deferens, κ from ketocyclazocine and μ from morphine. Each one of them has further subtypes, different locations and roles [7]. As a result, an intracellular domain of the receptor activates a G protein, which further activates signaling events. The structure of G proteins is crucial for the function and the signaling pathways, as they act as molecular switches by binding to and hydrolyzing GTP to GDP. When they bind to GTP, they are activated but not when they bind to GDP. As a result, they exchange GDP for GTP and fi nally they dissociate to activate other proteins in the opioid-mediated signal transduction. Generally, GPCR internalization, traffi cking and redistribution are the key mechanisms of the regulatory responses. The internalized G protein-coupled receptors can be either recycled to the cell surface or processed to endocytic pathways. Specifi cally, δ-opioid receptors are targeted for lysosomal degradation via the endosomal sorting complex required for transport (ESCRT) machinery using ubiquitination-dependent or independent mechanisms [8,9,10].

It has been shown that the pharmacological blockade of δ-opioid receptors reduces the rewarding properties of cocaine. Infusion of δ-opioid agonists, antagonists or enkephalinase inhibitors in specifi c brain sites, alter cocaine self-administration or cocaine-seeking behavior. As a result, even though a number of studies have shown that delta antagonists do not affect the rewarding properties of cocaine, there is evidence suggesting that δ-opioid receptors infl uence the rewarding and addictive effects of phychostimulants [11,12]. It is known that cocaine dependence is infl uenced to a great extent by genetic factors. We have currently focused on the highly polymorphic OPRD1 gene and its polymorphism rs678849 in chronic cocaine users.

Page 305: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

303

1. Materials and methods

1.1. Study subjectsA total of 73 individuals participated in this study (European population). Forty (40)

individuals were non-users (control group) and thirty three (33) individuals were ex- cocaine users. All ex- cocaine users were in the process of detoxifi cation, following either a buprenorphine substitution program, or a “dry” rehabilitation program. Urine, blood and nail samples were collected from the subjects. Twenty eight (28) urine, fourteen (14) blood and thirty one (31) nail samples were collected and used in the present study. Twelve (12) urine samples and twenty one (21) nail samples were obtained from ex- cocaine users, while the remaining samples were collected from non- cocaine users. Urine and blood samples were stored at 4oC and nails samples were stored at room temperature until DNA extraction. The study was approved by the Ethics Committee of the University of Crete and a written informed consent was obtained from all participants.

1.2. DNA extractionAs previously mentioned, nail samples were used as the source of DNA. The

QuickGene DNA tissue kit S was used for DNA extraction. Briefl y, ten mg of nails were washed with 100% ethanol and then purifi ed with water, cut into slices and placed in a 1.5 ml micro tube. 200 μl of 2.3M DTT (dithiothreitol) solution were added and the samples were incubated at 55o C overnight. Following centrifugation at 5000 rpm, at room temperature, the supernatant (DTT) was removed. A volume of 180 μl and 20 μl of MDT and EDT respectively, were added and the samples were further incubated overnight at 55oC. The supernatant was subsequently transferred into a new 1.5 ml tube, 180 μl of LDT were added and the samples were mixed by vortexing for 15 seconds. Another incubation at 70oC for 10min was then performed. A volume of 240 μl 99% ethanol was added, the samples were mixed by vortexing and were ready for the use of the QuickGene-Mini 80 Workfl ow in order to elute 50 μl of genomic DNA.

DNA was also extracted from urine and blood samples using the QuickGene kit. The samples were centrifuged at 5000 rpm, at room temperature for 2 min and the supernatant was removed in order to keep the leukocytes. EDTA (200 μl) and 10μl of EDT were added and the samples was slowly shaken for 2 hours at 55oC. LDT (180 μl) was added and the samples were then incubated at 70oC for 10 min. A volume of 240 μl of 99% ethanol was added before the use of the QuickGene-Mini 80 Workfl ow in order to elute 50μl of genomic DNA.

The DNA concentration and purity were measured with the ND-10006 spectrophotometer (NanoDrop Technologies, Wilmington, DE).

1.3. PCR- RFLP

The rs678849 polymorphism of OPRD1 was studied. PCR primers were especially designed for this purpose (Table 1).

Table 1. Primers for the amplifi cation of the selected sequence.

rs678849 Reverse primer Forward primer

5’-AGGAAAGGCGTCGTAAGCAG-3’ 5’-GTATCTCCTCAGGAATCATG-3’

Page 306: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

304

The DreamTaq Green PCR Master Mix (DreamTaq Green buffer, DreamTaq DNA polymerase, 0.4 mM of dATP, dCTP, dTTP, dGTP and 4 mM MgCl2) from Thermo Scientifi c was used in amplifaction. The cycling conditions were: initial denaturation at 95°C for 5 min, followed by 35 cycles of denaturation at 95°C for 30 sec, annealing at 58°C for 30 sec and extension at 72°C for 30 sec. Our products were analyzed on 2% (w/v) agarose gels, stained with ethidium bromide and photographed on an AlphaImager™ (Alpha Innotech, Santa Clara, CA) UV transilluminator.

The PCR product was digested using the Rsa1 restriction enzyme from Thermo Scientifi c, which binds and subsequently cleaves the following sites:

5’...G T→A C...3’3’...C A→T G...5’

Two μl of Buffer Tango, 1.5 μl Rsa1 and 12 μl water were added to 10 μl of our PCR product. The samples were then incubated at 37oC for 3 hours and the resulting DNA fragments were separated on a 2.5% agarose gel.

1.4. Cocaine detection Urine and blood samples were analyzed for the presence of cocaine and its metabolites

with FPIA (Fluorescence Polarization Immuno Assay) on an Abbott Diagnostics System (AxSYM), as well as liquid chromatography-mass spectrometry (LC/MS). Briefl y, in one ml of urine, 400 ng of internal standard (imipramine d3) and 20 μl β-glucuronidase were added. Spiked urine samples were prepared and used for the preparation of the spiked curve. Inallsamples (spikedandactualurine samples) 1 mlof phosphate buffer pH=7.0 was added and the samples were incubated for 45 minat 56oC. The clean up step was performed with solid phase extraction (SPE). The columns (1 ml, 100mg) were preconditioned with 1 ml of methanol and phosphate buffer, rinsed with 1 ml phosphate buffer and dried under full vacuum. The analytes were eluted with 0.8 ml 1% ammonia in methanol. The organic phase was evaporated to dryness under a stream of N2 and reconstituted in 0.4 ml of methanol. Cocaine and its metabolites were extracted from blood samples by liquid-liquid extraction with choroform-ethyl acetate (1:1).

Analysis was performed with liquid chromatography-mass spectrometry. A gradient of 0.1% formic acid in water (solvent A) and acetonitrile (solvent B) was selected as the mobile phase at a fl ow rate of 0.6 ml/min. A mass spectrometer with atmospheric pressure chemical ionization (APCI) was used to detect and quantify all analytes. Ion signals were acquired in time selected ion monitoring (SIM) mode with ions m/z = 290.15 for BE, 304.20 and 182.05 for cocaine and 284.20 and 222.10 for IS (imipramine d3)

1.5. Statistical analysisThe allelic association of this SNP with cocaine dependence was determined by using

the Chi-square. Deviation from Hardy-Weinberg was assessed in the total population, as well as separately for cases and controls. The SNP across the set of samples was in Hardy-Weinberg Equilibrium (p ≥ 0.05).

Page 307: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

305

2. Results

All analyzed urine and blood samples were negative for cocaine and BE both by FPIA, as well as by LC/MS.

In the present study, the rs678849 polymorphism was examined using RFLP- PCR in 73 participants, 33 ex- cocaine users and 40 controls. Our results showed that 23% of the participants were homozygotes for the polymorphism, 13% were homozygotes for the non polymorphic allele and the remaining 54% were heterozygotes for the examined polymorphism. Our results showed that there is no signifi cant relationship between the rs678849 SNP alleles and cocaine use. For the rs678849 T allele the p- value was 0.580 and the odds ratio (Cl) 1.346 (0.468-3.871), while for the C allele the p-value was 0.761 and the odds ratio (Cl) 0.870(0.353-2.142).

3. Discussion

The present study focused on the association of Oprd1 polymorphism with chronic cocaine use in European populations. The rs678849 SNP showed no association with cocaine dependence and no signifi cant association with chronic cocaine use. The two alleles of rs678849, T and C, neither seemed to be over-presented in the set of groups. The statistical analysis demonstrated that for the T allele the p- value was 0.580, while for the C allele the p-value was 0.761, indicating that there is no statistically signifi cant difference between the controls and the cases.

Gelernter and Kranzler [13], sequenced the OPRD1 coding region in six individuals with differing T921C alleles and identifi ed one novel variant in exon 1, 80T→G, which predicts a change in the amino acid sequence from phenylalanine (80T) to cysteine (80G) (F27C). They also demonstrated a signifi cant heterogeneity and different allele frequencies amongst Japanese and Europeans of about 0 and 0.09 respectively, and the frequency of the allele 921C, in African-Americans and Japanese with values 0.34 and 0.82 respectively. They also proposed that these differences, between people, could affect the receptor function and therefore might result in every condition related with delta opioid receptors.

Eight years later, Zhang et Al. [14] demonstrated the existence of an association between a 6 SNP haplotype and alcohol, cocaine and opioid dependence in 1506 European-American subjects. In this case it was shown that the allele 80G at rs1042114 was more frequent in opiate addicts (21%) compared to controls (13.2%). The frequency of the allele varied in different populations (it was 0.09 in European-American populations and 0.03 in African-American populations) but the polymorphic allele was absent in Japanese and Chinese populations.

Crist et.al [15] showed a strong association between the rs678849 polymorphism and cocaine addiction in the African-American populations. The samples consisted of 553 European-American and 503 African-American cocaine-addicted subjects, while 656 European-American and 503 African-American individuals served as the control group. They analyzed this SNP in a larger independent population of cocaine addicted African Americans and controls and the association was confi rmed, allelic p = 4.53 Ч 10-5. The T allele was over-represented in the controls compared to cases, indicating a protective role. Moreover, they proposed that the surrounding intronic region of rs678849 is of a great interest in addiction.

Page 308: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

306

Our results demonstrate that rs678849 is only related with cocaine dependence in African-American and not in European populations and are in agreement with the fi ndings of Crist et al. [15] who reported that rs678849 is related to cocaine use only in African-Americans. The reason for this is unknown, implicating that more research is required towards that direction.

References

[1] L.R. Goldfrank, R.S. Hoffman. The cardiovascular effects of cocaine, Ann Emerg Med20 (19910, 165-175.

[2] W.H. Spivey, B. Euerle. Neurologic complications of cocaine abuse.Ann Emerg Med19 (1990), 1422-1428.

[3] S.M. White, C.J.T. Lambe. The pathophysiology of cocaine abuse.J Clin Foren Med10 (2003), 27–39.[4] P.C.S. Chung, B.L. Kieffer. Delta opioid receptors in brain function and diseases.Pharmaco & Therap140

(2013), 112-20[5] P. Franke, M.M. Nothen, T . Wang, et al. Human δ-Opioid Receptor Gene and Susceptibility to Heroin and

Alcohol Dependence.Am J Med Genet (Neuropsych Genet )88 (1999), 462–464.[6] F. Simonin, K. Befort, C. Gaveriauxrute, H. et al. The Human b-Opioid Receptor: Genomic Organization,

cDNA Cloning, Functional Expression, and Distribution in Human Brain. Molecul Pharmac46 (1994), 1015-1021.

[7] R.M. Quock, T.H. Burkey, E. Varga, et al.The δ-Opioid Receptor: Molecular Pharmacology,Signal Transduction, and the Determination ofDrug Effi cacy. Pharmac Reviews51 (1999), 503-532

[8] A.A. Pradhan, A.J.Becker, G. Scherrer, et al. In vivo delta opioid receptor internalization controls behavioral effects of agonists. PLoS ONE4 (2009), e5425 DOI: 10.1371/journal.pone.0005425

[9] A.G. Henry, I.J. White, M. Marsh, M. von Zastrow, J.N. Hislop.The role of ubiquitination in lysosomal traffi cking of delta-opioid receptors.Traffi c12 (2011), 170–184.

[10] A.A. Pradhan, K. Befort, C. Nozaki, C. Aaveriaux- Ruff and B.L. Kieffer. The delta opioid receptor: an evolving target for the treatment of brain disorders. Trends Pharmacol Sci32 (2011), 16459-16468.

[11] S.J. Ward, D.C. Roberts. Microinjection of the delta-opioid receptor selective antagonist naltrindole 50 -isothiocyanate site specifi cally affects cocaine self-administration in rats responding under a progressive ratio schedule of reinforcement. Behav Brain Res 182 (2007), 140–144

[12] D. Simmons, D.W. Self. Role of mu- and delta-opioid receptors in the nucleus accumbens in cocaine-seeking behavior.Neuropsychopharmac34(2009), 1946–1957.

[13] J. Gelernter, H.R. Kranzler. Variant detection at the δ opioid receptor ( OPRD1) locusчand population genetics of a novel variant affecting protein sequence.Hum Genet107 (2000), 86–88.

[14] H. Zhang, H.R. Kranzler, B.Z. Yang, X. Luo, J. Gelernter. The OPRD1 and OPRK1 loci in alcohol or drug dependence: OPRD1 variation modulates substance dependence risk. Mol Psychiatry13 (2008), 531–543.

[15] R.C. Crist, L.M. Ambrose-Lanci, M. Vaswani, et al. Case- Control Association Analysis of Polymorphisms in the Delta- Opioid Receptor, OPRD1, with Cocaine and Opioid Addicted Populations.Drug Alcoh Depend127 (2013),122-128.

Page 309: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

307

Chapter 40

Effect of Myosmine, Administered Alone and on Tert-Butyl Hydroperoxide-Induced Toxicity

in Isolated Rat Hepatocytes

Magdalena KONDEVA-BURDINAa 1, Galina GORNEVAb, Mitka MITCHEVAa

a Laboratory “Drug metabolism and drug toxicity”, Department of Pharmacology, Pharmacotherapy and Toxicology, Medical University-Sofi a,

Faculty of Pharmacy, Sofi a, BULGARIAbInstitute of Molecular Biology, Bulgarian Academy of Sciences, Sofi a, BULGARIA

Abstract. The alkaloid Myosmine [3-(1-pyrrolin-2-yl) pyridine] is presented not only in tobacco products but also in various foods. The cytotoxicity of Myosmine was compared to Nicotine, using isolated rat hepatocytes. It was investigated also for possible protective effect on tert-butyl hydroperoxide-induced toxicity. The effects of Myosmine were evaluated on rat hepatocytes, a well-controlled biological model system in vitro, with high metabolizing capacity, isolated by two-stepped collagenase perfusion. Hepatocytes were treated with equi toxic concentrations of Myosmine (50 μM - 2 mM) and Nicotine (5 nM – 250 μM). In tert-butyl hydroperoxide (75 μM) model of toxicity, Myosmine was in concentrations 100 μM and 250 μM. Administered alone, Myosmine revealed statistically signifi cant, cytotoxic effects, which were concentration dependent and less toxic to those of Nicotine. In tert-butyl hydroperoxide model of toxicity, Myosmine had statistically signifi cant cytoprotective effects. According to these results, we can suggest that such cytoprotective effect of Myosmine might be due to an infl uence on tert-butyl hydroperoxide metabolism in rat hepatocytes.

Key words. Myosmine, isolated rat hepatocytes, cytoprotection

Introduction

Myosmine is the second tobacco alkaloid to nicotine and has been detected in nuts and nut products. However, myosmine has a much wider distribution over a wide variety of food products such as maize, rice, wheat, cocoa and milk [1].

1 Corresponding author: Magdalena Kondeva-Burdina, Dunav 2, 1000 Sofi a, Bulgaria; E-mail: [email protected]

Page 310: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

308

In vitro experiments with calf thymus DNA have shown that Myosmine is rapidly nitrosated at pH 2 to pH 4 and forms DNA adducts [2]. Using the Comet assay in human lymphocytes and upper aerodigestive tract epithelial cells, Kleinsasser et al., have demonstrated dose- and time-dependent genotoxic effects of Myosmine [3]. Doering & Richter (2009) found that Myosmine inhibit human aromatase [4].

According these literature data, the aim of our study was to investigate Myosmine for possible cytoprotective effects on tert-butyl hydroperoxide (t-BuOOH)-induced oxidative stress in rat hepatocytes.

1. Materials and methods1.1.Chemicals Myosmine was synthesized at the Institute of Molecular Biology, BulgarianAcademy

of Science (Figure 1).

Figure 1. Structure of Myosmine

1.2.AnimalsMale Wistar rats (body weight 200–250 g) were housed in plexiglass cages (3 per

cage), under standard laboratory conditions (ambient temperature 20°C ± 2°C and humidity 72 % ± 4 %) with free access to water and standard pelleted rat food. Animals were purchased from the NationalBreedingCenter, Sofi a, Bulgaria. All performed procedures were approved by the Institutional Animal Care Committee and the principles stated in the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientifi c Purposes (ETS123) were strictly followed throughout the experiment.

1.3. Isolation and incubation of hepatocytesRats were anesthetized with sodium pentobarbital (0.2 ml/100 g). In situ liver perfusion

and cell isolation were performed as described by Fau, with modifi cations [5, 6].

1.3.1. Lactate dehydrogenase releaseLactate dehydrogenase (LDH) release in isolated rat hepatocytes was measured as

described by Bergmeyer [7].

1.3.2. Cell viability, GSH depletion and MDA assayCell viability and the levels of reduced glutathione (GSH) and malondialdehyde

(MDA) were measured as described by Fau [5].

1.4. Statistical analysisStatistical analysis was performed using statistical programme ‘MEDCALC’. Results

Page 311: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

309

are expressed as mean ± SEM for 6 experiments. The signifi cance of the data was assessed using the nonparametric Mann–Whitney test.

2. Results and DiscussionIn experimental toxicology the in vitro systems play an important role for the

investigation of xenobiotic biotransformation and reveal the possible mechanisms of toxic stress and its protection.

Myosmine and Nicotine, administered alone, showed concentration-dependent toxic effects, as statistically signifi cant decreased cell viability and GSH level, increased LDH activity and lipid peroxidation in isolated rat hepatocytes, compared to the control (Tables 1, 2). These results correlate with other our experiments, that confi rms that single i.p. and oral administration of Myosmine signifi cantly increases lipid peroxidation and compromises the enzymatic and non-enzymatic antioxidative defence systems. Its pro-oxidant effects are comparable with those of Nicotine [1].

Table 1. Effect of Myosmine (M) and Nicotine (N), administered alone, on cell viability and LDH leakage in isolated rat hepatocytes

Group Cell viability (%) Effect vs control (%)

LDH (μmol/min/mill cells) Effect vs control (%)

Control 86 ± 5,5 100 0,073 ± 0,02 100

50 μM M 80 ± 3,1 ↓ 7 0,086 ± 0,02 ↑ 18

100 μM M 77 ± 4,1 * ↓ 10 0,112 ± 0,01 * ↑ 53

250 μM M 75 ± 2,1 * ↓ 13 0,130 ± 0,02 ** ↑ 78

500 μM M 59 ± 4,2 ** ↓ 31 0,141 ± 0,02 ** ↑ 93

1 mM M 52 ± 2,6 ** ↓ 40 0,186 ± 0,01 *** ↑ 155

1,5 mM M 42 ± 5,3 *** ↓ 51 0,243 ± 0,03 *** ↑ 234

2 mM M 40 ± 2,1 *** ↓ 53 0,271 ± 0,04 *** ↑ 271

5 nM N 56 ± 2,6 ** ↓ 35 0,141 ± 0,07 * ↑ 93

500 nM N 47 ± 4,2 *** ↓ 45 0,238 ± 0,03 *** ↑ 226

1 μM N 44 ± 5,2 *** ↓ 49 0,271 ± 0,04 *** ↑ 271

10 μM N 40 ± 4,6 *** ↓ 53 0,294 ± 0,05 *** ↑ 303

30 μM N 37 ± 5,2 *** ↓ 57 0,339 ± 0,03 *** ↑ 364

100 μM N 32 ± 6,1 *** ↓ 63 0,362 ± 0,05 *** ↑ 496

250 μM N 30 ± 7,2 *** ↓ 65 0,407 ± 0,06 *** ↑ 457

*p< 0,05; ** p < 0,01; *** p < 0,001 vs control

Page 312: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

310

Table 2. Effect of Myosmine and Nicotine, administered alone, on GSH level and lipid peroxidation in isolated rat hepatocytes

Group GSH (nmol/mill cells) Effect vs control (%)

MDA (nmol/mill cells)

Effect vs control (%)

Control 15 ± 3,2 100 0,073 ± 0,002 100

50 μM M 13 ± 2,5 ↓ 13 0,081 ± 0,005 ↑ 11

100 μM M 10 ± 2,9 * ↓ 33 0,083 ± 0,003 ↑ 14

250 μM M 9 ± 1,7 * ↓ 40 0,112 ± 0,01 * ↑ 54

500 μM M 8 ± 4,1 ** ↓ 47 0,136 ± 0,001 *** ↑ 83

1 mM M 7 ± 2,7 ** ↓ 53 0,185 ± 0,02 ** ↑ 153

1,5 mM M 6 ± 3,5 *** ↓ 60 0,201 ± 0,004 *** ↑ 175

2 mM M 5 ± 5,0 *** ↓ 67 0,241 ± 0,001 *** ↑ 230

5 nM N 10 ± 3,2 * ↓ 33 0,101 ± 0,01 * ↑ 38

500 nM N 9 ± 2,6 * ↓ 40 0,121 ± 0,01 ** ↑ 66

1 μM N 8 ± 3,2 ** ↓ 47 0,178 ± 0,01 ** ↑ 144

10 μM N 7 ± 4,2 ** ↓ 53 0,278 ± 0,01 *** ↑ 281

30 μM N 6 ± 3,8 *** ↓ 60 0,289 ± 0,02 *** ↑ 296

100 μM N 5 ± 4,1 *** ↓ 67 0,306 ± 0,02 *** ↑ 319

250 μM N 4 ± 3,5 *** ↓ 73 0,456 ± 0,03 *** ↑ 525

*p< 0,05; ** p < 0,01; *** p < 0,001 vs control

It’s known that one of mechanisms for cytotoxicity of tert-butyl hydroperoxide are decreased level of GSH and increased lipid peroxidation [8].

The incubation of hepatocytes with tert-butyl hydroperoxide (75 μМ) resulted in statistically signifi cant reduction of: cell viability by 68 %; GSH level by 91 %; increased: LDH leakage by 196 %; MDA level by 216 %.

Pre-incubation of the hepatocytes with Myosmine, signifi cantly protected them against this model of oxidative stress. The compound, during tert-butyl hydroperoxide hepatotoxicity, preserved the cell viability and GSH level, signifi cantly decreased LDH leakage in the medium and MDA level, compared to those measured in the samples incubated with the toxic agent only. The protective effects were lower to those of silymarin and were concentration-dependent, most prominent at the highest concentration 250 μM (Tables 3, 4).

According to our results, we can suggest that the observed cytoprotective effect of Myosmine on tert-butyl hydroperoxide-induced oxidative stress, might be due to infl uence on the lipid peroxidation process and liver glutathione concentration in rat hepatocytes.

Page 313: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

311

Table 3. Effect of Myosmine (M) and Silymarin (S) in t-BuOOH-induced oxidative stress on cell viability and LDH leakage in isolated rat hepatocytes

Group Cell viability (%)

Effect vs t-BuOOH (%)

LDH (μmol/min/mill cells)

Effect vs t-BuOOH (%)

Control 87 ± 5,1 0,163 ± 0,02

75 μM t-BuOOH 28 ± 5,3 *** 100 0,483 ± 0,07 *** 100

100 μM M + t-BuOOH 44 ± 3,1 ++ ↑ 57 0,377 ± 0,06 ++ ↓ 22

250 μM M + t-BuOOH 59 ± 4,2 +++ ↑ 111 0,320 ± 0,05 +++ ↓ 34

100 μM S + t-BuOOH 49 ± 2,5 +++ ↑ 75 0,240 ± 0,04 +++ ↓ 50

250 μM S + t-BuOOH 61 ± 4,6 +++ ↑ 118 0,186 ± 0,07 +++ ↓ 61

*** p < 0,001 vs control++ p < 0,01 ; +++ p < 0,001 vs t-BuOOH

Table 4. Effect of Myosmine (M) and Silymarin (S) in t-BuOOH-induced oxidative stress on GSH level and lipid peroxidation in isolated rat hepatocytes

Group GSH (nmol/mill cells)

Effect vs t-BuOOH (%)

MDA (nmol/mill cells)

Effect vs t-BuOOH (%)

Control 22 ± 3,7 0,051 ± 0,02

75 μM t-BuOOH 2 ± 4,7 *** 100 0,161 ± 0,02 *** 100

100 μM M + t-BuOOH 6 ± 3,9 +++ ↑ 200 0,133 ± 0,01 ++ ↓ 17

250 μM M + t-BuOOH 8 ± 4,3 +++ ↑ 300 0,127 ± 0,01 ++ ↓ 21

100 μM S + t-BuOOH 12 ± 5,7 +++ ↑ 500 0,085 ± 0,03 ++ ↓ 47

250 μM S + t-BuOOH 17 ± 3,4 +++ ↑ 750 0,057 ± 0,04 +++ ↓ 65

*** p < 0,001 vs control++ p < 0,01 ; +++ p < 0,001 vs t-BuOOH

3. ConclusionIn isolated rat hepatocytes, administered alone, Myosmine was less toxic then Nicotine

in equi toxic concentrations. In combination with tert-butyl hydroperoxide, Myosmine was shown to be an effective cytoprotector and antioxidant. The effects were lower to those of the fl avonoid silymarin – a classical hepatoprotector and antioxidant.

4. AcknowledgementsThis work was supported by the National Fund for Scientifi c Research grant no. TK-

L-1608/06 of the Bulgarian Ministry of Education and Science. The authors would like to thank Professor Elmar Richter for his continuous interest in this work and for helpful discussions, Dr Wolfgang Zwickenpfl ug for the HPLC analysis of Myosmine and Rumyana Gugova for its synthesis.

Page 314: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

312

References

[1] R. Simeonova, V. Vitcheva, G. Gorneva, M. Mitcheva, Effects of Myosmine on antioxidative defence in rat liver, Arh Hig Rada Toksikol63 (2012), 7-14.

[2] J. Wilp, W. Zwickenpfl ug, E. Richter, Nitrosation of dietary Myosmine as risk factor of human cancer, Food Chem Toxicol40 (2002), 1223-1228.

[3] N.H. Kleinsasser, B.C. Wallner, U.A. Harreus, W. Zwickenpfl ug, E. Richter, Genotoxic effects of Myosmine in human lymphocytes and upper aerodigestive tract epithelial cells, Toxicology192 (2003), 171-177.

[4] I.L. Doering & E. Richter, Inhibition of human aromatase by Myosmine, Drug Metab Lett3(2) (2009), 83-86.

[5] D. Fau, A. Berson, D. Eugene, B. Fromenty, C. Fisch, D. Pessayre, Mechanism for the hepatotoxicity of the antiandrogen, nilutamide. Evidence suggesting that redox cycling of this nitroaromatic drug leads to oxidative stress in isolated hepatocytes, J Pharmacol Exp Ther263(1) (1992), 69-77.

[6] M. Mitcheva, M. Kondeva, V. Vitcheva., P. Nedialkov, G. Kitanov, Effect of benzophenones from Hypericum annulatum on carbon tetrachloride-induced toxicity in freshly isolated rat hepatocytes, Redox Report11(1) (2006), 1-8.

[7] H. U. Bergmeyer, K. Gawehn, M. Grassl, Methods of Enzymatic Analysis. Ed. By H. U. Bergmeyer , Verlag Chemie, Weinheim, vol.1 (1974), 481-482.

[8] S. Vidyashankar, S.K. Mitra, K. S. Nandakumar, Liv.52 protects HepG2 cells from oxidative damage induced by tert-butyl hydroperoxide, Mol Cell Biochem333(1) (2010), 41-48.

Page 315: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

313

Chapter 41

Cytoprotective Effects of Saponarin from Gypsophila Trichotoma on Tert-Butyl Hydroperoxide

and Cocaine-Induced Toxicity in Isolated Rat Hepatocytes

Magdalena KONDEVA-BURDINAa1, Rumyana SIMEONOVAa, Vessela VITCHEVAa, Ilina KRASTEVAb, Mitka MITCHEVAa

a Laboratory “Drug metabolism and drug toxicity”, Department of Pharmacology, Pharmacotherapy and Toxicology, Medical University - Sofi a,

Faculty of Pharmacy, Sofi a, BULGARIAb Department of Pharmacognosy, Medical University - Sofi a,

Faculty of Pharmacy, Sofi a, BULGARIA

Abstract. Saponarin, isolated from Gypsophila trichotomaWend., was investigated for its protective effects on two models of oxidative stress – tert-butyl hydroperoxide (t-BuOOH) (75 μM) and cocaine (50 μM) in rat hepatocytes, isolated by two-stepped collagenase perfusion. The effects of saponarin were compared with those of silymarin. Cell incubation with t-BuOOH and cocaine led to a signifi cant decrease in cell viability, increased LDH leakage, decrease levels of cellular GSH and elevation in MDA quantity. Cells pre-incubation with saponarin (60 μg/ml; 6 μg/ml; 0,06 μg/ml; 0,006 μg/ml) signifi cantly ameliorated, in a concentration-dependent manner, the toxicants-induced hepatocytes damage. The effects are similar to those of silymarin (50 μg/ml; 5 μg/ml; 0,05 μg/ml; 0,005 μg/ml). Our results suggest that saponarin, isolated from Gypsophila trichotoma Wend., showed cytoprotective and antioxidant activity against those two models of oxidative stress, possibly by affecting the metabolism of tert-butyl hydroperoxide and cocaine.

Key words. saponarin, Gypsophila trichotoma, isolated rat hepatocytes, cytoprotection, antioxidant

IntroductionIsolated hepatocytes are a well-controlled, biological model system with high drug-

metabolizing capacities. This in vitro system is included in the battery of recommended tests from the European Centre for the Validation of Alternative Methods (ECVAM).

1 Corresponding author: Magdalena Kondeva-Burdina, Dunav 2, 1000 Sofi a, Bulgaria; E-mail: [email protected]

Page 316: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

314

There are data that some types of genus Gypsophila have antimicrobial, anti-cancer and hepatoprotective (in alcohol-induced hepatic fi brosis) effects and pancreatic lipase-inhibiting activity [1, 2, 3, 4].

According these literature data, the aim of our study was to investigate the saponarin, isolated from Gypsophila trichotoma Wend., for possible cytoprotective effects on two models of oxidative stress – tert-butyl hydroperoxide (t-BuOOH) and cocaine (50 μM) in rat hepatocytes.

1. Materials and methods1.1. Plant material, extraction and isolation of saponarinPlant material (overground parts) was collected in August 2008 at the Black Sea

coast, Bulgaria. A voucher specimen (SO 103887) has been deposited at the Herbarium of Faculty of Biology, Sofi aUniversity. Structural assessment of the compound was effected by acid hydrolysis and analysis of MS (ESI-MS: 595.1690 [M+H]+) and 1H and 13C NMR spectroscopic data [5] (Figure 1).

Figure 1. Structure of saponarin

1.2. Animals Male Wistar rats (body weight 200–250 g) were housed in plexiglass cages (3 per

cage), under standard laboratory conditions (ambient temperature 20°C ± 2°C and humidity 72 % ± 4 %) with free access to water and standard pelleted rat food. Animals were purchased from the NationalBreedingCenter, Sofi a, Bulgaria. All performed procedures were approved by the Institutional Animal Care Committee and the principles stated in the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientifi c Purposes (ETS123) were strictly followed throughout the experiment.

1.3. Isolation and incubation of hepatocytesRats were anesthetized with sodium pentobarbital (0.2 ml/100 g). In situ liver perfusion

and cell isolation were performed as described by Fau, with modifi cations [6, 7].

7

6

5

8

43

2O1

1'

6'

5'

4'

3'

2'

O

O

1"

1'"

O5'"

4'"

3'"

2'"

OHHO

HO

6'"

OH

O5"

4"

3"

2"

OH

HOHO

6"OH

OH

OH

Page 317: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

315

1.3.1. Lactate dehydrogenase releaseLactate dehydrogenase (LDH) release in isolated rat hepatocytes was measured as

described by Bergmeyer [8].

1.3.2. Cell viability, GSH depletion and MDA assayCell viability and the levels of reduced glutathione (GSH) and malondialdehyde

(MDA) were measured as described by Fau [6].

1.4. Statistical analysisStatistical analysis was performed using statistical programme ‘MEDCALC’. Results

are expressed as mean ± SEM for 6 experiments. The signifi cance of the data was assessed using the nonparametric Mann–Whitney test.

2. Results and DiscussionIsolated liver cells are used as a suitable model for evaluation of the cytoprotective

effect of some perspective biologically active compounds, both newly synthesized and plant isolated.

It’s known that one of mechanisms for cytotoxicity of both cocaine and tert-butyl hydroperoxide are decreased level of GSH and increased lipid peroxidation [9, 10, 11].

The incubation of hepatocytes with tert-butyl hydroperoxide (75 μМ) and cocaine (50 μM) resulted in statistically signifi cant reduction of: cell viability by 65 % and 71 %; GSH level by 81 % and 79 %; increased: LDH leakage by 223 % and 112 %; MDA level by 220 % and 330 %, respectively.

Pre-incubation of the hepatocytes with saponarin, isolated from Gypsophila trichotomaWend., signifi cantly protected them against both models of oxidative stress. The compound, during tert-butyl hydroperoxide and cocaine-induced hepatotoxicity, preserved the cell viability and GSH level, signifi cantly decreased LDH leakage in the medium and MDA level, compared to those measured in the samples incubated with the toxic agent only. The protective effects were similar to those of silymarin and were concentration-dependent, most prominent at the highest concentration 60 μg/ml (for saponarin) and 50 μg/ml (for silymarin)(Tables 1-4).

According to our results, we can suggest that the observed cytoprotective effect of the saponarin, isolated from Gypsophila trichotoma Wend., on both models of toxicity, might be due to infl uence on the lipid peroxidation process and liver glutathione concentration in rat hepatocytes.

Table 1. Effect of saponarin (SG) and silymarin (S) in t-BuOOH model of cytotoxicity on cell viability and LDH leakage in isolated rat hepatocytes

Group Cell viability (%)

Effect vs t-BuOOH (%)

LDH (μmol/min/mill cells)

Effect vs t-BuOOH (%)

Control 82 ± 5,5 0,141 ± 0,05

75μM t-BuOOH 29 ± 1,1 *** 100 0,456 ± 0,07 *** 100

Page 318: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

316

60 μg/ml SG + t-BuOOH 57 ± 1,1 +++ ↑ 97 0,195 ± 0,01 +++ ↓ 57

6 μg/ml SG + t-BuOOH 55 ± 2,1 +++ ↑ 90 0,272 ± 0,004 +++ ↓ 40

0,06 μg/ml SG + t-BuOOH

49 ± 4,2 +++ ↑ 69 0,364 ± 0,01 ++ ↓ 20

0,006 μg/ml SG + t-BuOOH

32 ± 2,6 +++ ↑ 10 0,397 ± 0,01 ↓ 13

50 μg/ml μM S + t-BuOOH

62 ± 5,3 +++ ↑ 113 0,148 ± 0,01 +++ ↓ 68

5 μg/ml S + t-BuOOH 56 ± 2,1 +++ ↑ 93 0,271 ± 0,04 ++ ↓ 41

0,05 μg/ml S + t-BuOOH 43 ± 2,6 +++ ↑ 48 0,326 ± 0,07 + ↓ 29

0,005 μg/ml S + t-BuOOH

34 ± 4,2 ++ ↑ 17 0,381 ± 0,03 + ↓ 16

***p< 0,001 vs control+ p< 0,05; ++ p < 0,01; +++ p < 0,001 vs t-BuOOH

Table 2. Effect of saponarin (SG) and silymarin (S) in t-BuOOH model of cytotoxicity on GSH level and lipid peroxidation in isolated rat hepatocytes

Group GSH (nmol/mill cells)

Effect vs t-BuOOH

(%)

MDA (nmol/mill cells)

Effect vs t-BuOOH

(%)

Control 26 ± 7,9 0,08 ± 0,002

75μM t-BuOOH 5 ± 0,5 *** 100 0,240 ± 0,005 *** 100

60 μg/ml SG + t-BuOOH 13 ± 0,9 +++ ↑ 160 0,083 ± 0,003 +++ ↓ 65

6 μg/ml SG + t-BuOOH 9 ± 0,7 +++ ↑ 80 0,142 ± 0,001 +++ ↓ 41

0,06 μg/ml SG + t-BuOOH 8 ± 1,1 +++ ↑ 60 0,156 ± 0,001 +++ ↓ 35

0,006 μg/ml SG + t-BuOOH 7 ± 0,7 +++ ↑ 40 0,185 ± 0,02 +++ ↓ 23

50 μg/ml S + t-BuOOH 14 ± 0,5 +++ ↑ 180 0,082 ± 0,004 +++ ↓ 66

5 μg/ml S + t-BuOOH 13 ± 1,0 +++ ↑ 160 0,141 ± 0,001 +++ ↓ 41

0,05 μg/ml S + t-BuOOH 8 ± 3,2 +++ ↑ 60 0,161 ± 0,01 +++ ↓ 33

0,005 μg/ml S + t-BuOOH 7 ± 1,6 +++ ↑ 40 0,178 ± 0,01 +++ ↓ 26

***p< 0,001 vs control+++p< 0,001 vs t-BuOOH

Table 3. Effect of saponarin (SG) and silymarin (S) in Cocaine model of cytotoxicity on cell viability and LDH leakage in isolated rat hepatocytes

Group Cell viability (%) Effect vs Cocaine (%)

LDH (μmol/min/mill cells)

Effect vs Cocaine (%)

Control 79 ± 4,8 0,305 ± 0,01

50μM Cocaine 23 ± 5,3 *** 100 0,648 ± 0,02 *** 100

Page 319: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

317

60 μg/ml SG + Cocaine 62 ± 1,1 +++ ↑ 170 0,402 ± 0,06 +++ ↓ 38

6 μg/ml SG + Cocaine 46 ± 4,2 +++ ↑ 100 0,478 ± 0,1 +++ ↓ 26

0,06 μg/ml SG + Cocaine 41 ± 0,5 +++ ↑ 78 0,534 ± 0,06 +++ ↓ 18

0,006 μg/ml SG + Cocaine 33 ± 2,6 +++ ↑ 43 0,636 ± 0,06 ↓ 2

50 μg/ml S + Cocaine 66 ± 10,0 +++ ↑ 187 0,459 ± 0,1 +++ ↓ 29

5 μg/ml S + Cocaine 48 ± 2,6 +++ ↑ 109 0,507 ± 0,1 ++ ↓ 22

0,05 μg/ml S + Cocaine 39 ± 1,1 +++ ↑ 70 0,532 ± 0,06 +++ ↓ 18

0,005 μg/ml S + Cocaine 34 ± 4,2 ++ ↑ 48 0,602 ± 0,2 ↓ 7

***p< 0,001 vs control++p< 0,01; +++ p < 0,001 vs Cocaine

Table 4. Effect of saponarin (SG) and silymarin (S) in Cocaine model of cytotoxicity on GSH level and lipid peroxidation in isolated rat hepatocytes

Group GSH (nmol/mill cells)

Effect vs Cocaine (%)

MDA (nmol/mill cells)

Effect vs Cocaine (%)

Control 19 ± 0,7 0,043 ± 0,01

50μM Cocaine 4 ± 0,7 *** 100 0,185 ± 0,004 *** 100

60 μg/ml SG + Cocaine 14 ± 0,7 +++ ↑ 250 0,06 ± 0,01 +++ ↓ 68

6 μg/ml SG + Cocaine 10 ± 0,7 +++ ↑ 150 0,140 ± 0,01 +++ ↓ 24

0,06 μg/ml SG + Cocaine 8 ± 1,1 +++ ↑ 100 0,161 ± 0,001 ++ ↓ 13

0,006 μg/ml SG + Cocaine 6 ± 1,1 + ↑ 50 0,177 ± 0,01 ↓ 4

50 μg/ml S + Cocaine 13 ± 0,7 +++ ↑ 225 0,055 ± 0,01 +++ ↓ 70

5 μg/ml S + Cocaine 8 ± 1,2 +++ ↑ 100 0,135 ± 0,002 +++ ↓ 27

0,05 μg/ml S + Cocaine 7 ± 1,5 ++ ↑ 75 0,166 ± 0,002 ++ ↓ 10

0,005 μg/ml S + Cocaine 6 ± 1,5 ++ ↑ 50 0,178 ± 0,01 ↓ 4

***p< 0,001 vs control+p< 0,05; ++ p < 0,01; +++ p < 0,001 vs Cocaine

3. ConclusionIn isolated rat hepatocytes, in combination with tert-butyl hydroperoxide and cocaine,

the saponarin, isolated from Gypsophila trichotomaWend., was shown to be an effective cytoprotector and antioxidant. The effects were close to those of the fl avonoid silymarin –a classical hepatoprotector and antioxidant.

4. AcknowledgementsThe authors would like to thank the Medical Science Committee for funding this

research (Project № 42 /2011) through Concurs Grant 2011.

Page 320: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

318

References

[1] Q. Zheng, W. Li, L. Han, K. Koike, Pancreatic lipase-inhibiting triterpenoid saponins from Gypsophila oldhamiana., Chem Pharam Bull55(4) (2007), 646-650.

[2] A. Weng, K. Jenett-Siems, P. Schmieder, D. Bachran, C. Bachran, C. Gorick, M. Thakur, H. Fuchs, M.F. Melzig, A convenient method for saponin isolation in tumor therapy, J Chromatogr B Analyt Technol Biomed Life Sci878 (7-8) (2010), 713-718.

[3] A. Shafagha, M. Shafaghaltionbar, Antimicrobial activity and chemical constituents of the essential oils from fl ower, leaf and stem of Gypsophila bicolor from Iran, Nat Prod Commun6(2) (2011), 275-276.

[4] Q.F. Huang, S.J. Zhang, L. Zheng, M. Liao, M. He, R.B. Huang, L. Zhuo, X. Lin, Protective effect of isoorientin-2”-O-α-L-arabinopyrranosyl, isolated from Gypsophila elegans on alcochol-induced hepatic fi brosis in rats, Food Chem Toxicol50 (2012), 1992-2001.

[5] M. Yotova., I. Krasteva, K. Jenett-Siems, P. Zdraveva, S. Nikolov, Secondary metabolites in Gypsophila trichotoma Wend, Pharmacognosy Magazine22 (2010), 159 (Suppl.).

[6] D. Fau, A. Berson, D. Eugene, B. Fromenty, C. Fisch, D. Pessayre, Mechanism for the hepatotoxicity of the antiandrogen, nilutamide. Evidence suggesting that redox cycling of this nitroaromatic drug leads to oxidative stress in isolated hepatocytes, J Pharmacol Exp Ther263(1) (1992), 69-77.

[7] M. Mitcheva, M. Kondeva, V. Vitcheva., P. Nedialkov, G. Kitanov, Effect of benzophenones from Hypericum annulatum on carbon tetrachloride-induced toxicity in freshly isolated rat hepatocytes, Redox Report11(1) (2006), 1-8.

[8] H. U. Bergmeyer, K. Gawehn, M. Grassl, Methods of Enzymatic Analysis. Ed. By H. U. Bergmeyer , Verlag Chemie, Weinheim, vol.1 (1974), 481-482.

[9] D. A. Donnelly, C. S. Boyer, D. R. Petersen, D. Ross, Cocaine-induced biochemical changes and cytotoxicity in hepatocytes isolated from both mice and rats, Chem Biol Interact 67(1-2) (1988), 95-104.

[10] C.R. Gцldlin, U. A. Boelsterli, Reactive oxygen species and non-peroxidative mechanisms of cocaine-induced cytotoxicity in rat hepatocyte cultures, Toxicology69(1) (1991), 79-91.

[11] S. Vidyashankar, S.K. Mitra, K. S. Nandakumar, Liv.52 protects HepG2 cells from oxidative damage induced by tert-butyl hydroperoxide, Mol Cell Biochem333(1) (2010), 41-48.

Page 321: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

319

Part 4

CHEMICALS TOXICITY

Page 322: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

320

Page 323: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

321

Chapter 42

Toxic Effects of Benzene Metabolites Hydroquinone, Catechol and Phenol in HL60 Cells

Virginia TZANKOVAa,1,Boyan VELICHKOVa, Massimo VALOTIb,aMedical University of Sofi a, Faculty of Pharmacy, Dunav 2 Str. 1000 Sofi a, Bulgaria,

bDipartimento di Neuroscienze, Sezione di Farmacologia, Universitа di Siena, viale A. Moro 2, 53100 Siena, Italy.

AbstractBenzene is industrial and environmental contaminants which continue to be of toxicological concern. The myelotoxic effects of benzene are well established, however the mechanism involved in benzene long-term toxicity –acute myeloid leukemia and myeloplastic syndromes remains unclear. HL-60 human promyelocytic leukemia cell was used as an experimental model to explore possible toxic effects of three benzene metabolites, hydroquinone, catechol and phenol. Screening using fl ow cytometry (FCM) was applied to evaluate the glutathione content (GSH), cell viability and mitochondrial potential. HL-cells were treated with either phenol, catechol or hydroquinone, and stained then with propidium iodide, JC-1 (5,5’,6,6’-tetrachloro-1,1’,3,3’ tetraethyl benzimidazolylcarbocyanine iodide) or CellTracker™ Green CMFDA (5-chloromethylfl uorescein diacetate) and sorted by FCM according to emitted fl uorescence intensity. Hydroquinone [100 μM] and catechol [75-100 μM] induced time dependent (0-24h) decrease in GSH levels, increased mitochondrial membrane potential and decreased cell viability, whereas phenol did not. In the lower concentrations of hydroquinone [100 μM] and catechol [75-100 μM] no statistically signifi cant effects on GSH and mitochondrial membrane potential was observed. Key words: benzene, catechol, toxicity

IntroductionBenzene is an important industrial chemical and environmental pollutant. It is

well know that benzene is myelotoxic and leukemogenic in humans. It is metabolized in liver by CYP2E1 to various metabolites. The main metabolites of benzene include 1,4-benzoquinone, hydroquinone, catechol, and phenol [1]. Some of these metabolites are formed in the liver and others through subsequent metabolism in the bone marrow [2, 3]. Bone marrow contains high levels of myeloperoxidase, which can catalyze the further metabolism of phenolic compounds to reactive free radical species and the generation of reactive oxygen species (ROS). Evidence for ROS formation after benzene exposure

1 Corresponding Author: Virginia Tzankova, Medical University of Sofi a, Faculty of Pharmacy, Dunav 2 Str. 1000 Sofi a, Bulgaria; e-mail: [email protected]

Page 324: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

322

has been shown through in vitro models as benzene’s metabolites increase cellular ROS and oxidative DNA damage in HL60 cells [4]. Benzene or benzene’s metabolites’ ability to induce DNA damage has been shown in vitro, in mice, and in humans [5, 6, 7].The production of ROS is tightly regulated and the cell possesses detoxifying mechanisms such as catalase, glutathione (GSH) and superoxide dismutase (SOD) to maintain appropriate cellular ROS levels. Cellular GSH has been shown to be crucial for regulation of cell proliferation, cell cycle progression and apoptosis [8, 9]. An alteration in GSH commonly occurs in response to ROS formation and oxidative stress, probably as an antioxidant defense mechanism[10, 11].

In the mid- 90s became also clear that mitochondria are active participants in the process of apoptosis. Mitochondrial dysfunction is an early event that preceded changes in nuclear and plasma membranes. It is characterized by an increase in the permeability of the mitochondrial membrane, and loss of membrane potential, which connects the release of cytochrome c into the cytoplasm. According to some studies there is a link between GSH levels and mitochondrial membrane potential [12].

In the present study we evaluated the effects of benzene’s metabolites hydroquinone (HQ), catechol (CAT) and phenol (PH) on GSH and the mitochondrial membrane potential in HL-60 cells. HL-60 cells were used as an in-vitro model system because they contain a high level of myeloperoxidase (MPO).

1. Materials and methods

1.1. Chemicals

Phenol, hydroquinone, catechol were purchased from Sigma Chemical Co. (St. Louis, MO). High purity distilled phenol was purchased from International Biotechnologies, Inc. (New Haven, CT). All other chemicals and solvents were of the highest grade commercially available.

1.2. Cell culture

HL60 cells obtained from the American Type Culture Collection (Rockville, MD) were cultured in RPMI 1640 supplemented with fetal bovine serum albumin (10%) and gentamicin sulfate (50 μg/ml). Cells were grown in a humidifi ed atmosphere in 5% CO2 and 37°C. Cell viability was determined using trypan blue exclusion in which 200 cells/ culture were analyzed. All initial viabilities were greater than 95%.

1.3. In Vitro Studies

HL60 cell cultures were preincubated for 0.5 h at 37°C before dosing with various metabolites of benzene. Cells (0.5 X I06/ml) were incubated in PBS (pH 7.4) with HQ, CAT, or PH [10-100 μM] up to 24 h.

1.4. Detection of intracellular glutathione (GSH) and cell viability

Cellular GSH levels were analyzed using 5- chloromethylfl uorescein diacetate (CMFDA, Molecular Probes) or the Glutathione Assay Kit (Sigma, St. Louis, MO). CMFDA

Page 325: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

323

is a membrane-permeable dye for determining levels of intracellular glutathione [13]. In brief, cells were incubated with hydroquinone, catechol or phenol for 24 h. Cells were then washed in PBS and incubated with 5μM CMFDA at 37◦C for 30 min according to the instructions of the manufacturer. Cytoplasmic esterases convert nonfl uorescent CMFDA to fl uorescent 5-chloromethylfl uorescein, which can then react with the glutathione (Ex/Em = 522 nm/595 nm).

Propidium Iodide (PI; (1μg/ml)) (Ex/Em = 488 nm/617 nm) was subsequently added, and CMF fl uorescence and PI staining intensity were determined using a FACStar fl ow cytometer (Becton Dickinson) and calculated by CellQuest software. PI was used to differentiate dead and normal cells. This agent is membrane impermeant and generally excluded from viable cells. For each sample, 5000 or 10,000 events were collected [14].

1.5. Measurement of mitochondrial membrane potentialA new method for the cytofl uorimetric analysis of mitochondrial membrane potential

in intact cells has been applied by using the lipophilic cationic probe 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolcarbocyanine iodide (JC-1), whose monomer emits at 527 nm after excitation at 490 nm. Depending on the membrane potential, JC-1 is able of forming J-aggregates that are associated with a large shift in emission (590 nm). The color of the dye changes reversibly from green to greenish orange as the mitochondrial membrane becomes more polarized [15].

1.6. Statistical analysis

Results represent the mean of at least three independent experiments; bar, S.D. Microsoft Excel or Instat software (GraphPad Prism4, San Diego, CA, USA) was used to analyze the data. Student’s t-test was used for parametric data. Statistical signifi cance was defi ned as P < 0.05.

2. Results and discussion

2.1. Effect of of hydroquinone (HQ), catechol (CAT) and phenol (PH) on GSH level, cell viability and mitochondrial membrane potential in HL-60 cells.

In the preliminary experiments the concentration - dependent effects of hydroquinone [1-100 μM] catechol [1-100 μM] and phenol [1-100 μM] on cell viability of HL-60 cells, were assessed after 15 min incubation time. The analysis of the results shows high cytotoxic effects of hydroquinone [50-100 μM ], while in the concentration range [0.1 - 25 μM], the effect was less pronounced. Catechol had no statistically signifi cant cytotoxicity, even in the highest concentrations [50 -100 μM]. Phenol had no statistically signifi cant effects. Based on the results from the preliminary data, our study was focused on the effects of hydroquinone [10 μM] and of catechol [50 and 100 μM] on glutathione (GSH) levels, cell viability the mitochondrial intermembrane potential evaluation.

In series of experiments, HL-60 were incubated with HQ [10 μМ], and CAT [50 μМ] and CAT [100 μМ] for 24 h. We measured glutathione status by CMF fl uorescence, cell viability by PI fl uorescence, and mitochondrial membrane potential by JC-1 fl uorescence.

HQ [10 μМ] exhibitseffects similar to that of CAT [50 μМ] on GSH levels (Fig. 1

Page 326: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

324

and Fig.2). Bothmetabolitesshowed time-dependent reduction in GSH levels to 27.19% ± 9.97 and to 11.17% ± 5.28, respectively, after6 hours of incubation. The effect was more pronounced in the higher concentration of CAT [100 μМ] causing a decrease to 3.66% ± 2,51 compared to the controls. Interestingly, the subsequentregenerationof the celltorestorethe level ofcellularglutathioneandmaintenanceofthe viability of thecellswas observed.

To evaluate whether the population of cells in the negative CMF fl uorescence region were dead, the cells were stained with additional PI for verifying disruption of the plasma membrane. Cell viability decreased with the increasing of the incubation time for both benzene metabolites. It dropped to 69.73%±10.85 after HQ [10 μМ] treatment, to 77.58%±8.96 and 38.49%±1.24, after CAT [50 μМ] and CAT [100 μМ] treatment, respectively. After 4h of incubation the negative CMF fl uorescence cells mostly showed PI-positive staining, which indicates that the cells showing GSH depletion were dead. These results confi rm that after 6 h of incubation, HQ treatment signifi cantly reduced GSH levels in HL-60 cells, as confi rmed by the direct measurement of glutathione content in cells (data not shown).

Severaldata suggest thatmitochondria play animportant rolein signal transductionand theamplifi cation of theapoptoticresponse.It appears that reactive oxygen species (ROS) produced by the mitochondria can be involved in cell death. The mitochondrial permeability transition is an important step in the induction of cellularapoptosis. During this process the electrochemical gradient (referred to as ΔΨ) across the mitochondrial membrane collapses. Evidence for ROS formation after benzene exposure has been shown through in vitro models as benzene’s metabolites increase cellular ROS and oxidative DNA damage in HL60 cells [4, 16].

Mitochondrial membrane potential (ΔΨm) is generated by mitochondrial electron transport chain, which drives a proton fl ow from matrix through inner mitochondrial membrane to cytoplasm, thus creating an electrochemical gradient. This gradient is in turn responsible for the formation of ATP molecules by F0-F1 ATP synthase. For this reason ΔΨm is an important parameter for mitochondrial functionality and an indirect evidence of energy status of the cell. Therefore, we also examined whether HQ and CAT affect mitochondrial membrane potential (ΔΨm). Both metabolites HQ and CAT caused loss of mitochondrial intramembranous potential in HL-60 cells, showing the lowest level after 6h of incubation (HQ to 14.67% ± 2.1; CAT to 49.05% ± 1.84), followed by a partial recovery during the next hours. Even at the lowerconcentrationof 10 μM, HQ caused time-dependent and statistically signifi cant breackdownin the mitochondrial membrane potential. The statistically signifi cant loss of the potential of HQ was evident after only 1 h of incubation, whilst effect of CAT was delayed compared to the HQ.

CAT can be converted to 1,2-benzoquinone by myeloperoxidases, and the reverse reaction is mediated by NAD(P)H-quinone oxidoreductase. Hydroquinone and 1,4-benzoquinone can also undergo this interconversion with the same enzymes [17]. Both quinones can undergo NADH-dependent redox cycling, via a semiquinone radical, to produce ROS [18]. Catechol and hydroquinone have been shown to have different redox properties, with catechol displaying an initially slower ability to generate superoxide via the oxidation of Cu2+ and subsequent reduction of cytochrome c in vitro[19]. This difference

Page 327: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

325

in superoxide generating ability could be a reason for attenuate effect of catechol on loss of mitochondrial membrane potential in respect to hydroquinone. Most probably there are additional and / or differentmechanisms underlying the loss of mitochondrial membrane potential caused by hydroquinone.

Conclusion

This study show that the benzene metabolites hydroquinone and catechol cause loss in the levels of glutathione (GSH), decrease in the cell viability, and loss of the mitochondrial intermembrane potential in human promyelocytic cell line HL60. We observed a decrease in cellular GSH that coincided with the loss of mitochondrial membrane potential. Both metabolites – HQ and CAT have shown a strong correlation between the loss of mitochondrial membrane potential and breakdown in GSH levels. No cells with high GSH values showed loss of mitochondrial membrane potential, excluding the possibility that the permeability transition is the cause of the GSH depletion. More likely, the permeability transition is due to failure of antioxidant mechanisms within the mitochondrial matrix space, as has been reported previously [1]

Figure 1. Effect of of hydroquinone (HQ) on GSH level (CMF fl uorescence) (■), cell viability (PI fl uorescence) (▲) and mitochondrial membrane potential (JC-1 fl uorescence) (▲) in HL-60 cells. Near-confl uent HL-60 cells were incubated for 24 h without (control, ctrl) or with HQ [10 μМ]. CMF fl uorescence cells and PI-positive staining and JC-1 fl uorescence cells were measured with FACSCalibur fl ow cytometer as described in Material and Methods. The graph shows the respective viable cells the levels of GSH content and the mitochondrial membrane potential, expressed as a percentage of the control value. Each value represents the mean ± S.D. of 4 determinations.

Page 328: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

326

Figure 2. Effect of of catechol (CAT) on GSH level (CMF fl uorescence) (■), cell viability (PI fl uorescence) (▲) and mitochondrial membrane potential (JC-1 fl uorescence) (▲) in HL-60 cells. HL-60 cells were incubated for 24 h without (control, ctrl) or with CAT [50 μМ] (fi gure 2A) and CAT [100 μМ] (fi gure 2B). CMF fl uorescence cells and PI-positive staining and JC-1 fl uorescence cells were measured with FACSCalibur fl ow cytometer as described in Material and Methods. The graph shows the respective viable cells the levels of GSH content and the mitochondrial membrane potential, expressed as a percentage of the control value. Each value represents the mean ± S.D. of 4 determinations.

References

[1] R., Snyder, Hedli, C.C., 1996. An overview of benzene metabolism. Environ. Health Perspect. 104 (Suppl. 6), 1165–1171.

[2] D.G. Schattenberg, Stillman, W.S., Gruntmeir, J.J., Helm, K.M., Irons, R.D., Ross, D., 1994. Peroxidase activity in murine and human hematopoieti progenitor cells: potential relevance to benzene-induced toxicity. Mol. Pharmacol. 46, 346–351.

[3] D.J Thomas, Sadler, A., Subrahmanyam, V.V., Siegel, D., Reasor, M.J., Wierda, D., Ross, D., 1990. Bone marrow stromal cell bioactivation and detoxifi cation of the benzene metabolite hydroquinone: comparison of macrophages and fi broblastoid cells. Mol. Pharmacol. 37, 255–262.

Page 329: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

327

[4] P. Kolachana, Subrahmanyam, V.V., Meyer, K.B., Zhang, L., Smith, M.T., 1993. Benzene and its phenolic metabolites produce oxidative DNA damage in HL60 cells in vitro and in the bone marrow in vivo. Cancer Res. 53, 1023–1026.

[5] Y. Hiraku, Kawanishi, S., 1996. Oxidative DNA damage and apoptosis induced by benzene metabolites. Cancer Res. 56, 5172–5178.

[6] H.Z. Pan, Na, L.X., Tao, L., 2003. DNA damage and changes of antioxidative enzymes in chronic benzene poisoning mice. Zhonghua Laodong Weisheng Zhiyebing Zazhi 21, 423–425 (abstract).

[7] D. Sul, Lee, E., Lee, M.Y., Oh, E., Im, H., Lee, J., Jung, W.W., Won, N., Kang, H.S., Kim, E.M., Kang, S.K., 2005. DNA damage in lymphocytes of benzene exposed workers correlates with trans,trans-muconic acids and breath benzene levels. Mutat. Res. 582, 61–70.

[8] M. Poot, H. Teubert, P.S. Rabinovitch, T.J. Kavanagh, 1995. De novo synthesis of glutathione is required for both entry into and progression through the cell cycle, J. Cell. Physiol. 163 (3): 555–560.

[9] T. Schnelldorfer, S. Gansauge, F. Gansauge, S. Schlosser, H.G.Beger, A.K. Nussler, 2000. Glutathione depletion causes cell growth inhibition and enhanced apoptosis in pancreatic cancer cells, Cancer 89 (7):1440–1447.

[10] P. J. O’Dwyer, Yao, K-S., Ford, P., Godwin, A. K., and Clayton, M., 1994. Effects of hypoxia on detoxicating enzyme activity and expression in HT29 colon adenocarcinoma cells. Cancer Res., 54: 3082-3087.

[11] K. Salnikow, Gao, M., Voitkun, V., Huang, X., and Costa, M., 1994. Altered oxidative stress response in nickel-resistant mammalian cells. Cancer Res., 54: 6407-6412.

[12] P. Costantini, Chernyak, B. V., Petronilli, V., and Bernardi, P. 1996. Modulation of the mitochondrial permeability transition pore by pyridine nucleotides and dithiol oxidation at two separate sites. J. Biol. Chem., 271: 6746-6751.

[13] T. Macho, A.Hirsch, I. Marzo, P. Marchetti, B. Dallaporta, S.A. Susin, N. Zamzami, G. Kroemer, 1997. Glutathione depletion is an early and calcium elevation is a late event of thymocyte apoptosis, J. Immunol. 158 (10): 4612–4619.

[14] D.W. Hedley, S. Chow, 1994. Evaluation of methods for measuring cellular glutathione content using fl ow cytometry, Cytometry 15 (4):349–358.

[15] A. Cossarizza, M. Baccaranicontri,G. Kalashnikova, C. Franceschi. 1993. A New Method for the Cytofl uorometric Analysis of Mitochondrial Membrane Potential Using the J-Aggregate Forming Lipophilic Cation 5,5′,6,6′-Tetrachloro-1,1′,3,3′-tetraethylbenzimidazolcarbocyanine Iodide (JC-1)Biochemical and Biophysical Research Communications, 197, (1): 40–45.

[16] L. Zhang, Robertson, M.L., Kolachana, P., Davison, A.J., Smith, M.T., 1993. Benzene metabolite, 1,2,4-benzenetriol, induces micronuclei and oxidative DNA damage in human lymphocytes and HL60 cells. Environ. Mol. Mutagen. 21, 339–348.

[17] S. Kim, Vermeulen, R., Waidyanatha, S., Johnson, B.A., Lan, Q., Smith, M.T., Zhang, L., Li, G., Shen, M., Yin, S., Rothman, N., Rappaport, S.M., 2006. Modeling human metabolism of benzene following occupational and environmental exposures. Cancer Epidemiol. Biomark. Prev. 15, 2246–2252.

[18] J.L. Bolton, Trush, M.A., Penning, T.M., Dryhurst, G., Monks, T.J., 2000. Role of quinones in toxicology. Chem. Res. Toxicol. 13, 135–160.

[19] K. Hirakawa, Oikawa, S., Hiraku, Y., Hirosawa, I., Kawanishi, S., 2002. Catechol and hydroquinone have different redox properties responsible for their differential DNA-damaging ability. Chem. Res. Toxicol. 15, 76–82.

[20] B. Kristal, Park, B. K., and Yu, B. P., 1996. 4-Hydroxyhexenal is a potent inducer of the mitochondrial permeability transition. J. Biol. Chem., 271: 6033-6038.

[21] C. Pieri, Recchioni, R., Marcheselli, F., Moroni, F., Maim, M., and Benatti, C. 1995. The impairment of mitochondrial membrane potential and mass in proliferating lymphocytes from vitamin E-defi cient animals is recovered by glutathione. Cell. Mol. Biol., 41:755-762.

Page 330: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

328

Chapter 43

Environmental Chemical Pollution Impact on Italian Campania Region

Dr Giussepe NOSCHESE, MD1, Col Dr Rostislav KOSTADINOV,MD, PhD2

1AORN “A.Cardarelli”, Naples, Italy2Military MedicalAcademy, Sofi a, Bulgaria

Abstract Introduction: For decades Campania region accepted clandestinely hundreds of tons of Toxic Industrial Materials – waste of the industrial production of Northern Italy, that were buried in various known and unknown location in the region. Moreover, the the waste management crisis is leading to burning the domestic waste, resulting in severe pollution of the air. Several national morbidity surveys are revealing signifi cant increase in the different neoplastic malignant processes in the Campania region in comparison with the average of the Italy.The aim of the publication is to present the new initiative started by medical and non medical entities in Campania region in order to establish a Standard Operating Procedures for monitoring the impact of environmental pollution on the Campania population health status. By the means of descriptive method toxic materials environmental pollution was described. Comparative method and deductive analyzes were applied in order to depict the main challenges health status monitoring system have to resolve.As a conclusion the authors are presenting the main elements, tasks and objectives of the e-health system to be established.Key words: Toxic Industrial Materials; e-health system; Environmental Pollution

Introduction

Contemporary world processes of industrialization and globalization are leading to establishment of different installation utilizing, processing and storing Toxic Industrial Materials – chemical elements and/or compounds. These chemical facilities are widespread globally with great variety of implemented safety and security protocols. From the time of notorious Bopal chemical incident the world is almost constantly witnessing chemical incidents with signifi cant impact on environment and population. Such incidents are not rare even in the most developed countries.

Naples is not only famous with the Vesuvius volcano and the Pizza Margherita, but also with its medieval hero Pulcinella. The mask carriers, vested in white clothes were Neapolitans expressing the population anger against the rulers in the 17 and 18 centuries. From those times an expression “Secret of Pulcinella” is used for describing something that is exposed to everyone, but no one wants and dare to speak about it (this was linked to the personality of the mask Pulcinella, in order to protect the brave citizens from the authorities revenge).

Page 331: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

329

Nowadays, unfortunately, the secret of Pulcinella is once again used, but in its contemporary sense, no one is revealing the well-known truth, because of either desperation – nothing could be changed, or because of the unwillingness to confront the mighty clandestine system established for extracting enormous lucrative profi t via waste management. [1] The topic is one of the greatest environnemental disasters in Italy – the clandestine burial of more than 13 million tons of toxic industrial materials (TIM) in the last 20 years. The TIM were dispersed in the Campania region, in the North Naples and Southern Caserta, in particular. According the offi cial data provided by the Italy’s Institute for Environmental Protection and Research (ISPRA) the production of TIM in Italy is about 135 million tons for one year, that is 4 times the production of the so-called urban waste (arround 30 millions tons). Out of this 135 millions tons more than 30 millions literally disapeared betwen the site of origin and the site for distruction. More than 1 million ton of them are particular dangerous for the human life and health, if are not destruct in accordance with the safety protocols. [1]

The reason of this is due to the fact that the Italian municipalities are allocating great ammount of funds for TIM and urban waste management. Both types of the waste is in constant increase due to the industrial production and the changes in the populations customs the widespread of the so called throwaway products, and therefore, the funds allocated for their destruction are also in constant grow. The clandestine organizations for years have focused their activities in becoming main players in the waste management market. The result of the clandestine waste management, in one of the worse hit Italian regions (Campania), is the increase of the morbidity and mortality amongs the population.

Surpisingly, very few surveys regarding the population morbidity and mortality are carried in the Campania region. Despite the fact of well-known health threat, related to the extremely dangerous substances found in the air, earth and water basins [2, 3, 4], relatively little researchs were conducted to analyse the impact of the recorded severe environmental pollution on the population health. The conducted surveys [5, 6] have revealed increased levels of cancer morbidity, mostly by liver cancer, lung cancer, breast cancer, cancer of the stomac, limphoma leukosis and sarcoma, and what is more, the places whith registred elevated levels of morbidity were coresponding with the sites where TIM were buried or managed by clandestine owned insenerators. But maybe due to the Secret of Pulcinella fenomenom these results provoked extremely scarce media coverege and scientifi c interest. But the fi gures are frightening - mortality rates for males with 9.2%, and with 12.4% for the females, higher than the average for Italy. Regarding the substances discovered –heavy metals as cadmium, mercury and asbestos in the ground, asbestos and 2,3,7,8-tetrachlordibenzo-p-dioxine in the air, methilen chloride in the waters of the bay, as just a few examples.

The aimThe aim of the publication is to present the new initiative started by medical and non

medical entities in Campania region in order to establish a Standard Operating Procedures for monitoring the impact of environmental pollution on the Campania population health status.

Results and DiscussionDespite the fact that highly toxic materials have been discovered as a environmental

Page 332: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

330

pollutants in Campania region for years, there is no comprehensive study performed in order clearly to demonstrate the link between recoded high mortality and morbidity levels among the Campania population and the poisonous environment, result of the waste mismanagement. What is more several published studies [8] are trying to convince the population that the recorded levels are due to particular Neapolitan life style.

Based on the results obtained in the Canadian survey about the impact of the waste sites on the population health (), a nongovernmental group of experts and companies are to establish a centre for health monitoring and prevention in the so-called “triangle or quadrangle of death” [9, 10, 11, 12, 13, 14].

The objective of the project “HEALTH TROUGH MONITORING AND PREVENTION” is to defi ne the most contaminated locations in the Campania region and analyzing the available environmental pollutants to defi ne the preventive measures required for decreasing the health risk levels, related to the environmental pollution. As the project main task experts set the monitoring of the population health indicators, thus obtaining data about the effi cacy and effi ciency of the implemented preventive measures.

The topicality of the Project is due to:1. Data scarcity regarding the environmental pollution in the Campania region – the

available map of the toxic sites is incomplete that is proven by the almost monthly discovery of new highly dangerous for the human health and animal life foci;

2. Lack of comprehensive study regarding the link between the elevated levels of neoplastic diseases, malformations and mortality of the population and environmental pollution present in the region;

3. The requirement for analysis on the effi ciency and safety of the implemented in the region methods for destruction of the urban and industrial waste;

4. The need for preventive measures to be implemented in order the deadly trend to be changed;

5. The capabilities of the modern nanotechnologies for monitoring different human and natural parameters;

6. The capacities of the e-health, telemedicine and telemonitoring for data obtaining, analyzing, sharing and dissemination.

The Project team is confi dent that via monitoring the levels of the environmental pollution the adequate preventive measures could be designated for implementation by the local authorities.

In order to achieve the set goal the project has to address not only environment by sampling the air, earth and water, but also has to monitor and assess the population health. From the utmost importance are what criteria for monitoring and what parameters will be monitored. Therefore, the fi rst step is the profound analyses on what side health effects could be expected when the human is constantly exposed to the detected toxic materials. Based on the results of these analyses and in regards to found toxic compounds the health parameters will be selected for every region. In the criteria selection the early cancer screening for entire population at risk has to be included.

When criteria are set and respective equipment for their monitoring is available the focused media campaign addressing the population at risk is to be performed, in order to gain the social support for project activities. Some of the topics of the campaign are:

Page 333: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

331

1. Providing information about the available environmental pollution;2. Presenting the possible health impact of the pollutants;3. Advertising the benefi ts from preventive measures implementation;4. Providing guidance on how the impact of environmental pollution could be

minimized;5. Advertising the monitoring as essential step in ameliorating the healthy conditions

in one polluted and dangerous environment;6. Providing possibilities for direct contact with the project team. For fulfi lling the tasks the project team plans to establish a robust scientifi c, research

and analytical net. The hub of this net will be the Virtual Health Centre where all the results from the performed analyses and shared data will be processed in order to create a region focused preventive program which implementation results will be assessed for adjusting the measures and signifi cantly decreasing the morbidity and mortality levels, success already achieved in Texas.

The desired structure of this scientifi c, research and analytical net is projected as minimum to include:

• 24/7 Call Consulting Center• Medical Assistance Coordinating Center• Telemonitoring • Telemedicine• Research on ambient and health protection • Laboratories and sampling teams• IT equipment including nanotechnologies for precise and continuing health

parameters’ and environment monitoring• Virtual Clinic – advice and monitoring• Established net for medical assistance – MEDEVAC and TreatmentThe structure is presented on fi gure 1:

Figure 1

Page 334: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

332

By establishing this Centre for Monitoring and Prevention the project authors will fi ll the gap between the available heavy toxic pollution of the Campania region and its impact on the population health and the limited, scarce population reaction to the increasing health threat, by providing on one hand, the required for adequate prevention data regarding the pathways polluted environment is affecting our health, and on the other hand the solution how to ameliorate the ambient and to decrease the health risks levels.

Reference

[1] Marfella Antonio La sovrapposizione dei fl ussi di rifi uti urbani e speciali industriali: il “segreto di Pulcinella” delle origini di Gomorra

[2] Terra di morte. La necessitа di agire. Il biocidio in Campania // http://www.contropiano.org/interventi/item/18785-terra-di-morte-la-necessita-di-agire-il-biocidio-in-campania

[3] Giordano A., Tarro G. Campania, Terra di veleni. Deanarro Libri, Napoli, 2012 [4] Giordano A Vivere in un mondo di rifi uti tossici: un silenzio assordante // Giordano A., Tarro G. Campania,

Terra di veleni. Deanarro Libri, Napoli, 2012 pp 23-29[5] Tarro G. Luci ed ombre della sanita a Napoli // Giordano A., Tarro G. Campania, Terra di veleni. Deanarro

Libri, Napoli, 2012 pp 31-38[6] Mazza A. Il Triangolo della morte // Giordano A., Tarro G. Campania, Terra di veleni. Deanarro Libri,

Napoli, 2012 pp 49-53[7] Senior K, Mazza A. Italian “Triangle of death” linked to waste crisis // The Lancet Oncology, Volume 5,

Issue 9, September 2004, Pages 525–527[8] Un triangolo della morte con il record dei tumori.// http://archiviostorico.corriere.it/2004/agosto/31/

triangolo_della_morte_con_record_co_9_040831031.shtml[9] http://www.oltregomorra.it/un-triangolo-della-morte-con-il-record-dei-tumori/[10] Nel triangolo della morte dove case, asili e strade sono costruiti con rifi uti tossici http://www.corriere.

it/ inchieste/nel-triangolo-morte-dove-case-asili-strade-sono-costruiti-rifi uti-tossici/7c27cbf0-5827-11e2-9a31-1eca72c52858.shtml

[11] Tumori/ Anomalie in Campania. Il triangolo della morte // http://archivio.rassegna.it/2007/attualita/articoli/campania2.htm

[12] Maio M. Tumore alla mammella: mai un caso! // Giordano A., Tarro G. Campania, Terra di veleni. Deanarro Libri, Napoli, 2012 pp 81-88

[13] Scodellaro D. Un cielo di veleni sopra la mia terra: la terra dei fuochi // Giordano A., Tarro G. Campania, Terra di veleni. Deanarro Libri, Napoli, 2012 pp 99-106

[14] Dessм I. Don Patriciello: “Nella Terra dei fuochi la strage degli innocenti. E quei veleni interrati provenienti dal Nord” // http://notizie.tiscali.it/articoli/interviste/13/10/don-patriciello-intervista-terra-fuochi.html

Page 335: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

333

Chapter 44

An Update of Issues Regarding Hair Analysis of Organic Pollutants

Matthaios P. KAVVALAKISa, Brice M.R. APPENZELLERb and Aristidis M. TSATSAKISa,1

aCenter of Toxicology Science & Research, MedicalSchool, University of Crete, GreecebLaboratory of Analytical Human Biomonitoring, Public Research Center –

Health, Luxembourg

Abstract. This paper presents a brief overview of all the published studies concerning hair analysis of organic pollutants associated with environmental and occupational exposure. Organic pollutants have an impact on human health after exposure to very low doses from environmental origin, food supply or occupational exposure, and these adverse health effects are still not known in depth. Dioxins, polychlorobiphenyls, polycyclic aromatic hydrocarbons, polybrominated diphenyl ethers and of course pesticides are the main groups of the most commonly measured pollutants in hair. This study underlines the importance of analytical sensitivity in hair analysis and the expected increase for assessing people exposure in future epidemiological studies.

Keywords.Hair, biomonitoring, organic pollutants, exposure.

1. Introduction

Organic Pollutants (OPs) are chemical substances that persist in the environment, bioaccumulate through the food web and pose a risk of causing adverse effects to human health and the environment [1]. Exposure to chemicals and OPs is a fact since humanity improves technological, agricultural and social activities. Because of the needing of the improvement and evolution of the humans, the burden of the environment in chemicals is of a great concern for the society. According to U.S.EPA, OPs are of a great concern because of their toxicity, their persistence, their long-range transport and of course their bioaccumulation [2].

OPs are toxic chemicals that several laboratory, fi eld and health studies have been linked to certain adverse health effects in people and wildlife [3]. They are highly stable chemicals that resist the natural processes of degradation. Once introduced into the environment, they can persist for a long time. They may be released in one part of the world but can travel

1 Center of Toxicology Science & Research, Medical School, University of Crete, 71003 Heraklion, Crete, P.O. Box 1393, GREECE. Tel.: +30-2810-394870 Fax: +30-2810-542098Email: [email protected]

Page 336: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

334

far from their original source via wind, water and to a lesser extent, migratory species. The major dust transport systems shown in Figure 1 are an evidence for that long-range transport of these substances. They are readily absorbed in fatty tissue and accumulate in the body fat of living organisms. These substances become more concentrated as they move up the food chain, especially into larger, longer-living organisms like humans.

Figure 1. The major global dust transport systems. An evidence for long-range transport of airborne gaseous and particulate substances in worldwide range. [4]

Concerning humans, several studies have already associated OPs exposure to many adverse health effects [3]. Reproductive, developmental, behavioral, neurologic, endocrine, immunologic, genetic damage are some of these adverse health effects that the organic pollutants contaminated in the environment are responsible for. Human activity expands in many fi elds and is responsible for environmental damage by contaminating soil, water and air with numerous organic pollutants.

The sources and the pathways of exposure to OPs is a multifaceted scenario. Food from the process of cooking to the packaging procedure and even the existence of additives is one of the most common sources of exposure to OPs. Outdoor environment (cities, villages, even country-side) is contaminated from several human activities (such as transportation, agriculture, industry, etc). Moreover, indoor environment is already contaminated with a variety of OPs that fulfi lls the list of possible sources of exposure to OPs. Furthermore, occupational exposure is of a great concern, while workers in facilities or fi elds of production and application of several chemical products are exposed in high concentrations for a long time. Biomonitoring of occupational groups to OPs and their major metabolites is of a great interest, since the opportunity to assess the risk of the burden to these chemicals is possible and also the possible correlation of high exposure to OPs and health issues or diseases could lead to signifi cant and safe conclusions.

2. Organic Pollutants

2.1. DioxinsDioxins are by-products of industrial and combustion processes or industrial

compounds. Dioxins are one of the fi rst OP group analyzed in human hair by Schramm

Page 337: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

335

and colleagues [5]. A total of 11 published studies detecting dioxins in hair can be found in literature. Two hundred sixty seven (267) people have been tested so far for dioxin levels in hair. Most of the published works were feasibility studies, except the most recent ones which reported higher concentration of both PCDD and PCDF in municipal solid waste incineration workers in Japan [6] and in people working in electronic waste recycling facilities in China [7-9], which clearly points out recycling/waste treatment activities as signifi cant sources of occupational exposure to dioxins.

2.2. Polychlorinated biphenyls (PCBs)Another OP group that was fi rstly detected in human hair was PCBs by Zupancic and

his colleagues also in 1992 [10]. In literature, 18 studies can be found concerning hair analysis of PCBs [11-24]. In total 707 people have tested for PCB levels in hair so far. The highest concentration in hair is reported for children living in urban region of Beijing (China) by Zhang and colleagues [11] and for people living close to e-waste disassembly sites in China [20].

Another interesting study on assessment of PCBs exposure using human hair using and comparing two analytical techniques (GC-DFHRMS and GC-MS) is that published by Barbounis and colleagues [25]. Hair samples from two agricultural regions of Greece (Crete and Peloponnesus) were collected and analyzed. Peloponnesus’ samples were positive to PCBs from 86.1 to 94.4%, while Crete’ samples were less contaminated. Researchers concluded that GC-DFHRMS provided higher rates of positive samples and lower median values due to higher sensitivity and interferences from isobaric ions in GC-MS technique. Therefore GC-DFHRMS is considered as a powerful tool for such assessments in hair specimens.

2.3. Polycyclic aromatic hydrocarbons (PAHs)Polycyclic aromatic hydrocarbons (PAHs) are by-products of incomplete combustion

of organic matter (fossil fuel combustion, metallurgy industry, smoking, etc). In literature only 3 studies can be found and 155 people are already tested for PAHs levels in hair. Parent molecules were measured only by Toriba and colleagues [26]. On the other hand, the other two studies detected monohydroxy-metabolites of PAHs in hair [27,28]. All tested hair samples were found positive to PAHs, a fact maybe attributed to the overburdened environmental air because of the transportation, the increased industrial activity and of course the personal habit of smoking.

2.4. Polybrominated diphenyl ethers (PBDEs)The most recently tested in hair polybrominated diphenyl ethers are used commonly

as fl ame retardants. A total of 8 studies published can be found in literature and 384 individuals have been tested for PBDEs. The highest concentration found in hair observed for BDE-47 (188pg/mg) was in people living in an area nearby e-waste recycling facilities in Taizhou (China) [17].

2.5. PesticidesOne of the major OP group is pesticides, a chemical class with variability in

structures. Some of the most commonly detected in hair pesticides are the organochlorines,

Page 338: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

336

organophosphates, carbamates, pyrethroids and recently the neonicotinoids [12-18,21-36]. These groups of pesticides are used in several agricultural and non-agricultural activities. The need of increasing agricultural production and also the overuse of insecticide products in pest control have increased the production and the use of the pesticides.

Pesticide usage is not a trend followed the last decades or centuries. Agriculture is a human activity that its beginning lies in 1800 BC. Almost 3 centuries later, Egyptians made insecticides to prevent lice, fl eas and wasps. Later in 1000 BC the Greek author of Iliad and Odyssey, Homer, preferred to use sulphur. The Roman statesman Cato in 200 BC told vineyard farmers to burn bitumen to remove insects. In early 1700s, John Parkinson said to use a mixture of vinegar, cow dung, and urine to be put on trees with canker to help them kill the “virus”. Later in 1885 French Prof. Millardet, found a copper mixture to prevent mildew. The “big bang” in pesticides came in 1939. World War II caused three discoveries in the fi eld of pesticides: the DDT, the organophosphorus pesticides and the phenoxyacetic herbicides (Figure 2). Until the dawn of the twentieth century, all food is considered to be “organic”. But then comes the invention and increasing use of synthetic nitrogen fertilizers and pesticides. DDT (organochlorine) was introduced the market in 1944 and banned in the 1970s. Organophosphates (introduced the market in 1940s) and carbamates (in 1950s) were banned in 2000s. The latest pyrethroids were introduced market in 1960s and some of them banned the current decade (Figure 2). Finally, the newest neonicotinoids entered the market in 1980s, while thoughts exist for banning them because of their effect on bees. The evolution of pesticides is remarkable in the late century, refl ecting the evolution to all the organic pollutants.

Figure 2. Timeline of pesticides history – shows when the major pesticide classes introduced worldwide market and when EU legislation began ban of some of them

The fi rst study for detection of pesticides in human hair was reported in 1998 [29], where Dauberschmit and colleagues actually analyzed lindane but did not have a positive case. The highest concentration ever measured was for diuron (4610 ng/g) [32]. Most frequently analyzed group of pesticides are the organochlorines (18 studies involving more than 2000 individuals). Lindane (γ-HCH) banned in Europe since 1998, currently detected in 100% of the hair samples tested. The newcomers: pyrethroids (2006) and neonicotinoids (2013) report so far low levels of positive detection. Organophosphates: due to their easy degradation, low level of positive detection for parent molecules, but high level of positive detection when analyzing their degradation products dialkylphosphates (DAPs).

Page 339: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

337

3. Discussion

Compared to drug hair analysis, OPs and especially dioxins and PCBs were fi rstly measured in hair in 1992 as aforementioned, and then followed the most recent reported hair analysis of organophosphates, pyrethroids, PBDE and neonicotinoids.

Figure 3. Timeline showing fi rst measurement of drug and organic pollutants using hair analysis.

The highest concentration reported in human hair among the OPs is observed for pesticides [3]. Compared to drugs of abuse, OPs appear in lower levels in hair. OPs concentration in hair is reported between 10 and 100 billion lower than illicit drugs (Figure 4).

Figure 4. Organic pollutants and some drugs of abuse concentration levels in hair (pg/mg) as reported in the current literature [3,37]

An important issue in hair analysis is the analytical method’s sensitivity. The improvement of method’s sensitivity is due to technical progresses, the usage of tandem mass spectrometry (MS/MS) instead of MS and the usage of solid phase microextraction (SPME). A general comment concerning sensitivity is that over time the LOD value decreases while the % of positive detection increases.

Another issue that should be mentioned is that of external contamination. As discussed in introduction, OPs are likely to be present in human surroundings. External decontamination aims to remove impurities without reaching internal stored chemicals. There’s no consensus yet on decontamination procedures and different procedures are presented in the publications so far, depending on the team and on the compounds to be analyzed.

Page 340: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

338

Concluding, due to increase in analytical performances and due to the increases awareness of adverse effects associated with chronic exposure to organic pollutants, the use of hair analysis instead of more classical matrices to assess people exposure in epidemiological studies will increase (Figure 5).

Figure 5. Studies published from 1992 to 2013 (so far) in the fi eld of OP detection in human hair.

References

[1] United Nations Environment Programme (UNEP), (http://www.chem.unep.ch/pops, Accessed Sept 2013)[2] U.S. Environmental Protection Agency, Offi ce of International Affairs, Persistent Organing Pollutants: A

global issue, a global response, EPA160-F-02-001, April 2002[3] B.M.R. Appenzeller and A.M. Tsatsakis, Hair analysis for biomonitoring of environmental and occupational

exposure to organic pollutants: State of the art, critical review and future needs, Toxicology Letters210 (210), 119-140

[4] C.A Kellogg and D.W. Griffi n, Aerobiology and the global transport of desert dust, Trends in Ecology & Evolution21, 11 (2006), 638-644

[5] K.W. Schramm, T. Kuettner, S. Weber, K. Lьtzke, Dioxin hair analysis as monitoring pool, Chemosphere24 (1992), 351–358

[6] T. Nakao, O. Aozasa, S. Ohta, H. Miyata, Survey of human exposure to PCDDs, PCDFs, and coplanar PCBs using hair as an indicator, Arch. Environ. Contam. Toxicol.49 (2005), 124–130

[7] J.K.Y. Chan, G.H. Xing, Y. Xu, Y. Liang, L.X. Chen, S.C. Wu, C.K.C. Wong, C.K.M. Leung, M.H. Wong, Body loadings and health risk assessment of polychlorinated dibenzo-p-dioxins and dibenzofurans at an intensive electronic waste recycling site in China, Environ. Sci. Technol.41 (2007), 7668–7674

[8] S. Wen, F.-X. Yang, Y. Gong, X.-L. Zhang, Y. Hui, J.-G. Li, A.-L. Liu, Y.-N. Wu, W.-Q. Lu, Y. Xu, Elevated levels of urinary 8-hydroxy-2’-deoxyguanosine in male electrical and electronic equipment dismantling workers exposed to high concentrations of polychlorinated dibenzo-p-dioxins and dibenzofurans, polybrominated diphenyl ethers, and polychlorinated biphenyls, Environ. Sci. Technol.42 (2008), 4202–4207

[9] J. Ma, J. Chenga,W. Wanga, T. Kunisueb, M. Wuc, K. Kannan, Elevated concentrations of polychlorinated dibenzo-p-dioxins and polychlorinated dibenzofurans and polybrominated diphenyl ethers in hair from workers at an electronic waste recycling facility in Eastern China, Journal of Hazardous Materials186, 2–3, (2011), 1966–1971

[10] L. Zupancic-Kralj, J. Jan, J. Marsel, Assessment of polychlorobiphenyls in human/poultry fat and hair/plumage from a contaminated area, Chemosphere25 (1992), 1861–1867

[11] H. Zhang, Z. Chai, H. Sun, Human hair as a potential biomonitor for assessing persistent organic pollutants, Environ. Int.33 (2007), 685–693

Page 341: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

339

[12] A. Covaci, P. Schepens, Chromatographic aspects of the analysis of selected persistent organochlorine pollutants in human hair, Chromatographia53 (2001), S366–S371

[13] A. Covaci, M. Tutudaki, A.M. Tsatsakis, P. Schepens, Hair analysis: another approach for the assessment of human exposure to selected persistent organochlorine pollutants, Chemosphere46 (2002), 413–418

[14] A. Covaci, C. Hura, A. Gheorghe, H. Neels, A.C. Dirtu, Organochlorine contaminants in hair of adolescents from Iassy, Romania, Chemosphere72 (2008), 16–20

[15] A.M. Tsatsakis, M.N. Tzatzarakis, M. Tutudaki, Pesticide levels in head hair samples of Cretan population as an indicator of present and past exposure, Forensic Sci. Int.176 (2008), 67–71

[16] A.M. Tsatsakis, M.N. Tzatzarakis, M. Tutudaki, F. Babatsikou, A.K. Alegakis, C. Koutis, Assessment of levels of organochlorine pesticides and their metabolites in the hair of a Greek rural human population, Hum. Exp. Toxicol.27 (2008), 933–940

[17] Salquebre, G., Schummer, C., Briand, O., Millet, M., Appenzeller, B.M.R.,. Multi-class pesticide analysis in human hair by gas chromatography tandem (triple quadrupole) mass spectrometry with solid phase microextraction and liquid injection. Anal. Chim. Acta (2011), In press.

[18] E.M. Ostrea, E. Villanueva-Uy, D.M. Bielawski, N.C.J. Posecion, M.L. Corrion, Y. Jin, J.J. Janisse, J.W. Ager, Maternal hair – an appropriate matrix for detecting maternal exposure to pesticides during pregnancy, Environ. Res.101 (2006), 312–322

[19] E.M. Ostrea Jr., D.M. Bielawski, N.C. Posecion Jr., M. Corrion, E. Villanueva-Uy, Y. Jin, J.J. Janisse, J.W. Ager, A comparison of infant hair, cord blood and meconium analysis to detect fetal exposure to environmental pesticides, Environ. Res.106 (2008), 277–283

[20] G. Zhao, Z. Wang, M.H. Dong, K. Rao, J. Luo, D. Wang, J. Zha, S. Huang, Y. Xu, M. Ma, PBBs, PBDEs, and PCBs levels in hair of residents around e-waste disassembly sites in Zhejiang Province, China, and their potential sources, Sci. Tot. Environ.397 (2008), 46–57

[21] L. Altshul, A. Covaci, R. Hauser. The relationship between levels of PCBs and pesticides in human hair and blood: preliminary results. Environ. Health Perspect.112 (2004), 1193–1199

[22] N.C. Posecion Jr., E.M. Ostrea Jr., D. Bielawski, M. Corrion, J. Seagraves, Y. Jin, Detection of exposure to environmental pesticides during pregnancy by the analysis of maternal hair using GC–MS, Chromatographia64 (2006), 681–687

[23] K. Neuber, G. Merkel, F.F.E. Randow, Indoor air pollution by lindane and DDT indicated by head hair samples of children, Toxicol. Lett.107 (1999), 189–192

[24] W. Tirler, G. Voto, M. Donega, PCDD/F, PCB and hexachlorobenzene levels in hair, Organohalogen Compd.52 (2001), 290–292

[25] E.G. Barbounis, M.N. Tzatzarakis, A.K. Alegakis, A. Kokkinaki, N. Karamanos, A. Tsakalof, A.M. Tsatsakis, Assessment of PCBs exposure in human hair using double focusing high resolution mass spectrometry and single quadrupole mass spectrometry, Toxicology Letters210, 2, (2012), 225–231

[26] A. Toriba, Y. Kuramae, T. Chetiyanukornkul, R. Kizu, T. Makino, H. Nakazawa, K. Hayakawa, Quantifi cation of polycyclic aromatic hydrocarbons (PAHs) in human hair by HPLC with fl uorescence detection: a biological monitoring method to evaluate the exposure to PAHs, Biomed. Chromatogr.17 (2003), 126–132

[27] C. Schummer, B.M.R. Appenzeller, M. Millet, R. Wennig, Determination of hydroxylated metabolites of polycyclic aromatic hydrocarbons in human hair by gas chromatography–negative chemical ionization mass spectrometry, J. Chromatogr. A1216 (2009), 6012–6019

[28] B.M.R. Appenzeller, C. Mathon, C. Schummer, A. Alkerwi, M. Lair. Simultaneous determination of nicotine and PAH metabolites in human hair specimen: A potential methodology to assess tobacco smoke contribution in PAH exposure, Toxicology Letters210, 2(2012), 211-219

[29] C. Dauberschmidt, R. Wennig, Organochlorine pollutants in human hair, J. Anal. Toxicol.22 (1998), 610–611

[30] D.B. Barr, R. Allen, A.O. Olsson, R. Bravo, L.M. Caltabiano, A. Montesano, J. Nguyen, S. Udunka, D. Walden, R.D. Walker, G. Weerasekera, R.D.J. Whitehead, S.E. Schober, L.L. Needham, Concentrations of selective metabolites of organophosphorus pesticides in the United States population, Environ. Res.99 (2005), 314–326

[31] T. Berman, D. Hochner-Celnikier, D.B. Barr, L.L. Needham, Y. Amitai, U. Wormser, E. Richter, Pesticide exposure among pregnant women in Jerusalem, Israel: results of a pilot study, Environ. Int.37 (2011), 198–203

[32] V. Cirimele, P. Kintz, B. Ludes, Assessment of pesticide exposure by hair analysis, Acta Clin. Belg.S1 (1999), 59–63

[33] C. Dauberschmidt, R. Wennig, Organochlorine pollutants in human hair, J. Anal. Toxicol.22 (1998), 610–611

Page 342: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

340

[34] M.G. Margariti, A.M. Tsatsakis, Analysis of dialkyl phosphate metabolites in hair using gas chromatography-mass spectrometry: abiomarker of chronic exposure to organophosphate pesticides, Biomarkers14 (2009), 137–147

[35] M.P. Kavvalakis, M.N. Tzatzarakis, E.P. Theodoropoulou, E.G. Barbounis, A.K. Tsakalof, A.M. Tsatsakis, Development and application of LC-APCI-MS method for biomonitoring of animal and human exposure to imidacloprid, Chemosphere (2013), In press

[36] M.P. Kavvalakis, M.N. Tzatzarakis, A.K. Alegakis, A.K. Tsakalof, A.M. Tsatsakis, Development and application of GC-MS method for monitoring of long-term exposure to cypermethrin, Drug Testing & Analysis (2013), Submitted

[37] F. Pragst, M.A. Balikova, State of the art in hair analysis for detection of drug and alcohol abuse, Clin. Chim. Acta 370 (2006), 17-49

Page 343: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

341

Chapter 45

Effect of Solvents on the Toxicity of Some L-Valine Peptidomimetics in Rats

Klisurov R.1, E. Encheva1, M.Genadieva 1, L. Tancheva2, D. Tsekova3

1Medical University of Sofi a, Medical Faculty2Institute of Neurobiology at Bulgarian Academy of Sciences3University of Chemical Technology and Metallurgy – Sofi a

Abstract.The effect of solvent on the toxicity of drugs and pharmacological agents is extremely relevant in the medical and pharmaceutical practice Two newly synthesized peptidomimetics derived from the amino acid L-valine exhibited signifi cant CNS pharmacological activity.Aim of this study is to compare the effects of some commonly used solvents (water, sunfl ower oil and Dimethyl sulfoxide (DMSO) on main toxic biochemical and histological parameters of laboratory rodents after 3-days of treatment with effective doses of the newly synthesized compounds.Methods: The experiments were conducted on mature male Wistar rats. The compounds with codes M6 and P6 (150 mg/kg, intraperitoneally, for 3 days) were dissolved in equal concentration in three different types of solvent - oil solution (sunfl ower oil), water (gum arabic) and an organic solvent, DMSO. Main biochemical parameters of toxicity in urine and histological samples of liver and kidneys were tested on day 4.Results: In oil solution and aqueous suspension, the compounds do not cause signifi cant changes in the studied biochemical urine parameters as well as in the hepatic and renal parenchymal histology. In contrast, dissolved in DMSO compounds demonstrated signifi cant hepatic and renal toxicity comparable for both studied compounds, and accompanied by some biochemical changes in the urine. The control group of animals treated only with DMSO had no signifi cant histological and biochemical urinary changes, demonstrating that the negative effect observed is not the result of solvent toxicity. Conclusion: Increased toxicity of newly synthesized compounds dissolved in DMSO happens under an unknown mechanism. It is probably due to improved solubility and facilitated penetration of M6 and P6 through membranes, as well as to other pharmacokinetic changes produced by DMSO. An important interaction solvent-compound is also possible to have occurred on the level of hepatic drug metabolism. This suggestion is supported by the established signifi cant parenchymal changes in the liver.

Key words. learning, memory, biogenic monoamines, hippocampus, L-valine

Introduction:

Liquid drugs pose many challenges to clinical practice, with major of them being toxicity and absorption of the drug. The problem of absorption is most easily overcome by administering the active substance into the systemic circulation parenterally, but even

Page 344: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

342

so the problem about toxicity of the solvent remains. In current clinical practice, liquid medications undergo a renaissance in the frequency of their use. Increasingly, new drugs may be used in the form of various liquid preparations. Implementation of very unstable, highly insoluble and problematic new molecules challenges the team of drug developers. The use of mixtures of solvents and other auxiliary substances is therefore mandatory. Each of the components may alter the biological behavior of the active ingredient and enhance or even cause occurrence of new toxic effects on the body. A full and an accurate survey should therefore be done carefully, in order to know the toxicity of each new preparation before clinical trial.

Aim of the present study was to investigate the role of the solvent for the toxicity of two newly synthesized peptidomimetic compounds, labeled M6 and P6(D. Tsekova et al., 2008) .

P6

M6

Fig. 1: The two peptidomimetics

In our prior studies we found that both compounds showed analgesic effect, they changed diversely cognitive processes and muscle coordination in laboratory animals. The experiments with isolated and collectively reared mice demonstrated improvement of memory processes after the administration of substances. Similar experiments with rats with altered cognitive functions showed normalization of memory performances in animals treated with the tested compounds (L.Tancheva et al., 2006, 2010). In the course of the experimental work the levels of serotonin and acetylcholine in hippocampus were studied. The initial ratios between the neurotransmitters in those animals that developed social isolation syndrome were found to be restored.

Previous toxicological studies in mice showed low oral toxicity of the studied compounds. This, together with accrued pharmacological data requires a more comprehensive study of their toxicity in different solvents.

Materials and MethodsThe aqueous suspension, oil and DMSO solutions of the compounds, prepared in the

same concentrations were studied. The experiment was performed on adult male Wistar rats of approximately the same weight and reared under normal conditions in accordance with

Page 345: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

343

the (ethical and laboratory) regulations. The solutions with compounds were administered daily for three days intraperitoneally (150 mg/kg). The histological preparations of liver and kidney and urinary biochemical parameters were examined after treatment. Each group of animals was compared to a control group which were administered pure solvent at the same volume for the same duration, and by the same route of administration.

ResultsThe aqueous suspension and oil solution of the compounds showed no toxicological,

macroscopic or histological changes in the histologic samples. Urinary parameters were also normal. In contrast, the solutions of the two compounds in DMSO showed signifi cant hepatic - and nephrotoxicity. Histologically we observed pathological changes such as centrilobular necrosis, lymphocytic fi ltration and destruction of liver lobules and visibly loosened parenchyma in the liver. We observed a necrotic renal parenchyma, with thickening and delamination of Bowman’s capsule in the kidney histology samples. The established lesions correlated well with biochemical changes in the urine - increased proteinuria, presence of erythrocytes and lymphocytes, increased bilirubin (Table 1). Solutions of M6 and P6 in DMSO resulted in 100% mortality of treated animals as the forementioned lesions were observed at necropsy. There were no signifi cant pathological changes in any parameter in the control group with pure DMSO. (Fig.2 C,D)

Figure 2. Comparison of the parenchyma tissues of kidney and liver of the treated with M6 dissolved in DMSO rats (A and B) A) Renal parenchyma (DMSO+M6), B) Liver parenchyma (DMSO+M6); and controls – C and D (these treated only with DMSO), C - renal parenchyma after DMSO, D - liver tissue slide after DMSO.

Table 1 – Biochemical parameters in urine after 3 days treatment with compound M6 in different solvents and controls treated with pure solvent DMSO

Parameters Water+M6 DMSO+M6 Controls (DMSO)Protein 0 100 0 Ketones 0 10 0 Glucose 0 5 0 Nitrites 0 0 0 Bilirubin 0 3 0 Leucocytes 0 5 0 Urobilinogen 0 1 0 Blood 0 50 0 SG 1,01 1,15 1,10

Page 346: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

344

Conclusions:

The conducted experiments confi rmed our working hypothesis on the crucial role of the solvent on the toxicity of the administered compounds. The most probable reason for the pathology observed are the changes in absorption of substances and in permeability of biological membranes under the infl uence of DMSO. A hypothesis is being discussed for the presence of a toxic liver metabolite - diamine, causing direct histological changes due to its high reactivity. Most likely, the increased peritoneal permeability under the effect of DMSO greatly increases the concentration of the active substance in plasma as well as the concentration of the toxic liver metabolite.

Further detailed experimental work in this direction would clarify the problem of toxicity observed and the overall metabolism of M6 and P6 in the body. It is advisable to examine additional solutions of active substances for potential toxicity to living organisms, which is a signifi cant problem in today’s environment and clinical practice.

References

[1] D. S.Tsekova, B. Escuder, J. F. Miravet, CrystGrowthDes, 8, 11 (2008).

[2] L. Tantcheva, V. V. Petkov, G. Karamukova, S. Abarova, Y. Chekalarova,D. Tsekova, B. Escuder, J. Miravet, K. Lyubomirova, Bulg. Chem. Comm., 38, 54 (2006).

[3] Tancheva L., M. Novoselski, E. Encheva, V. Petkov, G. Gorneva, D. Tsekova, J. Tchekalarova, I. Gurt, E Katz, and E. Pavlova, J. Med.CBR.,I, vol 8:277-284 (2010)

Page 347: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

345

Chapter 46

Health Risk Associated with Genetic Polymorphism of Apoe in Bulgarian Workers Exposed

to Carbon Disulfi de

Tzveta GEORGIEVA11, Zhivka GENOVA2, Vidka Peneva-NIKOLOVA1 , Teodor PANEV3, Anna MIKHAILOVA1, Todor POPOV1

1 The National Centre for public health and analysis Bull. “Accad.Yves. Geshov “15, Sofi a 1431, Tel: 028056239,

e-mail: [email protected] 2 Genika, Sofi a

3 Executive Environment Agency, Sofi a

Abstract: Numerous epidemiological studies have shown that some of the negative health effects of carbon disulfi de are based on infl uence of lipid metabolism and it may cause cardiovascular disease. Apolipoprotien E (APOE) plays a major role in plasma lipoprotein metabolism and is responsible for the transport of lipids in the bodies. APOE gene demonstrate three genetic polymorphic variants-APOE 2, APOE * 3, APOE * 4and the products of these three alleles differ in their functional characteristics. APOE * 4 allele is a risk factor for a predisposition to cardiovascular disease and Alcheimer’s disease. The purpose of this study was to determine is there a relationship between the presence of mutations in the genes, responsible for Apolipoprotin E synthesis and disturbance of lipid status in individuals exposed to carbon disulfi de. Object of study are 90 workers from the Bulgarian production, occupationally exposed to carbon disulfi de, distributed into 3 groups based on the level of exposure. The control group included 26, carbon disulfi de, unexposed individuals. Individual external and internal exposure was assessed. Biochemical indices were analyzed for illustration the status of lipid metabolism. Explored through RLFP- PCR method polymorphic gene variants of Apolipoprotein E genotype results demonstrate that 17% of studied population are with E3/E4 genotype. and 9% with E3/E2 genotype. The data show that there is an increased health risk for workers with E3/E4 genotype. The results of this study supplement the less data about Bulgarian population and demonstrate the need for additional investigation of individual susceptibility of exposed workers to chemical agents from the environment or workplace air.

Key words: Carbon Disulfi de, Genetic Polymorphism, APOЕ, health risk assessment

1. IntroductionThe studies of molecular targets of impacts as a result of occupational exposure to

various chemicals demonstrate their signifi cance for Molecular Toxicology and health risk assessment. There were identifi ed a large number of genetic factors leading to increased

1 Corresponding Author.

Page 348: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

346

individual susceptibility to chemical agents. Polymorphic variation of genes encoding enzymes or substrates involved in metabolizing of several xenobiotics may be markers for susceptibility. A large number of genes and their variants are already investigated. A number of isoforms of metabolizing enzymes and proteins involved in the metabolism of chemicals are explored.

Carbon disulfi de is widely used for rayon production, carbontetrachloride, rubber chemicals and cellulose fi lm, and as a by-product of dithiocarbamate pesticides. Chronic low level and long term exposure to CS2 can cause cardiovascular, reproductive and other negative health effects. [1, 2, 3, 4, 5].It is well known that carbon disulfi de has a harmful effect on the cardiovascular system.A number of studies have shown a signifi cant correlation between high levels of lipoproteins and coronary heart disease and Alzheimer’s disease.

Apolipoprotein E (AroE) participates in the binding, internalization, and degradation of lipoprotein particles. AroE is secreted by macrophages and hepatocytes and serves as a ligand for LDL (Aro B/E) and receptor of liver tissue for the specifi c type of AroE 1 and 2 receptors for triglyceride-rich residues [6,7]. The gene encoding the AroE is located in chromosome 19q 13.32 and contains several polymorphic locus. The most important isoforms of AroE are called E2, E3 and E4. ApoE3 (Cys112/Arg158) is the most common of the three described [8]. Within the isoform Cys112/1Cys158 AroE2 is connected with the remainder Hyperlipidemia .This isoform has a lower affi nity to AroE receptors. At AroE4 (Arg112/Arg158) isoform is associated with increased synthesis of VLDL and LDL as a risk factor for Alzheimer’s disease and other neurological disorders. There are other rare isoforms, which are associated with mixed Hyperlipidemia [6]

Heterozygous individuals have elevated serum cholesterol concentrations. ApoE is a involved in a clearance of HDL fractions that contributes to the reverse transport of cholesterol.[9] Genetic variants of ApoE locuses prerequisite on the level of plasma concentrations of cholesterol and for a relative risk of atherosclerosis. [10] On the other hand, the presence of carbon disulfi de exposure increases the risk of elevated serum cholesterol levels.

The identifi cation of the APO’s isoforms may be informative in assessing individual sensitivity to cardiovascular risk as a result of occupational exposure to carbon disulfi de.

2. Aim The purpose of this study was to determine whether there is a correlation between

the presence of the polymorphic variants in APOE genes, and disorders of lipid status in subjects exposed to carbon disulfi de.

3. Subjects Subjects of present study ware 86 workers from rayon production occupationally

exposed to carbon disulfi de. The Control group consists of 26 persons without occupational exposure.

The results presented in Table 1 show that the distribution by number and gender in the relevant groups is approximately equally, with the exception of the fi rst exposure group,

Page 349: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

347

where women predominate (third group consists only of men, due to the specifi c of the profession). The distribution by age, General and specialized work experience is also the same with the exception of the fi rst exposed group where the internship is smaller, but the differences are not statistically signifi cant (p > 0.05). High frequency makes the impression of smokers (80,8%) in the fi rst group, exhibited dominated by women.

Table1. Study population

Characteristic Control Group

(n = 29)IstExposure

group (n=28)

IIendExposure group (n=32)

IIIthExposure group(n=26)

SexWomen (n, %) Men (n, %)

11 (42.3 %)15 (57.7 %)

16 (61.5 %)10 (38.5%)

13 (52.0 %)12 (48.0 %) 26 (100%)

Age (X, SD) 45,6 ±9,2 40,8 ±6,1 46,0 ±6,7 47,4 ±9,4

Общ трудов стаж

(X, SD)26.1 ± 8.3 19.4 ± 6.9 26.08 ± 7.4 27.4 ± 10.7

Спец. трудов стаж

(X, SD)18.41 ± 10.44 13.23 ± 8.78 18.8 ± 9.41 17.8 ± 11.3

Smoking habits (n, %) дане

13 (52 %)12 (48 %)

21 (80.8 %)5 (19,2 %)

14 (56.0 %)11 (44.0 %)

15 (60.0 %)10 (40.0 %)

All persons involved in the study were informed of the purpose, benefi ts and risks of the study and participate voluntarily after signing the informed consent

4. Material and Methods 4.1. Questionnaire The survey includes data on age, gender and length of service, as well as smoking,

type and amount of used cigarettes, alcohol and other related lifestyle factors.4.2. Workplace Air Monitoring The personal exposure was measured. The samples for quantitative analysis were

collected on sorbent tubes fi lled with active charcoal. Chemical analysis was performed by NIOSH method [11] (1960/1994), verifi ed in Chemical laboratory of NCPHA with gas chromatography with mass spectrometric detector.

4.3. Internal exposure was assessed by examination of biomarkers – metabolites of carbon disulfi de in the urine. Samples were collected at the end of the working shift. There are measured:

• Common metabolites of carbon disulfi de by iodine-azidentest in urine[12] • Tiazolidin-4-carbon acid (TTCA) in urine by HPLC with UV detector [13,14]

Page 350: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

348

4.4. Biochemical investigation

Blood without additives to obtain the serum is collected by venipuncture VACUTAINERS (Beckton & Dickinson, UK). Lipid status were determined: total cholesterol (CHOL, mmol / l), cholesterol high density lipoprotein (HDL, mmol / L), cholesterol low density lipoprotein (LDL, mmol / L) and triglycerides (TRGL) (mmol / L) in sera with spectrophomoteric method. Analyses were performed on the test PointeScientifi c, USA, 180 Pointe chemistry analyzer (MFG, USA). The results obtained are compared with the received reference values in Bulgaria: CHOL - to 5,00 mmol / L, HDL - 1,70-4,60 mmol / L, LDL-0 0.78 to 1, 94 mmol / L, TRGL - 0 from 0.41 to 1, 88 mmol / L.

4.5. Individual susceptibility Blood for DNA extraction was collected by venipuncture (VACUTAINERS , Beckton

& Dickinson, UK) with EDTA anticoagulant. Molecular analysis of carriers of APOE*2 alleles, APOE*3, APOE*4 was conducted by polymerase chain reaction - PCR-RLFP [6]

4.6. Statistical analyses Database was made included results from external, internal exposure assessment,

questionnaire, biochemical indices and genetic analysis. Statistical analyses was performed with SPSS software version 15.00

5. Results and Discussion 5.1. Workplace air Results estimated the average shift exposure to carbon disulfi de show that in one of

the halls of the studied plant, concentrations of carbon disulfi de are highest - from 03.11 to 40.82 mg/m3. This group provisionally identifi ed as the Group 1. In a Group 2 most of the samples were in the normal range by one sample is 1.7 times above the limit value for an 8 hour exposure.

In the Group 3 of the measured concentrations of carbon disulfi de are lower than the limit value for an 8 hour exposure, and only two samples are 1.3 times higher than the limit value.

5.2. BiominoringThe results of the biological monitoring, distributed in groups, depending on the degree

of exposure is shown in Table 2.

Table 2. Biomarkers for internal exposure assessment

Biomarker Exposure group X SD Minimum Maximum Р value

TTCA/g creat Control 0,620 0,600 0,001 1,4801 2,087 0,96 0,001 7, 2484 0.001 (1 vs 0)2 0,689 0,50 0,001 1,895 0.001 (1 vs 2)3 0,720 0,63 0,103 2,393 0.001 ( 1 vs 3)

mol J2/mmolc Control 3,77 2,34 0,56 6,371 5,08 2,53 2,30 13,54 0.001 (0 vs 1)2 5,14 2,03 2,60 10,76 0.05 (0 vs 2)3 4,09 1,89 1,16 7,84 0.03 (0 vs 3)

Page 351: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

349

At concentrations of carbon disulfi de in working environment around 2 mg/m3, the total excretion of metabolites in urine TTCC ranges of physiological fl uctuations.

At two-fold increase in exposure to carbon disulfi de of level of 4 mg/m3, the total level of metabolites increases to that set for the control group (p <0,05), while urinary TTCC varies within normal values (p> 0,05).

The average concentrations of carbon disulfi de in the working environment of 20 mg/m3 exhibit biological responses on both metabolites. Among this group, marked the highest average contents TTCC and common metabolites in urine, compared with the designated control group (p < 0.001). “Dose-response” relationship is expressed in terms of the studied biomarkers. Displayed a statistically signifi cant correlation , characterized the relationship between TTCC and common metabolites as moderate (r = 0.478; P <0.05). In support the level of the correlation coeffi cient, determining the signifi cance of the relationship between exposure to carbon disulfi de and TTCC excretion in urine (r = 0.414; P <0.05).

The comparative analysis of data from exposer assessment and biological monitoring results allow exploring the dependencies associated with the level of exposure and the variance applied biomarkers for predicting health risk for chronic exposure to carbon disulfi de. Conducted correlation analysis showed a statistically signifi cant correlation between the concentrations of carbon disulfi de measured in the assessment of personal exposure and biomarkers determined in the urine of respondents : CS2mg/m3 vs TTCA Group 1 - r = 0.414, Group 2 - r = 0.428 (P <0.05) , Yodazide Test vs CS2 mg/m3 - group 3 -r = 0.506 (P <0.01);

5.3. Lipid status

In the Table 3 are given the results obtained in a study of the main parameters of the lipid metabolism - total cholesterol (CHOL), lipoproteins, high and low density (HDL respectively and LDL) and triglycerides (TIGL)

All tested parameters of lipid status were within the reference values for the method. A trend of correlation was found between the extent of exposure to carbon disulfi de in workplace air and levels of CHOL, HDL, LDL and TRGL. Analysis of the data showed that no statistically signifi cant differences were found in biochemical indices compared by the level of exposure.

Table 3. Results of biochemical tests (lipid status)

Biochemical Indices (mmol/L) Exposure group X SD

CHOL Control 4,96 0,79

1 4,06 1,09

2 4,85 1,07

3 4,22 0,82

Page 352: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

350

Reference value * до 5

HDL Control 1,58 0,411 1,22 0,282 1,18 0,463 1,29 0,29

Reference value 0.78 – 1.94

LDL Control 2,96 0,971 2,59 0,992 3,29 1,013 2,53 0,71

Reference value 1.7 – 4.6

TRGL Control 1,56 1,331 1,22 0,822 1,87 1,283 1,80 1,42

Reference value 0.41 – 1.88* Reference value are according to method used

5.4. ApoE gene polymorphismResults of molecular studies in the present study show that the highest incidence of

detection is presence of E3 allele.

Таble 4a Приблизително разпределение на човешките алелни честоти на АроЕ в световен мащаб [Farreret all, 1997] [15]

Alel E2 E3 E4

Frequency% 8.4 77.9 13.7

Table 4b Алална честота на изследваната в настоящето проучване, българска популация

Alel E2 E3 E4

Frequency % 9 74 17

When comparing the data from this study with data from other studies showed a trend towards a higher incidence of the studied Bulgarian population with carrier of E2 and E4 allele and a lower frequency of carriers of E3. The trend was not statistically signifi cant. As is clear from Tables 4a and 4b, a similar percentage of distribution was identifi ed in the literature data. [15]. For the evaluation of allele frequency whole exposed population and control group was analyzed.

For the purposes of this study we divided the subjects with occupational exposure by allelic frequency. Correlation and regression analysis of the results of measurements of the air of the working environment and biomarkers by genotype was conducted. In the study

Page 353: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

351

group with occupational exposure and carriers of Apo * 3 allele is established correlation between elevated levels of CHOL and LDL fractions and exposure levels and the values of TTCA - Chol: LDL (r = 0,688, p = 0,01), Chol: TTCA (r = 0,333, p = 0,01) Chol: CS2mg/m3 (r = 0,635, p = 0,01). The analysis of the group with occupational exposure and presence of Apo *4 alel demonstrated statistically signifi cant higher correlation between the levels of CHOL: LDL (r = 0,961, p = 0,05). In subjects with genotype Apo*2 we also found a statistically signifi cant correlation between the levels of CHOL: LDL (r = 0,909, p = 0,05).

Due to heterogenic distribution of the results from the internal and external exposure, the data are evaluated by the nonparametric tests (Mann-Whitney Test).

Figure 1. Level of CHOL, LDL, HDL and TRGL of occupationally exposure group, distributed by genetic variation of APOE

The comparative analysis of the levels of biochemical parameters, characterizing lipid status, depending on the genotype, was performed in the exposed groups. An Anova test was used. The results showed elevated levels of CHOL in those carriers of ApoE * 4 allele. Such dependence is registered for the control group (p = 0,03).

1,00 2,00 3,00

Apo E genotyping: 1= E*3; 2=E*4; 3= E*2

0,50

1,00

1,50

2,00

2,50

HDL

,mmol/L

1,00 2,00 3,00

Apr E genotyping: 1= E*3; 2=E*4; 3= E*2

1,00

2,00

3,00

4,00

5,00

6,00

LDL,

mmol/L

��

1,00 2,00 3,00

Apo E genotyping: 1= E*3; 2=E*4; 3= E*2

3,00

4,00

5,00

6,00

7,00

CH

OL

mm

ol/L

��

1,00 2,00 3,00

ApeE genotyping: 1= E*3; 2=E*4; 3= E*2

1,00

2,00

3,00

4,00

5,00

6,00

TRG

Lm

mol

/L

Page 354: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

352

Table 5. Distribution of lipid indices by genotype

Biocehm.Indices

Apo E Genotype Mean Std. Deviation Signidicance

CHOL E3/E3 4,51 0,98

E3/E4 4,99 1 0,07E3/E2 4,16 0,72

HDL E3/E3 1,28 0,41

E3/E4 1,32 0,31

E3/E2 1,53 0,41 0,16

LDL E3/E3 2,83 0,99

E3/E4 3,31 1,16 0,04E3/E2 2,31 0,85

TRIGL E3/E3 1,65 1,33

E3/E4 1,73 1,22 0,92E3/E2 1,53 1,12

The results showed statistically signifi cant differences in LDL values for different genotypes, they are highest in those exposed to E3/E4genotip. Registered trend increased levels of CHOL and TRGL and decreases in HDL, but they are not statistically signifi cant.

6. Conclusion The biological monitoring results confi rm appropriate selection of biomarkers to

evaluate the internal exposure. It should be emphasized that the high correlation between the internal and external exposure is evidence of the reliability of the strategy used for the exposure assessment and determination the concentrations of carbon disulfi de of the working environment. On the other hand the existing correlations rule out the possibility of „false” positives.

Concentrations of CHOL, LDL, HDL and TRGL are within referent values in accordance with the method used. However, the analysis carried out on the lipid status tends to increase the values of CHOL and LDL depending on the degree of exposure to carbon disulfi de.

Data from this study regarding the genetic polymorphism of ApoE gene corresponds with data from other studies and showing that heterozygotes subjects have elevated serum cholesterol. [16] In addition, the condition for increased health risk for them is the presence of occupational exposure, which also has detrimental effects on lipid metabolism. The analysis of the obtained data shows that there is an increased health risk for workers with genotype * E3 and * E4 .

Results of the polymorphic variants of the genes responsible for the synthesis of Apolipoprtein E showed similar frequency compared with data from other studies [ 17].

The results of this study complement the scarce data for Bulgaria population. Proved

Page 355: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

353

the importance of research at the molecular level, related to the assessment of individual sensitivity to chemical agents from the environment or the working environment and the information value of this type biomarkers for health risk assessments.

7. Reference

[1] Walker et all, 2001[2] Tanetal et all, 2001[3] WHO: World Health Organization. Task group on environmental health criteria for carbon disulfi de.

Environmental Health Criteria 10.Carbon disulfi de, Geneva (1979).[4] Kaloyanova, F.: Hygienic Toxicology. Generalities.Medicine and Sports, Sofi a (1981).[5] Kopcheva Chr., T.Panev, Tz.Georgieva, V.Nikolova, T.Popov, “External and internal exposure to Carbon

disulfi de at working place”, Proceedings of the NATO Advanced Study Instutute2006 , Springer, 401-508[6] Mahley RW, Rall SC Jr. Apolipoprotein E: farmorethan a lipid transport protein. Annu Rev Genomics Hum

Genet 2000;1:507–37; [7] Langer C, Huang Y, Cullen P, Wiesenhutter B, Mahley RW, Assmann G, et al. Endogenous apolipoprotein

E modulates cholesterol effl ux and cholesteryl ester hydrolysis mediated by high-density lipoprotein-3 and lipid-free apolipoprotein sin mouse peritoneal macrophages. J MolMed 2000;78:217–27

[8] Lambert-Hammill М, and A. S. Wierzbicki, Simple Sequence-specifi c-Primer- PCR Method To Identifythe Three Main Apolipoprotein E Haplotypes, PanagiotisPantelidis,1* ClinicalChemistry 49, No. 11, 2003 1945-1948

[9] Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science 1988;240:622-630

[10] Davignon J, Gregg RE, Sing CF. Apolipoprotein E polymorphism and atherosclerosis. Arteriosclerosis 1988;8:1-21

[11] NIOSH Pocket Guide to Chemical Hazards: www.skcinc.com.[12] Jakubowsk, M., Piotrowski, J.: Badania nad ocena stonia ehpozyci na dwusiarczek wegla. II J. Medycyna

Pracy, XVI, 2, 86-95 (1965).[13] Lauwerys, R. and Hoet, P: Industrial Chemical Exposure. Guidelines for biological monitoring.Carbon

disulfi de, Sec.edition, Lewis Puplishers, USA, 39-42 (1993).[14] Van Door R., Delbressine, L. P., Leijdekkers, C. M., Vertin, P.G., Henderson, P.Th.: Identifi cation and

determination of TTCA in urine of workers exposed to carbon disulfi de. Arch. Toxicol. 47, 51-58 (1981).[15] Farrer L.A., Cupples L.A., Haines J.L., Hyman B., Kukull W.A., Mayeux R., Myers R.H., Pericak-Vance

M.A., Risch N., van Duijn C.N. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease: a meta-analysis. (1997). JAMA 278: 1349–1356. doi:10.1001/jama.1997.03550160069041. PMID 9343467.

[16] Singh PP, Singh M, Mastana SS (2006). “APOE distribution in world populations with new data from India and the UK”. Ann.Hum. Biol. 33 (3): 279–308. doi:10.1080/03014460600594513. PMID 17092867.

[17] Eisenberg DTA, Kuzawa CW, Hayes MG. (2010). “Worldwide allele frequencies of the human apoliprotein E (APOE) gene: climate, local adaptations and evolutionary history”. American Journal of Physical Anthropology 143 (1): 100–111. doi:10.1002/ajpa.21298. PMID 20734437.

Page 356: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

354

Chapter 47

Chemical Risk Assessment an Imperative in Medical Training and Education

Col Dr Rostislav KOSTADINOV,MD, PhD1, Col (ret) Prof Dr Kamen KANEV, MD, PhD, DSc1,

Dr Giussepe NOSCHESE, MD2

1Military MedicalAcademy, Sofi a, Bulgaria2AORN “A.Cardarelli”, Naples, Italy

Abstract Contemporary world processes of industrialization and globalization are leading to establishment of different installation utilizing, processing and storing Toxic Industrial Materials – chemical elements and/or compounds. These chemical facilities are widespread globally with great variety of implemented safety and security protocols. From the time of notorious Bopal chemical incident the world is almost constantly witnessing chemical incidents with signifi cant impact on environment and population. Such incidents are not rare even in the most developed countries.The aim of this study is to present the required organizational skills and clinical knowledge medical specialist have to acquire in their education in order to be prepared to provide population with prompt and adequate medical support in case of chemical disaster. By the means of descriptive method the contemporary world chemical intoxication health risk related to the ongoing globalization is presented. Comparative method was applied in order to depict the main skills and knowledge requirements medical specialists have to obtain in order to respond to the contemporary world challenges. As a conclusion the authors are presenting a list of courses for addressing the imminent chemical threat.

Key words: Toxic Industrial Materials; Chemical Incidents; Medical Education and Training, Chemical Incident Medical Support and Management

Introduction: For describing the necessity of profound in-depth knowledge in chemical health risk

assessment, fi rstly is required to defi ne what the chemical disaster is and why it is of utmost importance for contemporary medical specialists to be trained in assessing the risk associated with the health hazards. One of the most popular and comprehensive defi nition of chemical disaster is the one given in Mosby’s Medical Dictionary, 2009 [1] – “chemical disaster is an accidental release of a quantity of toxic chemicals into the environment, resulting in death or injury to workers or members of nearby communities”. From this defi nition it is very easy to understand the increasing need of knowledge how to assess

Page 357: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

355

the chemical hazards and health risk in our contemporary world dominated by the process of globalization and related further industrialization. On one hand the globalization is benefi cial for both developed and developing countries, because this process is supporting the further developing of all economies, but on the other hand, the globalization is leading to implementation of highly sophisticated technologies in the countries. In the developing countries, in particular, not all the personnel involved in the production is trained to the level required for assuring the technological process safety and security, as well as not always, the infrastructure is suitable for safe transportation or storage of the precursors or fi nal products of the factories and plants. The globalization is leading also to increase in the speed of immigration, especially of the highly qualifi ed operators that are leaving their countries of origin looking for better paid assignments in the developed countries, thus creating serious gaps in the less economically developed and stable countries. These gaps are fi lled by less experienced operators with initial training but without practice and skills regarding the safety and security standard operating procedures. To summarize the effects of the globalization on the chemical industry:

1. Increase in the number of the chemical installations – for extracting or creation of the chemical elements and compounds, means and capabilities for transportation and storage of the extracted or created precursors, plants for processing of the chemicals and transportation and storage of the fi nal products;

2. Increased sophistication of the processes implemented for extracting, creation and processing of chemicals, leading to increased safety and security requirements and regulations;

3. Increase requirement for well trained and experienced operators;4. Increase operators’ migration towards developed countries;5. Establishment and operating of highly sophisticated industrial chemical installations

with personnel not suffi ciently trained and experienced for assuring the safety and security of the implemented chemical processes.

Contemporary world, especially in the last two – three decades, is facing and another phenomenon – the international terrorism. Despite the fact that chemical elements or chemical compounds have been used for terrorists’ purposes for millennia, [2] the real chemical terrorist’s threat is becoming almost imminent in last decades, when due to the development of the information technologies obtaining of highly toxic chemicals could be easily and not expensively achieved by those who would like to poses poisonous substances. Several are the evidence that the terrorists are eager, willing and capable to implement chemical compounds for their purposes [3]

Other contemporary world feature increasing the necessity of chemical health risk assessment is the recorded in the last decades trend in natural and man-made disasters frequency and severity. The recorded data [4, 5] presents grow in the number of occurred disasters, as well as increase in the disasters impact on population health – lives loses, disability etc.

Analyzing the medical community that is designated to decrease the chemical incidents and disasters health consequences it is easily to depict other factors increasing the need of postgraduate chemical risk assessment education and training for medical specialists.

Firstly, the great majority of graduating or new graduated medical specialists are

Page 358: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

356

attracted to follow postgraduate specialization in the fi elds of medicine with higher monthly income as neurosurgery, esthetic surgery, ophthalmology, orthopedic surgery etc, leaving the general practice, emergency medicine and toxicology with few candidates. Most of the programs, followed by the residents, are focused on the particular specialty skills and knowledge, where the principles of disaster medical management and health threat assessment are not included, or if included, their importance is diminished. All these are leading to gap in the physicians’ knowledge and practice how to assess the risk for themselves and the population in case of chemical incident. The same gap could be found and within the kills and knowledge of the other medical specialists. If we add to the above mentioned and the observed decrease in number of medical students and respectively in the graduated medical specialist, then the gap in the medical community readiness and preparedness to respond adequately and effi ciently to the health challenges of one chemical incident will become vast and frightening.

The aimThe aim of this study is to present the required organizational skills and clinical

knowledge medical specialist have to acquire in their education in order to be prepared to provide population with prompt and adequate medical support in case of chemical disaster.

Results and DiscussionIn order to defi ne what are the most important skills and knowledge every medical

specialist has to have, the question about what is required for provision of adequate and prompt medical support in case of chemical disaster has to fi nd its correct answer. When disaster strikes the time for reaction is limited, as well is limited and the time for medical support organization. As soon as possible the medical assistance is provided to the affected more lives could be saved. These basic principles are in particular valid in case of chemical disaster, when the life of the victims requires medical assistance in a minutes’ time frame. In regards to this principle it is reasonable to expect that the question related to the immediate, emergence treatment of chemical casualty should be in the focus of medical specialists’ education and training. But while evaluating the required knowledge and skills for proper and adequate provision of medical assistance to chemically affected population we have not to forget the other basic Disaster Medicine principle – Safety and Security. What is more important? Does to provide medical assistance to the peoples in need or to assure the safety fi rst? The answer is more than obvious – more important is to assure the safety of the teams in order they to be able to provide the required assistance. What is required for this activity? Firstly, knowledge about what type of chemical agent has affected the population. This so called hazard identifi cation is signifi cant for planning of the medical assistance activities – antidotes, clinical features, supportive therapy, detoxication etc.

Second, this hazard identifi cation is providing information about the required preventive measures against the released chemical – routes for intoxication, personal and collective protective equipment required for decreasing the number of affected and stopping the exposure to those already affected.

Third, this hazard identifi cation is providing the rescue teams with knowledge how they could protect themselves to not become victims, instead of providing support and assistance.

Page 359: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

357

Related to these observations there is no doubt about the primacy of hazard identifi cation and the following to it activities in comparison of the pure theoretical knowledge about toxicological treatment.

Once defi ned the medical education and training primary goal it is necessary to list the tasks to be fulfi lled in order this set goal to be achieved. From our analyses on the activities medical specialists are expected to perform in order to react safely and adequately in case of chemical disaster the following knowledge and skills should be acquired during the medical education and training:

1. Hazard Identifi cation2. Probability and Vulnerability Assessment3. Severity of consequences assessment4. Health Risk Assessment5. Health Risk CommunicationAfter the Bopal chemical incident [6] USA adopted Emergency Planning and

Community Right to Know Act (EPCRA) [7] where the need of community to know what is the possible chemical threat in every particular region is addressed. Moreover, the assessment and reporting tools are also described, in order to unify and facilitate the chemical disaster emergency planning and response. Addressing the increased probability of chemical incidence occurrence the WHO developed a manual regarding the chemical incident health risk assessment - The WHO Human Health Risk Assessment Toolkit [8]. This manual “provides users with guidance to identify, acquire and use the information needed to assess chemical hazards, exposures and the corresponding health risks in their given health risk assessment contexts at local and/or national levels. The Toolkit contains road maps for conducting a human health risk assessment, identifi es information that must be gathered to complete an assessment and lists electronic links to international resources from which the user can obtain information and methods essential for conducting the human health risk assessment.” [8]

A lot of others chemical risk assessment templates and guides are also developed by various scientifi c and business entities [9, 10 and 11]. What is the common between all these guidelines, templates and manuals? What is the essence every medical specialist has to know for performing proper chemical hazard health risk assessment, in order to protect rescue and medical teams and to organize effi cient medical support to the affected? All the guidelines are unanimous – for proper chemical health risk assessment is required data about the type and the characteristics of the chemical agent and data about the exposure of the affected.

By the defi nition human health risk assessment is a process intended to estimate the risk to a given target organism, system or (sub)population, including the identifi cation of attendant uncertainties, following exposure to a particular agent, taking into account the inherent characteristics of the agent of concern as well as the characteristics of the specifi c target system [12]

The risk assessment includes four steps: 1) hazard identifi cation, 2) hazard characterization, 3) exposure assessment and 4) risk characterization (fi g 1)

Therefore, the education and training of the medical specialist have to include these four steps:

Page 360: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

358

Collecting information and taking samples for identifying the present in the area of damage chemical agent. The medical specialists have to know where to look for such an information and that obtaining this information is the fi rst, basic, a must step in the medical support planning and execution.

The second step is to analyze the possible health harm the identifi ed chemical hazard could cause. In general the dangerous for the human health chemicals could be classifi ed in accordance with their toxicity in the following groups:

1. Cancer Developmental Toxicity;2. Reproductive Toxicity;3. Cardiovascular or Blood Toxicity;4. Endocrine Toxicity;5. Gastrointestinal or Liver Toxicity;6. Immunotoxicity;7. Kidney Toxicity;8. Musculoskeletal Toxicity;9. Neurotoxicity;10. Respiratory Toxicity;11. Skin or Sense Organ Toxicity. [13]

Fig 1. The environmental health chainpublished originally in EHC 214 (IPCS, 2000)

Page 361: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

359

After analyzing the probable effects of the present hazard the analysis continues by evaluating the population at risk exposure – does the hazard could enter by its route or the population has taken the protection and the route of exposure is blocked accordingly. Time of onset of the exposure has also to be considered, as well as duration of the exposure. All this data is essential for assessing the expected health damages to be created and related to them medical activities.

The last forth step is to educate the medical specialist on how to evaluate the risk level based on the obtained data regarding the hazard identifi cation and characterization and to exposure and severity assessment.

The key output from the training should be that the medical specialists are confi dent in their knowledge about the chemical hazards and poses suffi cient information about where to ask and fi nd valuable data about the chemicals characteristics and about the related to them negative impact on human health.

The authors, based on their study fi ndings, would like to propose establishing of working group for setting up of tutorial guidance and unifi ed program for medical specialists’ postgraduate training on chemical hazards health risk assessment.

References

[1] Mosby’s Medical Dictionary 8 edition Elsevier Health Sciences, 2008, ISBN 0323052908[2] De Noon Daniel J. Biological and Chemical Terror History Lessons Learned? // http://www.webmd.com/a-

to-z-guides/features/biological-chemical-terror-history[3] http://www.cbwinfo.com/History/Incidents.html[4] http://www.emdat.be[5] http://www.unisdr.org/we/inform/disaster-statistics[6] Lessons of Bhopal: 25 Years Later, U.S. Chemical Laws Need Strengthening http://www.foreffectivegov.

org/node/10585[7] Emergency Planning and Community Right to Know Act (EPCRA) http://www.epa.gov/osweroe1/content/

lawsregs/epcraover.htm[8] WHO Human Health Risk Assessment Toolkit: Chemical Hazards http://www.who.int/ ipcs/publications/

methods/harmonization/toolkit.pdf[9] http://www2.ul.ie/web/www/faculties/science_&_engineering/departments/chemical_&_environmental_

science/resources/chemical%20risk%20assessment%20template[10] http://www.ul.ie/hr/sites/default/fi les/docs/Health%20And%20Safety/Chemical%20Risk%20Assessment.

pdf[11] http://www.worksafe.vic.gov.au/__data/assets/pdf_fi le/0017/21617/checklist_for_chemicalsvs4.pdf[12] IPCS (2004) IPCS risk assessment terminology. Part 1: IPCS/OECD key generic terms used in chemical

hazard/risk assessment. Part 2: IPCS glossary of key exposure assessment terminology. Geneva, World Health Organization, International Programme on Chemical Safety (Harmonization Project Document No. 1; http://www.who.int/ipcs/methods/harmonization/areas/ipcsterminologyparts1and2.pdf

[13] http://scorecard.goodguide.com/

Page 362: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

360

Chapter 48

Neonicotinoid Pesticides – Some Medical Intelligence Concerns

Col Dr Rostislav KOSTADINOV, MD, PhD1, Dr Giussepe NOSCHESE, MD2;LTC Dr Georgi POPOV, MD, PhD1

1Military MedicalAcademy, Sofi a, Bulgaria2AORN “A.Cardarelli”, Naples, Italy

Abstract Despite the fact that the fi rst research efforts about implementation of new class insecticides started in the late 80-s of the last century, nowadays there are no doubt that the neonicotinoids are already the world’s most widely deployed insecticides.Most of the studies are assuring the low mammals and human toxicity of these insecticides, because of the different structure of the nicotine receptors in humans and interverbrals, but from the time of their implementation to day a lot of evidence about effects on human health have been reported.The aim of this study is to present some Medical Intelligence concerns about possible implementation of the neonicotinoids as chemical weapons. By the means of descriptive and comparative methods the available data about chemical features of this class insecticides and reported their impact on human health are analyzed. Deductive analysis was applied in order to depict the main challenges community and healthcare systems could face in case of implementation of neonicotinoids for terrorist’s purposes. As a conclusion the authors highlighted the requirement for detailed research on the neonicotinoid class insecticides regarding their possible implementation by terrorists organizations.

Key words: Neonicotinoids; Medical Intelligence, Human ToxicityIntroduction Neonicotinoids are pesticides created for altering the nervous system function of the

insects. [1] First experiments began in 80’s of the last centuries in PurdueUniversity, where fi rst neonicotinoid was synthesized – nithiazin. Its synthesis was result of research on the nicotine toxity to insects. It was known that the nicotine is toxic to the insects, but also cause severe damage to the mammals. The scientists began to search for compound that retains the insects’ toxity but is not or at least less harmful to the mammals, humans respectively. The fi rst insecticide from this new class called neonicotinoids, the nithiazin, is extremely toxic for insects, with moderate to low toxity for rats. This product was not utilized in the practice, because of its photo instability. But the achieved result – created insecticide, that is not blocking the cholinesterase activity, but reacting with the postsynaptic nicotine receptors and causing nerve paralysis and following death of insects with low mammal toxity, encouraged the scientists to continue the research, thus leading to synthesis, by

Page 363: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

361

Bayer, of photostable insecticide of the new class, suitable for industrial implementation –imidaloprid.

What is the difference between the widespread organophosphate insecticides and the newly created neonicotinoids that led to the change in the agriculture practice and the shift to the neonicotinoids? The answer is in the different mode of action of the two class insecticides. The organophosphates are binding with the cholinesterase, thus inactivating the enzyme resulting in excess of acetylcholine and Muscarin and Nicotine clinical picture leading to death of the poisoned insects and mammals. [2, 3 and 4] Despite the effi cacy of the organophosphate insecticides the health risk to the operators and consumers remained on high levels thus requiring specifi c precautions to be implemented while working with organophosphates. All this is due to the fact that the organophosphates are acting with the same mode of toxity both in the insects and in mammals.

Neonicotinoids are agonists at the nicotinic acetylcholine receptors (nAChRs), particularly the α4β2 subtype [5], [6], which induces neuromuscular paralysis and eventually death. They are highly selective for nAChRs in insects compared with mammals, which should reduce morbidity and mortality in cases of human poisoning. This diverse selectivity is explained by the structural differences in nAChRs that affect how strongly particular molecules bind, both in the composition of the receptor subunits and the structures of the receptors themselves.[7, 8] Therefore, most neonicotinoids show low affi nity for mammalian nicotinic acetylcholine receptors (nAChRs) while exhibiting high affi nity for insect nAChRs. The low mammalian toxicity of neonicotinoids can be explained in large part by its lack of a charged nitrogen atom at physiological pH that can penetrate the insect blood–brain barrier, but not the human one. Nicotine, like the acetylcholine, has a positively charged nitrogen (N) atom at physiological pH, which binds to the negatively charged site of the nicotine receptor nAChRs. What is more the nicotine is also highly lipophilic at physiological pH, therefore it is quickly and widely distributed, despite the fact that the blood–brain barrier reduces access of ions to the central nervous system. On the other hand neonicotinoids have a negatively charged nitro or cyano group that could only bind to the positively charged amino acid residue present on insect, but not mammalian nAChRs, resulting is species specifi c toxicity.

The described mode of action of neonicotinoid pesticides, modeled after the nicotine is affecting the central nervous system of insects, causing excitation of the nerves and eventual paralysis, which leads to death. Because this group is extremely effective against sucking and chewing insects, soil insects, whitefl ies, termites, turf insects, and Colorado potato beetle, in the last decades they become leaders on agricultural market - in 2008 they represented 24% of the global market for insecticides and were even more important in the market for seed treatments with almost 80% of the all seed treatment sales.

This widespread is related not only due to neonicotinoids insects’ high toxity but more due to its low mammals toxity - are classifi ed by the EPA as both toxicity class II and class III agents and are labeled with the signal word “Warning” or “Caution.” The recorded data regarding their toxity to warm blooded indicates that neonicotinoids are less toxic when absorbed by skin or inhaled, compared to ingestion. Signs of toxicity shown during experiments with rats presented minor eye reddening, lethargy, respiratory disturbances, decreased movement, staggering gait, occasional trembling, and spasms.

Page 364: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

362

Despite all this theoretical and experimental fi ndings in last decades a lot of evidence regarding unexpected neonicotinoids’ toxicity to others than insects species were recorded. It was proved that during the metabolism of imidoclopramid (the most widespread neonicotinoid) in the mammals’ bodies a desnitro-imidacloprid is formed. This compound has charged nitrogen and shows high affi nity to mammalian nAChRs [7], leading to symptoms of nicotine’s poisoning. The same product is formed and during the environmental breakdown of the imidoclopramid. What is more it is recorded that the time required for degradation of the neonicotinoids in the absence of sunlight and microorganism activity is 3,8 years and the insecticides could accumulate in the environment leading to the increased presence in the soil, surface and underground waters. Some independent studies’ results are concerning the eco activists, because of the possible accumulation of the neonicotinoids applied agriculturally in aquifers and from them to pose threat to the population utilizing the wells. [9] Other studies have proved the neonicotinoids highly, lethat toxicicty to the pollinators and especially to the bees. [10, 11, 12 and 13]

These fi ndings led to registration review in USA - “Some uncertainties have been identifi ed since their initial registration regarding the potential environmental fate and effects of neonicotinoid pesticides, particularly as they relate to pollinators. Data suggest that neonicotinic residues can accumulate in pollen and nectar of treated plants and may represent a potential exposure to pollinators. Adverse effects data as well as beekill incidents have also been reported, highlighting the potential direct and/or indirect effects of neonicotinic pesticides. Therefore, among other refi nements to ecological risk assessment during registration review, the Agency will consider potential effects of the neonicotinoids to honeybees and other pollinating insects.” [14] European Union also took measures – European Food and Safety Authority (EFSA) scientists have identifi ed a number of risks posed to bees by three neonicotinoid insecticides [15] that led to the EU “restriction that will prevent the use of three neonicotinoid products - clothianidin, imidacloprid and thiametoxam - in seed treatment, soil application (granules) and foliar treatment on plants and cereals (with the exception of winter cereals) that are attractive to bees”. [16]

A lot of studies have been also published regarding the neonicotinoid toxicity to humans resulting even in deaths. [17, 18, 19, 20, 21, 22, 23 and 24]

The aimThe aim of this study is to present some Medical Intelligence concerns about possible

implementation of the neonicotinoids as chemical weapons.Results and DiscussionsThe main objective of Medical Intelligence is to assess the health threat in particular

area of interest. With the world globalization nowadays it is becoming more diffi cult to identify the area of interest and to exclude from the analyses and monitoring the health hazards leading to increasing the health threat for the population in particular area of the world. Based on the interconnectivity of the world economies and countries stability the possibility of deteriorating medical situation in one country could lead to the effects in far located communities. Therefore, the newly published studies regarding the neonicotinoids’ toxicity have attracted the attention of the medical intelligence analysts. On the fi rst glance the articles published by eco-activists and bird association regarding the neonicotinoids effects on bees colonies and birds population is hardly to be assessed as medical

Page 365: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

363

intelligence topic of interest, but reading carefully the above mentioned publications and especially those describing the suicide attempts, some of them successful, by ingestion of neonicotinoids are leading the medical intelligence analysts to requirement to evaluate the probability these insecticides to be used deliberately as a chemical weapons.

The Organization for Prohibition of Chemical Weapons defi nes chemical weapons much more generally in comparison with the general and traditional defi nition of a chemical weapon as a toxic chemical contained in a delivery system, such as a bomb or shell. In the Convention defi nition “the term chemical weapon is applied to any toxic chemical or its precursor that can cause death, injury, temporary incapacitation or sensory irritation through its chemical action. Munitions or other delivery devices designed to deliver chemical weapons, whether fi lled or unfi lled, are also considered weapons themselves.” [25]

Analyzing this defi nition it is easy to put the neonicotinoids in the array of the chemical weapons. Scientists and researches in the studies mentioned above, have described the possible and recorded effects of ingested neonicotinoids on the human health. The recorded clinical picture depends on the ingested dose. The light clinical picture includes:

• Nausea; • Vomiting;• Headache;• and diarrhoea.The moderate one is manifested with:• Drowsiness;• Disorientation;• Dizziness;• Oral and gastroesophageal erosions to hemorrhagic gastritis;• Productive cough;• Fever;• Leukocytosis and Hyperglycemia. When the ingestion is massive, then a life threatening clinical picture, resembling the

nicotine poisoning is developed:• Diffi cult breathing with rapid breathing in the beginning, followed by respiratory

failure - decreased breath excursions and stop of the breathing • Heartbeat - pounding and rapid, followed by slow heart rate Coma• High blood pressure, which then drops• Depression and Confusion• Convulsions• Coma and death Aspiration pneumonia is also associated with the life threatening clinical manifestation

of the neonicotinoids poisoning. All the clinical manifestation could temporary incapacitate the affected population and

depending on the dose could lead to sensory irritation and death.What is more the neonicotinoids could be used not only as a weapon against the

individual health, but could be more effectively utilized for undermining the particular community health and wellbeing by affecting the pollinators’ colonies and water sources.

Page 366: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

364

Decreasing the pollinators colonies for years could lead to decreased harvest, thus creating food instability and related to it poverty, malnutrition, impaired individual and collective immunity, increase in the infectious disease morbidity, social unrests and migration. Contamination of the water sources will affect the population with chronic intoxication, poultry, and vegetation leading to the already described consequences.

The other source of medical intelligence concern is the availability of these products on the market. The neonicotinoids could be purchased all over the world in unrestricted quantities. There is no ban for trans-boundaries transportation and no one will doubt the seed treatment with the insecticides. All these conditions facilitating the deliberate use of neonicotinoids for individuals or population health damage are, from medical intelligence point of view, converting, transforming the neonicotinoids from insecticides with moderate to low mammals’ toxicity into powerful and covert terrorist chemical weapon. The deliberate utilization of neonicotinoids against entire tribe, national minority or population living in contested regions, even by unstable governments could not be excluded as an option.

Presenting this medical intelligence analysis the authors would like to increase the medical and agriculture communities’ awareness about the possible unconventional use of the at fi rst glance harmless chemical compounds, created for humanity wellbeing. The history of the organophosphate compounds fi rst synthesized for insecticide use, but rapidly convert into chemical weapons, utilized even nowadays, could be an example for transformation of the scientists’ achievements into terrorists’ means.

References

[1] Fishel Frederick M. Pesticide Toxicity Profi le: Neonicotinoid Pesticides // http://edis.ifas.ufl .edu/pi117[2] Leikin JB, Thomas RG, Walter FG, Klein R, Meislin HW. A review of nerve agent exposure for the critical

care physician. Crit Care Med. Oct 2002;30(10):2346-54.[3] Clark, RF. Insecticides: Organic Phosphorus compounds and Carbamates. In: Goldfrank LR, FlomenbaumNE,

Lewin NA, Nelson LS, Howland MA, Hoffman RS, eds. Goldfrank’s Toxicologic Emergencies. Stamford, Ct: Appleton & Lange. 8th ed. 2006:1497-1512.

[4] Barthold CL, Schier JG. Organic phosphorus compounds-- nerve agents.Crit Care Clin. Oct 2005;21(4):673-89, v-vi.

[5] Tomizawa M, Casida JE (2005) Neonicotinoid insecticide toxicology: mechanisms of selective action. Annu Rev Pharmacol Toxicol 45: 247–268.

[6] Tomizawa M, Casida JE (2003) Selective toxicity of neonicotinoids attributable to specifi city of insect and mammalian nicotinic receptors. Annu Rev Entomol 48: 339–364.

[7] Tomizawa, M. (2004). “Neonicotinoids and Derivatives: Effects in Mammalian Cells and Mice”. Journal of Pesticide Science 29 (3): 177–172.

[8] Tomizawa, Motohiro; Latli, Bachir; Casida, John E. (1999). “Structure and Function of Insect Nicotinic Acetylcholine Receptors Studied with Nicotinic Insecticide Affi nity Probes”. In Yamamoto, Izuru; Casida, John.Nicotinoid Insecticides and the Nicotinic Acetylcholine Receptor.Tokyo: Springer-Verlag. pp. 271–292.

[9] Intervju z mikrobiologom: Ta prostor je prestreljen z lobisti multinacionalk // http://www.delo.si/brez%20kategorije/intervju-z-mikrobiologom-ta-prostor-je-prestreljen-z-lobisti-multinacionalk.html

[10] Whitehorn, P. R.; O’Connor, S.; Wackers, F. L.; Goulson, D. (2012). “Neonicotinoid Pesticide Reduces Bumble Bee Colony Growth and Queen Production”. Science 336 (6079): 351–352.

[11] Tapparo, A.; Marton, D.; Giorio, C.; Zanella, A.; Soldа, L.; Marzaro, M.; Vivan, L.; Girolami, V. (2012).

Page 367: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

365

“Assessment of the Environmental Exposure of Honeybees to Particulate Matter Containing Neonicotinoid Insecticides Coming from Corn Coated Seeds”. Environmental Science & Technology 46 (5): 2592.

[12] Schneider, C. W.; Tautz, J. R.; Grьnewald, B.; Fuchs, S. (2012). “RFID Tracking of Sublethal Effects of Two Neonicotinoid Insecticides on the Foraging Behavior of Apis mellifera”. In Chaline, Nicolas. PLoS ONE 7 (1): e30023.

[13] Krupke, C. H.; Hunt, G. J.; Eitzer, B. D.; Andino, G.; Given, K. (2012). “Multiple Routes of Pesticide Exposure for Honey Bees Living Near Agricultural Fields”.In Smagghe, Guy. PLoS ONE 7 (1): e29268.

[14] Pesticides: Registration Review // http://www.epa.gov/oppsrrd1/registration_review/highlights.htm#nn[15] EFSA identifi es risks to bees from neonicotinoids // http://www.efsa.europa.eu/en/press/news/130116.htm[16] European Commission confi rms December 1 neonicotinoid ban // http://www.farmersguardian.com/home/

arable/european-commission-confi rms-december-1-neonicotinoid-ban/55842.article[17] Mohamed F, Gawarammana I, Robertson TA, Roberts MS, Palangasinghe C, et al. (2009) Acute Human

Self-Poisoning with Imidacloprid Compound: A Neonicotinoid Insecticide. PLoS ONE 4(4): e5127. doi:10.1371/journal.pone.0005127

[18] Agarwal R, Srinivas R (2007) Severe neuropsychiatric manifestations and rhabdomyolysis in a patient with imidacloprid poisoning. Am J Emerg Med 25: 844–845.doi:

[19] Proenca P, Teixeira H, Castanheira F, Pinheiro J, Monsanto PV, et al. (2005) Two fatal intoxication cases with imidacloprid: LC/MS analysis. Forensic Sci Int 153: 75–80. The imidacloprid blood concentrations found in the two-fatal cases postmortem were 12.5 and 2.05μg/mL.

[20] Wu IW, Lin JL, Cheng ET (2001) Acute poisoning with the neonicotinoid insecticide imidacloprid in N-methyl pyrrolidone. J Toxicol Clin Toxicol 39: 617–621

[21] Huang NC, Lin SL, Chou CH, Hung YM, Chung HM, et al. (2006) Fatal ventricular fi brillation in a patient with acute imidacloprid poisoning. Am J Emerg Med 24: 883–885.

[22] David D, George IA, Peter JV (2007) Toxicology of the newer neonicotinoid insecticides: imidacloprid poisoning in a human. Clin Toxicol (Phila) 45: 485–486.

[23] Faul J (1996) NTN 33893-Occupational medical experiences. Unpublished report from Bayer AG, Medical department, dated March 1996. Submitted to WHO by Bayer AG, Mannheim, Germany.

[24] Calumpang SM, Medina MJ (1996) Applicator exposure to imidacloprid while spraying mangoes. Bull Environ Contam Toxicol 57: 697–704

[25] Brief Description of Chemical Weapons // http://www.opcw.org/about-chemical-weapons/what-is-a-chemical-weapon/

Page 368: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

366

Page 369: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

367

Part 5

CHEMICAL TERRORISM, DIAGNOSIS AND TREATMENT OF EXPOSURE TO

CHEMICAL AGENTS

Page 370: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

368

Page 371: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

369

Chapter 49

Chemical Weapon Information Revealed by Aum Shinrikyo Death Row

Inmate Dr. Tomomasa Nakagawa

Anthony T. TU Department of Biochemistry and Molecular Biology,

Colorado State University, Fort Collins, USA

Abstract

The Toyko subway attack with sarin on March 20, 1995 killed 13 people and injured over 5,000. An earlier sarin attack on June 27, 1994 in Matsumoto City killed 7 people and injured over 500. All thirteen senior member of the cult were sentenced to death and much important information about the group’s criminal activities are still kept secret by the cult members who are on death row at the Tokyo Detention Center in Japan (photo 1).

Photo 1. Tokyo Detention Center in Kosuge, Japan where all 13 senior members of Aum Shinrikyo on deathrow

In order to learn more information about the cult’s chemical and biological weapons, I met with Dr. Nakagawa at the Tokyo Detention Center in Tokyo three times, December 14, 2011, June 11, 2012 and October 16, 2012. Normally the meeting time with a death row inmate is limited to 10 minutes, but I was granted a 30-minute meeting; this was a rather exceptionally long time. It might be due to the fact that I helped the Japanese Police identify the location of the Aum’s sarin manufacturing. I was anxious to answer many questions that I had, so I took the initiative to ask Dr. Nakagawa many questions (photo 2).

Page 372: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

370

1. How Did Aum Shinrikyo Get the Idea to Make Sarin and VXA. SarinAum got the idea to use sarin from a book that Tsuchiya had translated to Japanese,

“The Story of Poisons,” written by D. Vachivarov and G. Nedelchef in Bulgaria.

B. Large Production of Sarin and the 7th SatyanLarge scale production of sarin, with 70 tons being the target, was made at the 7th

Satyan in Kamikuishiki Village, in the Nagano Prefecture. Mr. Takizawa was in charge of the actual construction of the building and facilities. Dr. Nakagawa gave a book entitled, “A FOA Briefi ng Book on Chemical Weapon’s Threats, Effects and Protection (ISBN 91-7056-085, 4, 1992) published by the Swedish Defense Department. Inside the book there was photographs of sarin production in Iraq. Mr. Takizawa told Dr. Nakagawa that the book was extremely useful.

C. VXDr. Nakagawa said the idea of the use of VX was obtained from the article I wrote

in 1994 for the September issue of “Chemistry Today.” The Japanese Police read my article and asked me to help them. The Japanese Police used this method to fi nd the methylphosphonic acid from the soil in the area of 7th Satyan at Aum Shinrikyo’s 70-ton sarin manufacturing site. I also briefl y mentioned VX in this article. In order for it to not be misused by some crook, I deliberately mentioned it briefl y. I only mentioned that VX can be made from CH3P(O)Cl2 through CH3P (O) Cl, OC2H5. At that time Tsuchiya was making Sarin from PCl3 through many intermediates. One of the intermediate compounds they has was CH3P(O)Cl2 and Tsuchiya got the idea immediately that he could make a CH3P(O)Cl, OC2H5 from it and eventually make VX. This was quite a surprise to me. Tsuchiya started to make VX one month after he read this article.

D. Use of diethylaniline in Sarin Synthesis Aum Shinrikyo used diethylaniline twice; the fi rst and the last steps. They used

diethylaniline to neutralize the acid released.

E. Destruction of Sarin at the Beginning of 1995Yomiurui Shinbun, a leading newspaper in Japan published the news in the

January 1, 1995 issue of their paper, revealing that the Japanese Police analyzed the soil and found the organophosphate. This news really shocked Asahara who ordered all the sarin they manufactured to be destroyed. There were considerable amounts of difl uoromethylphosphonic acid left that is the product before fi nal sarin production. Later this difl uro compound was used for the Tokyo subway attack on March 20, 1995. I thought Dr. Nakagawa deliberately saved the difl uro compounds.

So, I asked Dr. Nakagawa, “Why did you hide the difl uro compound”? He said, “Oh no, we decided to destroy the sarin so that the Japanese Police would not know we made sarin when they came to our facility to inspect. But it was too much and simply we were no longer able to destroy all the sarin we had. Mr. Tsuchiya and I did not sleep for 3 days because we were trying to destroy all of the sarin we manufactured. I myself kept injecting

Page 373: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

371

PAM intravenously in order to destroy the sarin they had for 3 days. Simply there was too much sarin so we gave up and hid the unfi nished sarin”. This surprised me, but I was glad to fi nd out the truth.

F. More than Sarin and VX in the Aum Shinrikyo’s ArsenalI was shocked to learn that Aum Shinrikyo had more chemical weapons than anyone

anticipated. They actually synthesized 20g of soman (GD), 20g of tabun (GA) and 20g of cyclosarin (G7) in April 1994. Mustard gas was made in a larger quantity, 200kg, in December 1994. They also made phosgen and used it once. They did not use these chemical agents, with the exception of phosgen, because only relatively small quantities were synthesized (fi g 1.)

Fig 2. Chemical agents made by Aum Shinrikyo other than sarin and VX

These facts are not well known to the public. Why didn’t the Japanese know about these deadly chemical agents? There are probably two reasons that account for this. One is that the police investigation focused on the collection of information related to Aum’s criminal acts. If some chemical agents are not used then they remain outside of the police investigation. The second reason is attributed to the methods of Japanese Police investigations. In Japan, the special inquiry offi ce conducts questioning of a criminal; they do not know any science or chemistry. Therefore, when it comes to questions involving chemistry, the offi cer simply lets the criminals write the answers themselves. So the criminals do not have to answer if it was not asked.

G. Different Ways to Make SarinThe Aum Shinrikyo made sarin mainly in the laboratory at the Kushiti Galva prefab

(Tsuchiya’s lab.) Aum Shinrikyo made sarin using 5 steps, and they modifi ed the steps depending on the attack the sarin was to be used for.

Page 374: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

372

Photo 2. The Author in front of three sarin manufacturing sites at Kamikuishiki. The left prefab was used to manufacture the sarin used in the Tokyo Subway attack. The middle prefab was used by Tsuchiya to make sarin used in the Matsumoto City attack. The right building was called the 7th Satyan and was supposed to be used to manufacture 70 tons of sarin. But sarin production here was stopped at the 3rd step, so the 4th and 5th steps were never used.

1. Sarin Used for the Matsumoto City AttackThe sarin used for the Matsumoto City attack was made from the second step; the fi rst

step was omitted. They used it in Matsumoto City on the night of June 26, 1994.

2. Tokyo Subway AttackThey had a large stock of the sarin precursor, difl uro-methylphosphonic acid, so only

the fi nal step was used to make sarin overnight. Because they were pressed for time, they did not purify the fi nal product and used it in the condition in which it was obtained. If the sarin had been purifi ed, there would have been more casualties.

3.Sarin Manufacture at the 7th SatyanThe 7th Satyan was a huge building and Aum Shinrikyo tried to make 70 tons of sarin

here. They gained experience in making sarin on a laboratory bench starting from the second step, by purchasing P(OCH3)3 . They never had experience making the fi rst step, even at their lab.

Asahara was anxious to make sarin in large quantities at the 7th Satyan and 70 tons was their goal. Because of their lack of experience, they failed many times to make P(OCH3)3 from PCl3. This caused the fi rst step reaction mixture to leak through to the outside twice. Japanese Police were unable to detect the identity of noxious gases at that time. By trial and error, the Aum eventually succeeded to the third step of sarin production. On January 1, 1995 Yomiuri Shinbun published in a newspaper that the Japanese Police detected organophosphates from the soil of Kamikuishiki, where the 7th Satyan was located. This news alerted Asahara and he ordered the Aum to stop the manufacture of sarin and they tried to destroy all the sarin and its precursors that they possessed. So, at the 7th Satyan large-scale production of sarin was also stopped at the 3rd step.

Page 375: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

373

H. Where Did the Methylphosphonic Acid in the Soil Come From?In November 1994, there was a noxious gas leak from the 7th Satyan and the Japanese

Police were able to detect methylphosphonic acid in the soil.

1.Methylphosphonic Acid In the SoilThe discovery of methylphosphonic acid (MPA) in the soil as, reported by the Japanese

Police, really puzzled the senior members of Aum Shinrikyo. How can one discover the sarin degradation product, MPA, without sarin being produced? Aum Shinrikyo’s members thought the Japanese Police fabricated this information.

Matsumoto’s sarin was made at Tsuchiya’s lab. Tsuchiya’s lab and the 7th Satyan were next to each other. Dr. Nakagawa said the soil around Tsuchiya’s lab was heavily contaminated by sarin and its precursors. So he thinks the MPA that the Japanese Police found came from Tsuchiya’s lab, not the 7th Satyan.

2. Methylphosphonic Acid from Inside the 7th Satyan The Japanese Police said they detected methylphosphonic acid from the soil near the

7th Satyan. But the Japanese Police published two articles saying that they also found methylphosphonic acid inside the 7th Satyan. By interviewing Dr. Nakagawa I found a great discrepancy between the data published by the Japanese Police and the actual sarin manufacturing at the 7th Satyan. Japanese Police said they found methyphosphonic acid, thus they obtained direct evidence of sarin manufacturing at the 7th Satyan. But there was no sarin manufacture at the 4th and 5th steps. This mystery should be solved in the future by further investigation

Page 376: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

374

Chapter 50

Research of Mobility of Sodium Arsenite in Soils of Areas of Destruction of the Chemical Weapon

Vadim PETROV, Marina SHUMILOVA, Olesya NABOKOVA and Marina LEBEDEVA

Institute of mechanics of Ural Branch of the Russian Academy of Sciences

Abstract: The estimation in laboratory conditions of mobility of sodium arsenite in pollution soil is spent. Such technogenic pollution could result from works on destruction of lewisite and processing of reactionary masses of detoxifi cation of lewisite on method of alkaline hydrolysis. High mobility of sodium arsenite in the polluted soil samples and the tendency of pollution by arsenic to delocalization is shown, that confi rms data of monitoring of works on destruction of lewisite and at sanitation of the polluted territories.

Keywords: Chemical weapon destruction, technogenic emissions of arsenic, soil pollution

Introduction

In 2009 in Kambarka, the Udmurt Republic, at realization of the International Convention on the chemical weapon (CW) it has been destroyed more than 6 thousand tons of poison gas containing arsenic - lewisite. As a result of use of a method of alkaline decomposition of lewisite and the subsequent evaporation of reactionary masses it has been received more than 10 thousand tons of the dry salts containing sodium arsenite (SA) and sodium chloride. In 2011-2012 works on dismantle of a part of the equipment, neutralization of territory of object are spent. Also the question of a reshaping of object in connection with necessity of processing of a considerable quantity of toxic reactionary masses containing arsenic is considered. Thereupon the most dangerous pollution is арсенит sodium, because of its solubility and high toxicity [1]. We had been studied in laboratory conditions mobility in soil of SA for preparation of recommendations about neutralization of the polluted territories and organization of monitoring.

Methods and materials

Mobility of SA in soil it was studied at the experimental stand [2]. The stand is a design from several columns and a portioning device. Pollution is entered into columns with samples of soil. Then through polluted the sample the distilled water was passed from a portioning device. In the bottom of a column the fi ltering device was established and fractions of the water which has passed through polluted sample were selected. The stand modelled atmospheric

Page 377: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

375

infl uences in the form of a rain on soil. Speed of passage of water through polluted the sample, and also volume of the passed water was defi ned. Researches were spent at temperature of a laboratory room. For calculation of constants of speed of allocation of SA in water elements of the theory of heterogeneous chemical processes were used. During researches following substances, materials and reagents were used: the various samples of soil typical for region, with the various maintenance of humus; peat (the maintenance of humus not less than 25%, pH - 5,5 - 6,0); the washed out building sand; SA - NaAsO2, has been received from the Karaganda technological institute (Republic Kazakhstan). The arsenic maintenance defi ned a method atom absorption spectroscopy on device “Shimadzu” - АА7000.

Results and discussion

Decomposition of lewisite by method of alkaline hydrolysis can be described reaction:

C2H2AsCl3 + 6NaOH → 2C2H2 + Na3AsO3 + 3NaCl + 3H2O (1).

The reactionary masses then evaporated and received dry salts with the maintenance of SA ~ 52 % of weights. At research of mobility of SA samples of soil, sand and peat polluted different quantity of substance. Pollution of samples carried out of 10MAC (technological failure) and 100MAC (emergency situation) on Аs (MAC (maximum allowable concentration) Аs in soil = 2,0 mg/kg of dry soil) [3]. Further at the experimental stand allocation degree of SA from samples was defi ned at giving on them of the distilled water with certain speed (ω ~ 2,8.10-2 ml/s). On Figure 1-3 allocation degree of SA from the various polluted samples of soil is shown. Also allocation of SA from the polluted samples of peat and sand was investigated.

Figure 1. Dependence of degree of allocation of SA- α(in shares from the initial maintenance) from water volume - V (ml), the soil passed through

the sample № 1, with the maintenance of humus is 3,73 % of weights

Figure 2. Dependence of degree of allocation of SA- α (in shares from the initial maintenance) from water volume - V (ml), the soil passed through

the sample № 2, with the maintenance of humus is 5,96 % of weights.

0,00E+001,00E-022,00E-023,00E-024,00E-025,00E-02

5015

0250 35

045

055

065

0750 85

095

0105

01150

1250

10-MAC 100-MAC

0,00E+00

5,00E-02

1,00E-01

1,50E-01

50 150

250

350

450

550

650

750

850

950

1050

1150

1250

10-MAC 100-MAC

α

V

α

V

Page 378: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

376

Figure 3. Dependence of degree of allocation of SA- α (in shares from the initial maintenance) from water volume - V (ml), the soil passed through the sample № 3, with the maintenance

of humus is 2,55 % of weights.

For the experimental stand the constant of speed of process of allocation of SA from the sample of soil can be calculated at formula integration:

(2),

where α- quantity of the substance allocated from soil in shares from the initial maintenance, к - an observable constant of speed of allocation of substance from soil, ω-speed of passage of water through polluted the sample, n - degree of the kinetic equation, V - volume of submitted water.

On the basis of the data resulted on Figure 1-3, it has been defi ned, that degree of the kinetic equation of allocation of SA from the polluted samples it is close to 2. Constants of speed of allocation of SA, which are resulted in the table, have been defi ned. Values of constants of speed allow to make an estimation of the period of semideducing of substance of the soil sample at α = 0,5, that is an indicator of behaviour of technogenic pollution in environment. For defi nition of the period of semideducing the formula (2) can be written down in a following kind:

(3),

where ТY, α- time necessary for allocation of substance from the polluted soil to degree α, in years; S - the area of a soil cover on which technogenic infl uence has been had; НY, i - annual height of a separate kind of an atmospheric deposits in the form of a rain (a weak rain, a rain, a strong rain), in mm; ωi- speed of passage of water through the polluted soil, ml/s, m - quantity of kinds of deposits in the form of a rain.

For defi nition of the period of semideducing (α = 0,5) the formula (3) can be written down in a following kind, at n = 2:

0,00E+00

1,00E-02

2,00E-02

3,00E-02

4,00E-02

5,00E-02

50 100

150

200

250

300

350

400

450

500

550

600

650

700

750

800

850

900

950

1000

1050

1100

1150

1200

1250

10-MAC 100-MAC

α

V

dVdn ω

καα =

− )1(

��=

=−

m

i in

HS�d

0

iY,Y,

0 )1( ωκ

αα

α

α

Page 379: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

377

(4).

In the Table values of the period of semideducing of SA from various soil samples, and also from peat and sand are resulted. The quantity and character of an atmospheric deposits was defi ned on the basis of data on 2011 for territory on which the object on destruction of lewisite in Kambarka settled down.

From the Table it is visible, that at increase in level of pollution of soil of a constant of speed of allocation of SA at atmospheric infl uence in the form of a rain essentially increase. SA in depending on a kind of soil and degree of its pollution possesses enough high mobility. Pollution of SA aspires to delocalization, has low values Т 0,5 that proves to be true data [4,5]. For example for pollution of oxides of heavy metals, such as Cu, Cd, Mn, Cr, the semideducing period makes some tens and hundreds years. Such feature for SA is connected with its high solubility that demands special measures at the organization of monitoring of technogenic infl uence by this substance, sanitation of the polluted territory after works on destruction of CW that is important taking into account high toxicity of this substance. The increase of quantity of humus in the polluted soil reduces constants of speed of allocation of SA.

Table Constants of speed of allocation and the period of semideducing

of SA from the polluted samples of soil, peat and sand

The polluted sample Mobility indicators of SA

Pollution 10 MAC on As

Pollution 100 MAC on As

The sample of soil № 1, the maintenance

of humus - 3,73 %

к, s-1 1,733.10-7 7,880.10-7

ТY,0.5, years 3,07 0,66

The sample of soil № 2, the maintenance

of humus - 5,96%

к, s-1 1,677.10-7 2,772.10-6

ТY,0.5, years 3,17 0,19

The sample of soil № 3, the maintenance

of humus - 2,55%

к, s-1 1,770.10-7 1,776.10-6

ТY,0.5, years 3,01 0,30

Peat,the maintenance of humus

~ 25%

к, s-1 5,186.10-7 2,789.10-6

ТY,0.5, years 1,03 0,19

River sand к, s-1 4,449.10-7 1,017.10-5

ТY,0.5, years 1,19 0,05

ConclusionsMobility in pollution soil of sodium arsenite under the infl uence of an atmospheric

deposits in the form of a rain is studied. Such pollution can be formed as a result of works on destruction of poison gases containing arsenic. High mobility of pollution and their aspiration to delocalization is established. It should be considered at the organization of monitoring for revealing of objectivity of technogenic infl uence and at sanitation of the polluted territories.

�=

= m

i i

HS

0

iY,5.0Y,

1

ωκ

Page 380: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

378

References

[1] Petrov, V. G, Lipanov, A.M., Trubachev, A.V., Chechina, A.A. Neutralisation of dangerous substances on the reorientated object on destruction of lewisite. Chemical physics and mesoscopia11(2009), 54-58.

[2] Petrov, V. G, Shumilova, M. A. Way of studying in laboratory conditions of mobility of technogenic pollution in soil.Chemical physics and mesoscopia14(2012), 257-260.

[3] Medical instructions 2.1.7.730-99-RF. A hygienic estimation of quality of soil of the occupied places, are entered 5.04.99, Moscow, 1999.

[4] Shumilova, M. A, Nabokova, O. S, Petrov, V.G. Features of behaviour of technogenic arsenic in natural objects. Chemical physics and mesoscopia, 2011. 13 (2011), 262-269.

[5] Shumilova, M. A, Nabokova, O. S, Petrov, V. G, Frizorger, G.G. About some features of behaviour arsenic containing substances at monitoring of object on destruction of lewisite. Vestnik of the Udmurt university, Series: Physics, Chemistry,1(2011),125-129.

Page 381: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

379

Chapter 51

The Control of Formation of Dioxins in Incinerators for Reactionary Masses of Destruction of the Chemical Weapon

Vadim PETROVa, Semon STOMPELb and Vladimir BUKOVb

a Institute of mechanics of Ural Branch of the RussianAcademy of SciencesbCompany “Safe Technologies”, St. – Petersburg, Russia

Abstract: According to the Program of destruction of CW now in the Russian Federation neutralization of phosphororganic substances on technologies with use of reagents is spent. One of problems of use of such technologies of destruction of CW is necessity of neutralization of a considerable quantity of reactionary masses and the polluted materials. Application of technology of high-temperature incineration for neutralization of reactionary masses is considered. Advantage of use of a method of thermal neutralization to reactionary masses is also possibility of a reshaping of objects on destruction of CW after end of conventional works.

Keywords: Destruction of the chemical weapon, reactionary masses, thermal neutralization

Introduction

In the Russian Federation 4th stage of performance of the Program on the chemical weapon (CW) is at the moment carried out. Its purpose is end of destruction of all kinds of CW in 2015. Last object on which works on the CWD should be spent, the object in settlement Kizner in the Udmurt Republic on which an artillery ammunition will be destroyed, mainly about phosphorus organic CW (POS) is. However, as we informed earlier [1,2], terms of works on the CWD most likely will be prolonged for various reasons. One of such reasons is technical complexity of destruction of a considerable quantity of an artillery ammunition methods with use of chemical reagents. The same complexities are available at the moment in the USA because of what the Program on the CWD is prolonged to 2023 [3]. From the point of view of the decision of some technical problems use of thermal methods of neutralization of CW which have well proved in the USA is defensible, both at neutralization of directly fi ghting reagents, and at neutralization of formed reactionary masses and a waste. In the given work we will consider possibility of use of thermal methods for neutralization of reactionary masses of destruction of POS.

Page 382: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

380

Discussion

Methods with use of chemical reagents which are used in the Russian Federation for neutralization of POS , lead to formation of toxic reactionary masses for which the approximate structure and quantity are shown in Table 1 [4]. From Table 1 it is visible, that the structure of such masses includes the organic substances containing phosphorus, nitrogen, fl uorine. Besides, in works can be used chlororganic solvents which also after application are necessary for neutralizing. Thus, at use of thermal methods of neutralization of reactionary masses by the basic technical decisions should be: the guaranteed destruction of dangerous toxic substances, clearing of departing gases from phosphorus oxide, fl uorine substances, on-possibility, nitrogen oxides. Also a condition of safety of a way is the synthesis exception of dioxins and dioxin-like substances above norms of ecological safety.

Such requirements are answered with a method of high-temperature incinerating. We will consider possibility of use for neutralization of reactionary masses incinerators, let out by the company «Safe Technologies», St.-Petersburg. On Figure 1,2 the kind of complexes of thermal decomposition (CTD), by productivity of 50 and 150 kg/hour is resulted. The complex of thermal decomposition can work for neutralization both liquid, and a solid waste. The technology assumes burning of substances at temperature 850-950 °С, reburning of smoke gases is a lot of oxidizer at temperature 1100-1200 °C. Clearing of departing gases is spent in a scrubber at introduction to departing gases of an alkaline reagent [5].

Table 1

Approximate structure and quantity of reactionary masses at destruction of POS

POSTechnology ofdestructions Approximate structure of reactionary

masses after destruction process

Quantity of reactionary

masses, kg/kg POS

Sarin, Soman Decomposition of monoethanolamin in the presence of water

3

V-gas Decomposition by a mix of N -methylpirrilidion, kalium isobytilat and isobytil spirit

3

Page 383: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

381

Figure 1. A complex kind on high-temperature neutralisation of a waste of Company «Safe Technologies» (St.-Petersburg), productivity of 50-70 kg/h, CTD-50

Figure 2. A complex kind on high-temperature neutralisation of a waste of Company «Safe Technologies» (St.-Petersburg), productivity of 150 kg/h, CTD-150

Also formation studying of dioxins has been spent at complex work on a waste which contained chlororganic substances. Maintenance defi nition of dioxins in departing gases of a complex has been spent, and also the quantitative estimation of formation of these substances in various zones of incinerator on the basis of kinetic and thermodynamic characteristics of reactions of synthesis [6] is spent. The quantity of dioxins in zones of incinerator can be defi ned from the formula:

Page 384: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

382

where c - concentration of the sum of polychlorinated dioxins and furans (PCDD/F) in departing gases, с0 - equilibrium value of concentration of the sum dioxins, к0, Е - kinetic parametres of reactions of synthesis and decomposition of PCDD/F, τ - time of stay of gases in a critical interval of temperatures.

Comparison of results of calculation with experimental data for CTD-50 is shown in Table 2. In Table 3,4 distribution of dioxins in various knots of incinerator CTD-50 and CTD-150 is shown.

Table 2

Comparison of values of the maintenance of dioxins in the smoke gases received at the analysis for incinerator with productivity of 50-70 kg/h and calculated on thermodynamic

and kinetic parametres of reactions

Maintenance of dioxins, pg/nm3

Results of the analysis Calculations

Total 64,6 33,7The maintenance in a toxic equivalent 5,9 3,0

Table 3

Distribution of dioxins in various knots of incinerator CTD-50, received on the basis of calculations of reactions of synthesis

The knot name Quantity of dioxins, pg/nm3

The burning chamber ≥ 0The reburning chamber 0,0Dry scrubber 10,8-12,9Deduster 4,3-20,8

Table 4Distribution of dioxins in various knots of incinerator CTD-150,

received on the basis of calculations of reactions of synthesis

The knot name Quantity of dioxins, pg/nm3

The burning chamber 0,0The reburning chamber 0,0Recuperator 0,0Scrubber 70,4

From comparison of results of the analysis and use of calculations it is possible to draw a conclusion, that they well correspond among themselves. Some difference between calculations and experimental data can be connected that in real conditions of operation process parametres sometimes differ from the established production schedules of work of the device. The carried out analysis shows, that the maintenance of dioxins in departing gases considerably below existing norms of ecological safety of incinerators (0,1 ng in a

τ))exp()exp(( ,0,00 RT

Ek

RTE

k�� d

ds

s −−−=

Page 385: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

383

toxic equivalent /nm3). The control of parametres of process of work of incinerators allows to exclude formation of dioxins above norms of ecological safety

As was above marked, by the important condition of works on destruction of the chemical weapon, owing to essential expenses of budgetary funds for the Program of CWD, and also at possibility of preservation of workplaces, the prospect of a reshaping of objects of the CWD for other purposes is. The reshaping of objects of the CWD after performance of conventional problems for neutralization of various kinds of a waste can be one of such directions. In Table 5 possible kinds of a waste which can be neutralized at a reshaping of object of the CWD in settlement Kizner are shown [7]. From tab. 5 it is visible, that for these kinds of a waste the method of high-temperature neutralization can be used. Thus, the part of the equipment of object of the CWD, and also its infrastructure can be kept for the decision of other problems.

Table 5The short characteristic of process of the CWD on object in settlement Kizner and offers on a reshaping of object after end of works on destruction of CW

The characteristic of processes on object of the CWD

Chemical decomposition of highly toxic substances; methods of neutralization of reactionary masses with reception of a waste of low toxicity; thermal methods of neutralization of a waste

Offers on a reshaping of object of the CWD

1. Neutralization of a dangerous industrial organic waste;2. Neutralization of old poisonous chemicals and pesticides.3. Neutralization and recycling of a waste of oil extracting. 4. Neutralization of a medical waste

Conclusions

Possibility of application of a method of high-temperature incinerating of toxic reactionary masses of destruction of POS and a waste of the CWD is considered. Perspectivity of use of a method at which use recycling of products of clearing of departing gases is possible, the maintenance of dioxins does not exceed norms of ecological safety, is shown. Method use allows to reorientate object of the CWD after performance of conventional problems for neutralization of other kinds of a waste.

References

[1] Petrov V., Shumilova M. Whether all chemical weapon in Russia in 2015 will be destroyed? A view from Udmurtia// Book of abstracts of CBMTS- Industry VII Int. Symposium, Cavtat-Dubrovnik, Croatia, (2011), 47-48.

[2] Petrov V.G., Shumilova M.A. The analysis of some problem aspects of chemical disarmament in Russia// Book of abstracts of CBMTS-IX Int. Symposium,Spiez, Switzerland, (2012) 50.

[3] Petrov V.G. Analiz of application of technology of high-temperature incinerating at destruction of the chemical weapon in the USA//the Bulletin of the Udmurt university, Series: Physics, chemistry.4 (2012), 51-58.

[4] Petrov V. G., Trubachev A.V. Some questions of destruction of the chemical weapon. Izhevsk: IAM UB RAS, 2004.

[5] Production schedules of thermal neutralization of a waste on installations (complexes) CTD- 50. S.Peterburg, 2012.

[6] Petrov V.G., Chechina A.A. The analysis conditions of formation polychlorinated dioxins and furans in zones of cooling of incinerators for arms destruction // Proc. of NBC 2006 Int. Symposium, Tampere, Finland, (2006), 263-270.

[7] Petrov V. G, Trubachev A.V. Reshaping of objects on destruction of the chemical weapon in the UdmurtRepublic for neutralization of an industrial waste. Izhevsk: IAM UB RAS, 2009.

Page 386: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

384

Chapter 52

The Effi ciency of Carbamates as Components of Prophylactic Antidotes Against OP Poisoning

Valerii TONKOPII, Anatoliy ZAGREBIN

Institute of Limnology, RussianAcademy of Sciences,Sevastyanova street 9, 196105 St.Petersburg, Russia

Abstract The usual treatment for organophosphate (OP) poisonings is a combination of atropine and some oximes. This is not effective against poisoning by soman, cyclosarin and tabun because of the rapid dealkylation of acetylcholinesterase (AChE). Previously, it has been shown that pretreatment with certain cholinesterase reversible inhibitors, such as physostigmine, gave appreciable protection against poisoning by any OP, including soman. In this paper a number of carbamates have been tested for their ability to protect mice, rats, cats and dogs against paraoxon, armine and soman poisoning. A series of pyridyl, pyridine and quinoline carbamates, with different chains between rings and with different substituents on the rings, were tested. In the experiments in vitro the anticholinesterase activity of carbamates and the infl uence of compounds on phosphorylation of AChE by OP were studied also. The present study has shown that the protective effect of carbamates depends primarily upon the ability of the carbamate to inhibit the brain AChE with the partial protection of the enzyme against following phosphorylation. Many factors are involved in determining the protective effi ciency of the carbamates, such as their abilities to cross the blood - brain barrier, the duration of anticholinesterase action, the inhibition of brain AChE reversibly, the ability of acetylcholine to increase the speed of gradual decarbamylation of the AChE. It has been found that some carbamates, with irreversible modes of action, increased the toxicity of OP. The effi ciency of carbamates also depends on animal species. In mice and rats carbamates gave a weak protection, whereas in cats and dogs the protective effect was signifi cantly more. A few of the most effective carbamates, with a longer duration of protective action than physostigmine, were studied in more detail as possible components of prophylactic antidotes in conjunction with atropine.

Keywords.Carbamates, organopohosphates, antidotes , treatment, effi ciency

Inroduction In the treatment of organophosphates (OP) poisonings, the combination of muscarinic

cholinoreceptors antagonists, such as atropine and some oximes (obidoxime, pralidoxime, HI-6 etc.), has been used [1]. The effi ciency of oximes is not, however, satisfactory in the case of soman, cyclosarin and tabun poisonings, on account of the rapid dealkylation (“aging”) of acetylcholinesterase (AChE). The resulting methylphosphonyl-AChE appears to be resistant against the nucleophilic attack of oximes [2]. It was found that pretreatment

Page 387: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

385

with certain carbamates, such as physostigmine and pyridostigmine, in conjunction with atropine and oxime, gave appreciable protection against poisonings by any OP, including soman [3,4]. Since 1946 when G.Koster showed the protective effect of physostigmine against DFP poisoning, the mechanism of the prophylactic action of carbamates has been connected only with an ability of compounds to inhibit the AChE with the protection of the part of enzyme against following phosphorylation. The gradual spontaneous decarbamylation of the AChE-carbamate complex, in parallel with the relatively rapid removal or destruction of the OP, may release suffi cient AChE to maintain life. However, it can not explain the strongly pronounced protective action of carbamates.

Materials and Methods In order to get some new information about the effi ciency and the mechanism of

protective action of carbamates, we studied the effi ciency of different drugs in vitro and in vivo experiments. The following carbamates were used: physostigmine, pyridostigmine, pyridyl carbamates (aminostigmine, compounds T-66), piperidile pyridyl carbamates (T-80, T-85), pyridine carbamate (T-83), pyrrolidile pyridyle carbamate (T-87), quinoline carbamate (T-89) with different chains between rings and with different substituens on the rings. In the fi rst part of the study, the kinetics of the inhibition of the purifi ed human erythrocytes AChE by different carbamates was studied. The inactivation of the enzyme by carbamates was measured with the help of bimolecular rate constant of interaction of the AChE with compounds. AChE activity was assayed by the method of Ellman et al. (5). In the second part of the study, the effect of carbamates on phosphorylation of AChE by some OP (armine, soman, paraoxon ) was studied, too. After 30 minutes of incubation of carbamates with AChE, we investigated the biomolecular rate constant of interaction of the carbamylated AChE with OP. In the third part of the study, the activity of rat’s brain AChE, after the i.m. injection of different carbamates was measured by method of Ellman. In a fourth part of the study, the toxicity of carbamates in experiments with albino mice following subcutaneous injection of carbamates was determined from the LD50 values. And fi nally, in a study of the effi ciency of carbamates in the experiments on mice, animals were injected i.m. with a carbamate, with or without of atropine. After the appropriate pretreatment interval (30-60 min) the organophosphates ( soman, paraoxon, or armine) was given sc. LD50 values, based on 24 -hr mortalities, were calculated by the method of moving averages. The results of the protection experiments are expressed as the protective coeffi cient - PC (i.e., the ratio of LD50 value in treated and in untreated animals). The effi ciency of carbamates in experiments in rats, rabbits, cats and dogs has also been investigated.

Results and discussionIt has been found that the carbamates in the in vitro experiments acted as irreversible

inhibitors of AChE - the degree of enzyme inhibition increases with longer incubation time. It was found that the carbamates do not change the bimolecular rate constant of interaction of the AChE enzyme with soman . Thus showing that in the presence of carbamates, the OP ( soman, armine, paraoxon) interacted only with the free active centres of the enzymes, the active centres not occupied by the carbamates. There was no correlation between the

Page 388: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

386

protective activity of the carbamates and either their chemical structure or toxicity and .anticholinesterase activity. The protective effects of carbamates are due to their abilities to migrate across the blood-brain barrier and cause a reversible inhibition in an essential amount of brain AChE. The quaternary compound pyridostigmine, with a weak blood-brain permeability, does not protect the animals against OP poisonings when carbamate is used without other antidotes ( atropine and oximes). Of the nine carbamates tested, only three (physostigmine, aminostigmine and compound T-66) were effective in protecting mice against soman, armine and paraoxon poisonings. The effi ciency of the carbamates is related to the duration of the anticholinesterase action. Aminostigmine (in comparison with physostigmine and compound T-66) caused longer inhibition of mice brain enzyme (Table 1).

Table 1. The inhibition (%) of mice brain AChE after effective carbamates were injected i.m. at 0.25 LD50

Time (min) Carbamates

Physostigmine Aminostigmine T-6630 82.0 + 2.4 72.0 + 2.3 71.2 + 2.160 58.1 +1.5 65.0 + 1.8 45.4 + 1.290 33.0 + 2.3 58.0 + 1.4 30.0 + 1.3 120 20.0 + 1.2 50.0 +1.2 20.0 + 0.9150 0 40.5 + 3.5 10.0 + 0.7180 0 30.5 + 3.5 0

The prophylactic action of aminostigmine was also more strongly pronounced in comparison with physostigmine and T-66. The duration of inhibition of mice brain AChE lasted more than 24 hours after injection of carbamates T-80, T-85, T-87, T-89 (Table 2).

Table 2. The inhibition (%) of mice brain AChE after noneffective carbamates were injected i.m. at 0.25 LD50

Time( hrs ) T - 80 T – 85 T- 87 T - 89

0.51.03.06.012.018.024.0

82.0 + 1.379.0 + 2.164.0 + 1.955.0 + 1.826.0 + 0.915.0 + 0.810.0 + 0.5

78.0 + 2.179.0 + 2.569.0 + 2.360.0 + 1.853.0 + 2.040.0 + 1.534.0 + 1.3

84.0 + 1.980.0 + 2.375.0 + 1.978.0 + 2.358.0 + 1.955.0 + 1.640.0 + 1.2

80.0 + 2.185.0 + 2.578.0 + 2.169.0 + 2.260.0 + 1.862.0 + 1.953.0 + 1.7

In contrast with physostigmine and aminostigmine, these carbamates with almost irreversible action on AChE inceased the toxicity of the all tested OP including soman (Table 3).

Page 389: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

387

Table 3. The effects of carbamates were injected i.m. at 0.25 LD50 as a drugs for prophylaxis of soman poisoning in experiments on the mice.

Carbamates LD50 of somanmg/kg

PC

Control 0.135±0.003 -

Physostigmine 0.20±0.006 1.48

T-66 0.22±0.004 1.62

Aminostigmine 0.30±0.007 2.22

T-80 0.10±0.005 0.74

T-85 0.094±0.003 0.69

T-87 0.087±0.002 0.62

T-89 0.088±0.001 0.65

It is well known that carbamates have a broad spectrum of toxicity - from relatively nontoxic to highly toxic compounds comparable with nerve agents]. Treatment of carbamate poisonings is resistant to the effects of oximes . Due to these reasons, in the experiments on mice, the prophylactic action of two reversible cholinesterase inhibitors (galanthamine and tacrine) against poisonings by irreversible carbamates T-80, T-87 and T-89 was studied. After 30 minutes of mice pretreatment by galanthamine and tacrine in doses of 0.25 LD50, the average lethal doses of irreversible carbamates (T-80, T-87, T-89) was measured. For the fi rst time the prophylactic effi ciency of reversible inhibitors as drugs for prophylaxis of poisonings by irreversible carbamates have been shown (Table 4).

Table 4. The effi ciency of tacrine and galathamine were injected i.m. at 0.25 LD50 for prophylaxis of carbamate poisoning in experiments on the mice

Carbamates Control LD50 mg/kg

GalanthamineEffi ciency

PC TacrineEffi ciency

PC

T-80 12.8±0.18 25.0±1.05 1.95 40.0±2.5 3.12T-87 34.0±1.6 105.0±5.9 3.08 120.0±4.1 3.53T89 90.0±2.1 172.0±8.3 1.91 270.0±5.2 3.0

The effi ciency of carbamates also depends on animal species. In mice, rats and rabbits carbamates provide a weak protection, whereas in cats and dogs the protection effect was signifi cantly greater. This may due not only with different elimination time of carbamates but also with the peculiarities of pathogenesis by OP intoxication on different animal species.

In the experiments on rats, cats and dogs, we found that in the presence of effective carbamates, the inhibition of AChE of the brain (caused by reversible inhibitors) quickly diminished following introduction of OP. This effect was connected with an ability of acetylcholine surplus to displace relatively rapidly the part of enzyme occupied by

Page 390: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

388

carbamates. This free part of the AChE can hydrolyze the acetylcholine surplus and prevent the intoxication. Thus the protective action of carbamates against OP poisonings no doubt depends primarily upon the ability of the carbamates to inhibit brain AChE, forming a semistable complex of reversible inhibitor - enzyme, which can spontaneously breakdown to liberate the enzyme. The mode of connection (reversible or irreversible) of carbamates with AChE and the sensitivity of the complex of carbamate-enzyme to acetylcholine is very important for the prophylactic action. The preference of the carbamate, aminostigmine, for prophylaxis against OP poisonings was shown.

REFERENCES:[1] .E. Heilbronn and B. Tolagen, Tooxogonin in sarin, soman and tabun poisoning. Biochem.Pharmacol.,

14 (1965) 73-77.[2]. O. Wolthuis and F. Berends, Problems in the therapy of soman poisoning. Fundam.Appl.Toxicol., 1

(1981) 183-192. [3.] R. Koster, Synergisms and antagonisms between physostigmine and DFP in cats. J.Pharmacol.exp.

Ther., 88 (1946) 39-46.[4.] W. Berry. and D. Davies, The use of carbamates and atropine in the protection of animals against

poisoning by 1,2,2 trimethylpropyl methylphosphonofl uoridate. Biochem. Pharmacol., 19 (1970) 927-934.

[5]. G. Ellman. et al., A new and rapid colorimetric determination of acetylcholiesterase activity.Biochem. Pharmacol., 7 (1961) 88-95.

Page 391: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

389

Chapter 53

A New Possibility for Separate Detection of Organophosphates and Carbamates

Valerii D. TONKOPII and Anatoliy ZAGREBINInstitute of Limnology, RussianAcademy of Sciences, Sevastyanov str., 9,19605 St.

Petersburg, Russia

Abstract It is well known that the cholinesterase of fi sh’s brain is the typical acetylcholinesterase (AChE) with the same substrate specifi city. On the other hand, the ChE of some fi sh’s blood plasma has its own specifi city. Some years ago for the fi rst time we discovered that only the blood serum of freshwater fi sh from family of Cyprinidae (blue bream - Abramis ballerus, roach - Rutilus rutilus) contains nonordinary ChE with unusually high sensitivity to organophosphates - dipterex and DDVP and low sensitivity to carbamate neostigmine. This observation is of scientifi c and practical interest and so the fi sh’s blood plasma ChE of these species was purifi ed for study of kinetic behaviour and sensitivity to antiChE compounds. The sensitivity of enzyme to 45 organophosphates (including sarin, soman and Vx) and carbamates has been determined. The Russian commercial purifi ed lyophilized cholinesterases have been used for comparison: AChE from the erythrocytes of human, butyrylcholinesterase (BuChE) from the horse blood serum and propionylcholinesterases (PrChE) from the hen blood serum and from squid optic ganglion. We investigated the activity of new fi sh’s purifi ed enzyme relatively to a choline and thiocholine esters hydrolysis as a function of substrate concentration. The results of this and the following experiments indicate that the new ChE from the blue bream and roach blood serum can be classifi ed as BuChE, so the velocity of butyrylcholine and butyrylthiocholine hydrolysis is more than other substrates. . The sensitivity of fi sh’s ChE to organophosphates is in 100-2000 times higher than the sensitivity of all types of commercial ChE. On the other hand a new enzyme has an extremely low sensitivity to carbamates. It is very important that with the help of a new purifi ed fi sh’s ChE the separate identifi cation of organophosphorus CW, pesticides and carbamates may be carried out. The extremely low sensitivity of new enzymes to carbamates and very high sensitivity to organophosphates is particularly valuable for these purposes.

Key words: organophosphates, carbamates, detection, cholinesterases.

IntroductionAmong many xenobiotics entering aqueous media, anticholinesterase (antiChE)

compounds notable for their high toxicity and selectivity are of particular hazard. Many compounds of these classes are used as pesticides, drugs, chemical warfare agents and many have thus become environmental contaminants. Currently, dozens of pesticides ( organophosphorus compounds and carbamates) capable of polluting the aqueous media

Page 392: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

390

through the runoff from agricultural lands or as a result of chemical industry accidents are produced. The forthcoming destruction of chemical weapons, a major part of which constitutes antiChE compounds ( sarin and soman) can also lead to water pollution. Although the stability of these substances in water is not high (e.g., when compared to organochlorine compounds), certain xenobiotics of these classes are highly toxic for humans, animals, and particularly for hydrobionts. At present, various types of purifi ed commercial cholinesterases (ChE) are widely used to detect antiChE compounds. The detection is based on the property of xenobiotics to lower the activity of enzymes. However, this method is not universal because ChE has low sensitivity to many organophosphorus (OP) and carbamate pesticides [1].

It is known that the cholinesterase of fi sh’s brain is the typical acetylcholinesterase (AChE) with the same substrate specifi city and sensitivity to organophosphorus compounds as AChE from human erythrocytes [2]. The fi sh’s blood serum cholinesterase also has been identifi ed as AChE [3]. On the other hand the blood serum of only two freshwater fi shes (blue bream - Abramis ballerus and roach - Rutilus rutilus) contains mainly butyrylcholinesterase (BuChE) with unusually high sensitivity to organophosphorus pesticides - dipterex and DDVP [4]. This is of both scientifi c and practical interest.

Material and methods In this work we studied the substrate specifi city, kinetic behaviour and sensitivity of

blue bream blood serum ChE for some OP and carbamate pesticides. The earlier unstudied blood serum of blue bream was used as a source of ChE. The fi sh were collected during autumn - winter period from the Volga pool of the Rybinsk Reservoir. The blood was taken away from fi sh tail vein and the fi brin clot was separated from the serum. After isolation and purifi cation of blood serum by help of well - known methods, a stabilized lyophilized enzyme with activity of 5 - 10 units per mg of protein was obtained. The following Russian commercial purifi ed lyophilized cholinesterases have been used for comparison: AChE (acetylcholine acetylhydrolase, EC 3.1.1.7) from the erythrocytes of human, BuChE (acylcholine acylhydrolase, EC 3.1.1.8) from the horse blood serum and propionylcholinesterases - PrChE (acylcholine acylhydrolase, EC 3.1.1.8) from the hen blood serum (PrChE-1) and from the squid (Todarodes pacifi cus) optic ganglion (PrChE-2). The kinetics of cholinesterase hydrolysis of different substrates was investigated at pH-7.5 and at 25oC. Acetylcholine iodide (ACh), propionylcholine iodide (PrCh), butyrylcholine iodide (BuCh), acetylthiocholine iodide (AThCh), propionylthiocholine iodide (PrThCh), butyrylthiocholine iodide (BuThCh) were used as substrates. The initial rate of enzyme hydrolysis was measured by the following methods. In the experiments with choline esters it was measured by the potentiometric titration [5] in 1mM of phosphate buffer and 100 mM of potassium chloride. In the experiments with thiocholine esters it was measured by the photometric method of Ellman [6] in the 20 mM of phosphate buffer containing 100 mM of potassium chloride and 0.2 mM5’,5’-dithio-bis-(2-nitrobenzoic acid). The maximal rates of enzyme reaction and Michaelis constants (Km) were obtained by the Lineweaver - Burk method [7] using a computerized production control system.

The irreversible inactivation of the enzyme by OP and carbamates was measured with the help of bimolecular rate constant (K2) of interaction of the enzymes with inhibitors at pH 7.5 and 25oC in the 20 mM of phosphate buffer, containing 100 mM of

Page 393: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

391

potassium chloride and 0.2 M5’,5’- dithio-bis - (2-nitrobenzoic acid). In experiments were used the following organophosphates and carbamates. 1. Organophosphates: dipterex, DFP (diisopropylfl uorophosphate), dichlorvos (DDVP), paraoxon, phosphamidon, dimethoate, mevinphos, sarin, soman. 2. Carbamates: physostigmine, aminostigmine (2-dimethylaminomethylene-3-dimethylcarbamoyloxypyridine), bizerine (N,N’-hexamethylene-bis-[ N-methylcarbamic acid 3 - ( 2 - dimethylaminomethyl) pyridile ester] tetrahydrochloride), sevine, bis-quaternary carbamate - X-129 (1-[ N- (3-[dimethylcarbamoyloxy] - α - picolile) -N,N- dimethylammonio] - 10 - [N- oxyethyl - N,N - dimethylammonio]decabromine).

ResultsThe curve of the dependence between the hydrolysis rates of substrates by fi sh ChE

(FChE) and the value of pH looks like a bell and has a maximum at pH 8.5. The curves for pH- dependence in the experiments with FChE differed only slightly from these for purifi ed cholinesterases - AChE and BuChE. We investigated the activity of fi sh ChE (FChE) relatively to a choline and thiocholine esters hydrolysis as a function of substrate concentration. The optimal substrates for FChE are BuCh and BuThCh; the activity of enzyme rises with an increase of substrates concentration. The results of this and the following experiments indicate that the purifi ed ChE of blue bream blood serum can be classifi ed as a BuChE, so the velocity of butyrylcholine and butyrylthiocholine hydrolysis is more than other substrates. At the same time, this type of FChE differs from typical BuChE, so the hydrolysis rate of butyrylcholine by FChE is in 10 - 13 times more rapid as compared to hydrolysis of acetylcholine (Table 1).

Table 1. The kinetic parameters (Km μM, and relative rate of substrates hydrolysis –V) of different cholinesterases (Rates of hydrolysis expressed as percentages of the acetylcholine rate).

EnzymesACh PrCh

SubstratesBuCh AThCh PrThCh BuThCh

VFChE 100 450 1140 500 800 1250

Km 1300 560 170 430 57 16V

AChE 100 70 5 89 19 5

Km 190 260 - 88 160 -V

BuChE 100 173 260 141 127 210

Km 910 550 770 540 510 430

On the other hand the hydrolysis rate of butyrylcholine by BuChE is only in 1.2 - 2.6 more rapid as compared to hydrolysis of acetylcholine. The comparison of Michaelis constants for different substrates confi rms the differences between the FChE, horse blood plasma BuChE and human erythrocytes AChE. The value of Km for FChE is 5 times less in the case of butyrylcholine and 30 times less for butyrylthiocholine as compared with ordinary BuChE. The differences between FChE and other types of ChE are especially

Page 394: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

392

appreciable during the study of different inhibitors of cholinesterase. The FChE has a very high sensitivity to some organophosphorus compounds (Table 2).

The sensitivity of FChE is 70 times higher for dipterex, that of DDVP - 1500 times, phosphamidon - 160 times, dimethoat - 90 times, mevinphos - 100 times, sarin - 50 times than the sensitivity of BuChE. It is very unexpectedly and unordinary that FChE has a very small sensitivity to active carbamates.The sensitivity of FChE for physostigmine is 200, aminostigmine - is 700, bizerine- is 250, bis-quaternary carbamate X-129 - is 15 000 times lower than the sensitivity of BuChE.

Table 2. The bimolecular rate constant (k2 M-1 min-1) of interaction

of the enzymes with organophosphates and carbamates.

Compounds Types of cholinesterasesAChE BuChE PrChE-1 PrChE-2 FChE

Dipterex 2.103 2.6 103 1.2 103 2 104 1.8 105

DDVP 1.1.104 2.1.104 2.3.104 2.5.105 3.1.107

Paraoxon 3 .106 1.3.106 2.106 7.106 3.4.107

DFP 1.1.104 4.2.106 4.5.105 5.106 8.107

Phosphamidon 5.103 5.103 5.104 8.104 8.105

Dimethoate 6.103 1.104 2.104 3.5.104 9.105

Mevinphos 5.104 5.103 2.5.103 4.5.104 5.5 .105

Sarin 1.2.107 4.2.106 4.8.107 1.9.108 2.5.108

Soman 7.8.107 1.5.107 3.107 3.108 9.107

Aminostigmine 5.106 7.7.105 1.105 3.5.105 1.103

Physostigmine 8.106 2.106 1.2.106 1.6.105 1.1.104

Bizerine 3.3.106 9.105 4.105 8.105 4.103

X-129 2.6.109 4.107 6.106 5.106 2.4.103

Sevine 2.5.104 2.5.103 1.103 1.103 4.102

The results obtained with study of kinetic behaviour of FChE and its sensitivity to OP and carbamates suggested that there might be a essential difference between the active sites of FChE and another types of cholinesterases. It is very important that with the help of a new purifi ed FChE the separate bioidentifi cation of OP (especially the organophosphorus pesticides) and carbamates may be carried out. The extremely low sensitivity of new purifi ed enzyme to carbamates is particularly valuable for these purposes.

References

[1] Silver. Ann. The biology of cholinesterases. Elsevier publish Company, 1974, 236.[2] Augustinsson. K.-B. The evolution of esterases in vertebrates Homologous Enzymes and Biochemical

Evolution, Gordon and Breach, New York, 1968, 299 - 311.[3] Augustinsson, K.-B. Electrophoresis studies of blood plasma esterases. Acta Chem. Scand. 13, 2, 1959,

571 – 592.[4] Pavlov. D., Feeding behavior and brain acetylcholinesterase activity in bream (Abramisbrama) as affected by

DDVP. Comp. Biochem. Physiol., 103C, 3, 1992,563-568.[5] Kozlovskaya,V. et al. Cholinesterases of invertebrates. Rev. Environ. Contam. Toxicol. 132, 1993,117 - 142.[6] Ellman, G. et al. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem.

Pharmacol. 7, 2, 1961, 88 - 95.[7] Lineweaver, H., Burk, D. J. The determination of enzyme dissociation constants.J.Amer. Chem. Soc. 55,

6,1974, 658 - 666.

Page 395: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

393

Chapter 54

Biomarkers of Organophosphorus Compounds Poisoning and Exposure: a Review

Kambiz SOLTANINEJADLegal Medicine Research Center, Legal Medicine Organization, Tehran, Iran

Abstract: Organophosphorus compounds (OPs) are organic derivatives of phosphorus and used mainly as pesticides and chemical warfare agents. OPs considered as a toxicant to human and animals. One of the most important mechanisms of action of OPs is the inhibition of the acetylcholinesterase that leading to the accumulation of acetylcholine at cholinergic synapses. OPs poisoning is a worldwide health problem, especially in developing countries and affecting millions of people with a high fatality rate. From this view, predicting of OPs intoxicated cases outcome has a critical role for preparing of therapeutic measures. Biomarkers are one of the mainstays in the diagnosis and prognosis of poisoning. There are some of biomarkers for diagnosis of exposure and intoxication of OPs. But the usefulness of these biomarkers for diagnosis, prognosis and predicting of severity of OPs poisoning has been debated. A major challenge in OPs toxicology has been the identifi cation and utilization of reliable biomarkers for the rapid, sensitive, and effective detection of OPs exposure and toxicity. In this article, the main biomarkers which have been evaluated for diagnoses and prognosis of OPs poisoning are reviewed.

Key words: Organophosphorus, Intoxication, Biomarker, Exposure

1. Introduction Organophosphorus (OPs) compounds are esters, amides or thiolic derivatives of

phosphoric, phosphonic or phosphinic acids. They are organic substances which contain a phosphoryl or a thiophosphoryl bond [1]. OPs are used mainly as pesticides in agricultural and domestic settings worldwide [2, 3]. They are used as solvents, plasticizers, fi re retardants, drugs, additives to hydraulic fl uid and jet engine oil [2,3].OPs are used as chemical warfare agents in battlefi elds and terrorist attacks and they so-called nerve gases or nerve agents [1,4,5].

OPs considered as a toxicant to human and animals. One of the most important mechanisms of action of OPs is the inhibition of the acetylcholinesterase (AChE) that leading to the accumulation of acetylcholine at cholinergic synapses and overstimulation of cholinergic synapses in the parasympathetic system, neuromuscular junction and central nervous system [1-8]. Butyrylcholinesterase (BChE) is another esterase which inhibit by OPs, but in contrast, appears not to result in clinical effects [3]. Clinical signs and symptoms of OPs acute intoxication including muscarinic effects (e.g. nausea, vomiting,

Page 396: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

394

diarrhea, abdominal cramps, urinary incontinence, miosis, bradycardia, bronchorrhoea, bronchoconstriction, salivation, lacrimation, emesis, hypotension and cardiac arrhythmias), nicotinic effects (e.g. tremors, fasciculations, muscle weakness, respiratory failure, hypertension, tachycardia, sweating, mydriasis) and central nervous system presentations (e.g. altered level of consciousness, respiratory failure and seizures)[4,7,8]. Intermediate neurotoxic syndrome is a clinical feature of OPs poisoning that characterized by cranial nerve palsies, weakness of the neck and proximal limbs, and respiratory paralysis [8, 9]. Organophosphate-induced delayed neuropathy (OPIDN) is another clinical manifestation in poisoning induced by some types of OPs. OPIND appears 1-3 weeks after an acute exposure to neuropathic OPs, which characterized by muscle weakness, ataxia and paralysis of the limbs [3, 10, 11]. Inhibition of neuropathy target esterase ( NTE) which is an integral membrane protein found in central and peripheral nerve tissue by OPs is correlated with the ability of these OPs to induce OPIIDN [11].

OPs poisoning is a worldwide health problem, especially in developing countries due to their availability and affecting millions of people with a high fatality rate [12, 13]. Suicidal poisoning is the major type of acute OPs poisonings in developing countries [14]. In contrast, occupational and environmental exposures are a common source for OPs intoxications in industrial and developed countries [15, 16]. OPs acute poisoning considered as a cause of hundreds of thousands of death annually, worldwide [17]. From this view, predicting of OPs acute intoxicated cases outcome has a critical role for preparing of therapeutic measures [18]. Biochemical markers or biomarkers are one of the mainstays in the diagnosis and prognosis of disease and poisoning [19]. There are some of biomarkers for diagnosis of exposure and intoxication of OPs [19-26]. But the usefulness of these biomarkers for diagnosis, prognosis and predicting of severity of OPs poisoning and patient outcome has been debated. As there are limitations in each of biomarkers for diagnosis and prognosis of OPs poisoning, investigations for fi nding of new and reliable biomarkers in OPs intoxication have been welcome.

In this article, the main biomarkers which have been evaluated in diagnoses and prognosis of OPs poisoning are discussed.

2. OPs biomarkers

2.1. Enzymes as OPs biomarkers

2.1.1. Acetylcholinesterase and butyrylcholinesterase Acetylcholinesterase (AChE, E.C. 3.1.1.7) and butyrylcholinesterase (BChE, E.C.

3.1.1.8) are the enzymes which members of the cholinesterases group [1, 27]. AChE hydrolyses acetylcholine (ACh) and terminates the action of it at the post-

synaptic membrane in the neuromuscular junction and neuronal synapses [27]. ACh is released from the presynaptic nerve in response to an action potential, and diffuses across the synapse, binds to the ACh receptor. After binding to the post synaptic cholinergic receptor, it initiates several events that result in the beginning of the action potential in the cell. AChE acts mainly at the cholinergic receptors located in the central and peripheral nervous systems. The role of AChE in some tissues such as the red cell membrane is not clear [1, 27].

Page 397: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

395

BChE also known as plasma cholinesterase, nonspecifi c cholinesterase and pseudocholinesterase, hydrolyses butyrylcholine and is synthesized by the liver, and found in the plasma. The physiological function of BChE is not fully understood. It appears not to be essential for cholinergic transmission, but there are some indications of its participation in the initial stages of the nervous system development. BChE as AChE is found in neurons and glial cells of the human brain [1, 27].

AChE and BChE are the prototype of serine hydrolases family and exist as water-soluble forms. AChE and BChE hydrolysis the different types of cholines. AChE hydrolyses ACh quickly and propionylcholine with slower rate. It shows a loss of hydrolytic activity for butyrylcholine. In contrast, BChE hydrolyzes butyrylcholine and benzoylcholine [1, 26, 27].

OPs inhibit AChE and BChE enzymes [4,8]. However, AChE is a main target enzyme for OPs induced poisoning and it inhibition causes clinical effects [4,8]. In contrast, BChE inhibition appears not to result in clinical features [3, 28].

Determination of AChE and BChE activity in OPs intoxicated cases biosamples considered as biomarkers during OPs poisoning. BChE activity is more easily measured than AChE activity, and BChE assays are widely available and therefore commonly recommended in the early assessment of OPs intoxicated patients [18]. A severity scoring system according to the measurement of plasma BChE activity has been developed [29]. The evaluated BChE 20–50% of normal activity had in mild poisoning, moderate poisoning 10–20% and severe poisoning <10% of normal BChE activity [29]. The usefulness of BChE as biomarker for diagnosis and prognosis of OPs poisoned patients has been much debated. There are some physiological and pathophysiological statuses that could be changed the BChE plasma levels among patients. For example, the BChE concentration in human plasma is correlated with growth hormone levels, obesity, pregnancy and parturition [27]. Low level of plasma cholinesterase was found to be caused by pulmonary Koch’s and hepatitis B with associated malnutrition [19].

Some of the studies showed BChE to be a poor predictor of outcome and response to therapy [19, 30-32]. Despite of these studies, use of BChE activity as a biomarker in the early assessment of OPs poisoning severity has been used [24,33-38].It is possible that BChE activity may be useful for particular OPs, if they can be confi dently identifi ed on admission. Eddleston et al (2008) showed that plasma BChE activity on admission can provide useful biomarker for predicting fatality in patients to have poisoned with dimethoate or chlorpyrifos. BChE activity can only be used to predict death when the insecticide ingested is known and its sensitivity and specifi city for that insecticide has been studied [18].

In a retrospective study was conducted on 47 children with organophosphate and carbamate poisoning showed that low BChE levels support the diagnosis of OPs poisoning, but no signifi cant association is present between the severity of poisoning and BChE levels [20].

Generally, measurement of plasma BChE activity is a suitable biomarker of OPs exposure and poisoning diagnosis but it is not reliable biomarker for determining of OPs poisoning severity and patient outcome. The interpretation of BChE activity, sometimes presents signifi cant complexities [39].

Inhibition of AChE is regarded as the primary toxic mechanism of OPs, AChE activity, from this view, measurement of AChE activity could be considered a suitable biomarker

Page 398: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

396

for OPs poisoning severity [4,7,8]. In practice, the erythrocyte AChE activity is used to mirror effects on the nervous system and can be measured in clinical setting [8, 40]. The physiological function of erythrocyte AChE is not known, but the enzyme is the same as that involved in synaptic transmission. However, erythrocyte AChE has large inter- and intraindividual variation, and small changes are detectable by comparison with pre- exposure values. The relation between inhibition of erythrocytes and nervous tissue AChE depends on the pharmacokinetics of OPs. Usually, erythrocyte AChE inhibition overestimates that in the nervous system. Pharmacodynamic factors such as spontaneous reactivation and aging of inhibited enzyme should also be considered in assessing AChE inhibition. Other factors, such as timing of measurement, add complexity because erythrocyte AChE inhibition persists longer than that in the nervous tissues [40].

Brahmi et al (2006) showed that the marked decrease of erythrocyte AChE activity appeared as prognostic factor in acute OPs, and coma, respiratory failure, hemodynamic disturbances, and death are associated with a decrease of the erythrocyte AChE of less than 23.5 μmol/mL per hour at 37 °C [41].

In clinical setting, serial erythrocyte AChE activity measurements in OPs poisoned patients could be performed for evaluation of effi cacy of antidotal therapy and individual response during and after poisoning [42].

2.1.2. Beta-glucuronidaseBeta-glucuronidase (beta-G) is an enzyme which has been used as a biomarker of OPs

poisoning [43-49]. Beta-G is a liver microsomal enzyme that is stabilized within microsomal vesicles by complexation with egasyn [43-45]. Egasyn is an accessory glycoprotein of beta-glucuronidase in the liver microsomes and one of the carboxylesterase isozymes [43-46]. In human, it does not appear to use the esterase active site for binding to Beta-G [48].

In vivo and in vitro studies showed that when OPs are incorporated into the liver microsomes, they become tightly bound to egasyn, and subsequently, beta-G is dissociated and released into blood. Consequently, the increase in plasma beta-G activity becomes a good biomarker of OP exposure. Previous studies showed that the some of OPs such as EG (O-ethyl O-p-nitrophenylphenylphosphonothioate), acephate, fenitrothion, phenthionate, bis-beta-nitrophenyl phosphate (BNPP) and chlorpyrifos increased plasma BG activity in approximately 100-fold the control level in rats. The increase in plasma BG activity after OP exposure is a much more sensitive biomarker of acute OP exposure than AChE inhibition. In experimental studies, similarly, carbamate insecticides such as carbaryl caused rapid increase of plasma beta-G level. In contrast, no signifi cant increase of plasma beta-G level was observed when pyrethroid insecticides were administered to rats [43-47].

A study on 21 pest control operators and their co-workers who had not sprayed OPs (control group) within 3 days prior to sample collection, showed that plasma BG activity is more sensitive biomarker as well as urinary OP metabolites than BChE for low-level exposure in humans [49].

Soltaninejad et al (2007) showed that on total of 21 acute OPs poisoned patients were divided into two groups of with mild and sever poisoning according to clinical presentations,a signifi cant increase in serum beta-G was observed only in severely affected OPs patients as compared to controls. The results showed blood beta-G is very sensitive biomarker in severe OPs poisoning [25].

Page 399: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

397

However, recently some controversy has been observed to use beta-G as an OPs poisoning biomarker. For example, Sabbe et al (2008) in a study observed a decrease in plasma concentration of beta-G, even within normal range. These fi ndings do not support the hypothesis that beta-glucuronidase is a useful biomarker for acute OPs poisoning in humans [50].

2.1.3. Paraoxonase Paraoxonase 1 ( PON1) is a high-density lipoprotein (HDL)-associated serum enzyme

that hydrolyzes a number of OPs, and its role in modulating the toxicity of OPs [51]. The physiological function of PON1 has not yet been identifi ed. PON1 appears to be primarily a lactonase and also displays an anti-atherogenic activity closely linked to its localization on HDL particles [51]. Human serum paraoxonase (EC 3.1.8.1) is a Ca2+-dependent enzyme that hydrolyzes esters including OPs insecticides, nerve agents, oxidized lipids and a number of drugs or pro-drugs [52]. PON1 hydrolyses OPs entering the blood circulation and tissue fl uid thus limiting toxicity [53,54].

Several polymorphisms in the PON1 gene have been described, which have been shown to affect either the catalytic effi ciency of hydrolysis or the expression level of PON1. In humans, a substrate-dependent polymorphism of serum paraoxonase is observed, where one isoform of paraoxonase has a high turnover number for paraoxon and the other a low turnover number [54]. Both isoforms appear to hydrolyze chlorpyrifos-oxon and phenylacetate at the same rate. The PON1192Q/R polymorphism (that affects catalytic ability toward different substrates) and PON1 levels (which are modulated in part by a C-108T polymorphism) over straight genotyping [51,53]. The PON1 coding region has two polymorphisms involving the amino acids at position 55 and 192, giving rise to isoenzymes which differ in their catalytic rate for the hydrolysis of OPs [55]. Cloning and sequencing of the human paraoxonase cDNAs has elucidated the molecular basis of the polymorphism. Arginine at position 192 determines high paraoxonase activity, and glutamine at this position, low paraoxonase activity [55]. In addition to the polymorphism, a 13-fold variation in serum enzyme levels within a given genetic class is seen. Therefore, that hypothesized individuals inheriting low activity isoforms of PON1 would be more liable to report symptoms of OPs toxicity [54]. In another study observed the farmers reporting chronic poisoning due to OPs exposure has a higher proportion of the PON1-192R polymorphism associated with lower rates of diazoxon hydrolysis and lower rates of diazoxon hydrolysis than the controls and that their ill health may be explained by a lower ability to detoxify diazoxon [54].

A positive correlation was found between AChE activity and PON1 stimulation in the workers who has been exposed to OPs, chronically [56]. Recent study examining the involvement of PON1 status in determining OPs susceptibility of poisoned patients with nerve agents represent a step in the right direction, but more studies are needed [52].

Determination of PON1 status in farm worker mothers and their babies predicted a range of sensitivity to diazoxon of 65-fold from the baby with the lowest PON1 status to the mother with the highest PON1 status [56].

These studies suggest that PON1 status may act as a biomarker for OPs toxicity in humans.

Page 400: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

398

2.1.4. Antioxidant enzymes and oxidative stress parametersThere are some studies that demonstrated oxidative stress during OPs acute, subchronic

and chronic poisoning [57-62]. Malathion induced oxidative stress and ChE depression in saliva and plasma in rats following subchronic exposure (at doses of 100, 500 and 1500 ppm for 4 weeks, orally). The malondialdehyde (MDA) concentration, the end product of lipid peroxidation, was measured. Malathion increased plasma thiobarbituric acid reactive substances (TBARS) of control. The results showed that in OPs subchronic exposure, depression of ChE is accompanied by induction of oxidative stress [61].

In another study, malathion induced oxidative damage in different tissues of wistar rats, administered intra peritoneally at doses of 25, 50, 100 and 150mg malathion/kg, after acute and sub-chronic exposure. TBARS and protein oxidation by levels of carbonyl groups, and also on the activities of superoxide dismutase and catalase, two antioxidant enzymes that detoxify superoxide radical and hydrogen peroxide, respectively. The results showed that the most sensitive targets of oxidative damage were kidney, lung and diaphragm after acute treatment, and liver, quadriceps and serum after sub-chronic treatment. Also, in general, increased lipid peroxidation seems to be a better biomarker of oxidative stress compared to the contents of protein carbonyls after acute and sub-chronic malathion exposure [57].

Oxidative stress status and AChE activity were studied in blood samples OPs formulating pesticide workers. The results showed marked inhibition of AChE activity and increased TBARS, decreased ferric-reducing ability of plasma (FRAP) and decreased total thiol group levels in workers. The reduction in activity of AChE correlated well with increased TBARS and decreased FRAPS in OPs formulators [58].

Oxidative stress in workers who formulate OPs, synthetic pyrethroid and carbamate pesticides has been studied. In this survey, blood erythrocytes from a group of 94 pesticide-formulating workers (at least 5-years experience).Lipid peroxidation level, catalase, superoxide dismutase (SOD) and glutathione peroxidase activities in erythrocytes were analysed as biomarkers of oxidative stress. In addition, the AChE activity was measured as a biomarker of toxicity. Results indicated that chronic exposure to OPse, synthetic pyrethroid and carbamate pesticides were associated with increased activities of catalase, SOD and lipid peroxidation in erythrocytes (p < 0.05). AChE activity did not show any signifi cant differences between the two groups (p > 0.05). It is concluded that human chronic exposure to pesticides may result in stimulated antioxidant enzymes [59].

OPs induced genotoxicity has been done by oxidative stress in workers who formulate OPs pesticides. The results indicated that chronic exposure to OPs pesticides was associated with increased activities of catalase, SOD and glutathione peroxidase in erythrocytes. The level of lipid peroxidation and AChE activity did not show any signifi cant differences between the two groups. It is concluded that human chronic exposure to OP pesticides may result in stimulated antioxidant enzymes and increased DNA damage in the absence of depressed Ach [61]. In a study, has been demonstrated that xanthine oxidase (XO) plays an important role in the mechanism of toxicity of acute OPs pesticide poisoning. The results showed that serum XO and MDA activities were higher and the serum SOD, PON1 and BChE activities were lower in the poisoned patients compared with the controls. A signifi cant negative correlation between XO activity and the SOD, PON1 and BChE activities, but a signifi cant positive correlation between XO activity and MDA has

Page 401: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

399

been demonestrated. These results suggest that increased activity of XO and decreased antioxidant enzyme activity contribute to the development of oxidative injury in OPs acute poisoning cases [62].

2.2. Dialkyl phosphates (DAPs)DAPs are metabolic and environmental breakdown products of OPs. DAPs such as

dimethyl phosphate (DMP), diethyl phosphate (DEP), diethyl thiophosphate (DETP) and diethyl dithiophosphate (DEDTP) are used for a biomarker of OPs exposure. DAPs have high oral bioavailability and are ubiquitously present in produce at concentrations several-fold greater than parent OPs [22, 63, 64]. Recently, urinary DAPs have considered as a biomarker assessing exposure to OPs in epidemiological investigations. The in vivo metabolism of OPs yields different DAPs, depending upon whether they undergo bioactivation or detoxifi cation. The detection of urinary DAPs does not provide specifi city with respect to the OPs from which they were derived, or their toxicological potency. Several recent studies documented the common presence of DAPs in residential environments and foods [64]. Experimental studies support that DAPs have signifi cant oral bioavailability, and undergo little to no metabolism prior to urinary excretion. There are not a dose-response relationship between DAPs and ChE inhibition and evidences of a threshold level of DAPs excretion at which clinical cholinergic signs or symptoms have been observed. No conducted analyses are that distinguish different DAPs that refl ect bioactivation versus detoxifi cation pathways [22].

The limitations of the use of DAPs as biomarkers of OPs exposure including a lack of specifi city with respect to the OPs source and toxicologically irrelevant DAPs are commonly encountered in food and the environment. Substantial intra- and inter-day variability has been demonstrated for DAPs excretion in humans. A relationship between DAPs as biomarkers of exposure and ChE activity has not been established [22].

In general, DAPs have very limited utility in clinical toxicology or in the risk assessment process for OPs [22].

2.3. OP- Albumin adducts as a new biomarker of OPs exposureAlbumin is a non-glycosylated single chain protein consists of 585 amino acids with

17 intra-chain disulfi de bonds. Albumin is the most prominent protein in plasma that is synthesized in the liver and discharged into the blood [65]. Albumin is responsible for the plasma colloid osmotic pressure and for maintenance of blood volume and buffering action of plasma proteins. Albumin binds and transports metabolites and drugs [66]. Binding of OPs to albumin could act as OPs scavenger and therefore reduce the amount of OPs available for reaction with AChE [65]. The affi nity of different OPs for albumin is depended to the identity of the OPs [65].

Using of OP-Albumin adduct as an sensitive exposure biomarker has been demonstrated for nerve agents. The OPs nerve agents sarin, soman, cyclosarin and tabun phosphylate a tyrosine residue on albumin in human blood. These adducts may offer relatively long-lived biomarkers of nerve agent exposure that do not ‘age’ rapidly, and which are not degraded by therapy with oximes. Sensitive methods for the detection of these adducts have been developed using liquid chromatography-tandem mass spectrometry. Adducts of all four

Page 402: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

400

nerve agents were detected in the blood of exposed guinea pigs being used in studies to improve medical countermeasures. The tyrosine adducts with soman and tabun were detected in guinea pigs receiving therapy 7 days following subcutaneous administration of fi ve times the LD50 dose of the respective nerve agent. VX also forms a tyrosine adduct in human blood in vitro but only at high concentrations [66].

Although Tyr 411- OP adducts to human serum albumin (HSA) have been suggested to be one of the most robust biomarkers in the detection of OPs exposure, the analysis of HSA-OP adduct detection has been limited to techniques using mass spectrometry. A method described the procurement of two monoclonal antibodies (mAb-HSA-GD and mAb-HSA-VX) that recognized the HAS Tyr 411 adduct of soman (GD) or S-[2-(diisopropylamino)ethyl]-O-ethyl methylphosphonothioate (VX), respectively. The mAb-HSA-GD was able to detect the HSA Tyr 411 OP adduct at a low level that could not be detected by mass spectrometry. mAb-HSA-GD and mAb-HSA-VX showed an extremely low-level detection of GD and VX adducted to HSA (as picograms). The ability of the two antibodies to selectively recognize nerve agents adducted to serum albumin suggests that these antibodies could be used to identify biomarkers of OPs [21].

3. ConclusionA major challenge in OPs toxicology has been the identifi cation and utilization of

reliable biomarkers for the rapid, sensitive, and effective detection of OPs exposure and poisoning. Although, AChE and BChE have been utilized as routine biomarkers of OPs exposure and toxicity in clinical setting, but there are limitations for application and interpretation of these biomarkers. Therefore, identifi cation and evaluation of sensitive, rapid and reliable biomarkers in this fi led has been noted. From this view, determination of OPs-albumin adducts in blood considered as a new and sensitive OPs exposure biomarkers especially for detection of nerve agents in intoxicated patients. In clinical toxicology, common utilization of antioxidant enzymes and oxidative stress biochemical markers and bet-G as OPs toxicity biomarkers need to more and further investigations.

References:

1. Delfi no RT, Ribeiro TS, Figueroa-Villar JD. Organophosphorus Compounds as Chemical Warfare Agents: A Review. J Braz Chem Soc, 2008; 20(3):407-428.

2. Newmark J. Therapy for nerve agent poisoning. Arch Neurol, 2004; 61:649-652.3. Aardema H, Meertens JHJM, Ligtenberg JJM, Peters-Polman OM, Tulleken JE, Zijlstra JG.

Organophosphorus pesticide poisoning: cases and developments. Netherlands J Med, 2008; 66(4): 149-153.

4. Bey T, Sullivan J, Walter F. Organophosphate and carbamate insecticides. In: Sullivan J, Gray K (eds.) Clinical environmental health and toxic exposures, 2nd edn. Lippincott Williams & Wilkins, Philadelphia, 2001; p 1046-1057.

5. Balali-Mood M, Balali-Mood K. Neurotoxic disorders of organophosphorous compounds and their managements. Arch Iranian Med, 2008; 11(1): 65-89.

6. Roberts DM, Aaron CK. Managing acute organophosphorus pesticide poisoning. BMJ , 2007; 334:629-34.

7. Gupta RC. Toxicology of Organophosphate and Carbamate Compounds. Gupta R. C., ed., 1st ed., Elsevier Academic Press: London, 2006, pp. 5-24.

Page 403: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

401

8. Carlton F, Simpson W, Haddad L. The organophosphates and other insecticides. In: Haddad L, Shannon M, Winchester J (eds) Clinical management of poisoning and drug overdose, 3rd edn. W.B. Saunders, Philadelphia, 1998; p 836-845.

9. He F, Xu H, Qin F, Xu L, Huang J, He X. Intermediate myasthenia syndrome following acute organophosphates poisoning--an analysis of 21 cases. Hum Exp Toxicol , 1998; 17:40-5.

10. Nand N, Aggarwal HK, Bharti K, Chakrabarti D. Organophosphate induced delayed neuropathy. J Assoc Physicians India, 2007; 55:72-3.

11. Thomas TC, Ishikawa Y, McNamee MG, Wilson BW. Correlation of neuropathy target esterase activity with specifi c tritiated di-isopropyl phosphorofl uoridate-labelled proteins. Biochem J, 1989; 257, 109-116.

Jeyaratnam J. Acute pesticide poisoning: a major global health problem. World Health Stat Q, 1990; 43:139–44.

12. Van der Hoek W, Konradsen F, Athukorala K, Wanigadewa T. Pesticide poisoning: a major health problem in Sri Lanka. Soc Sci Med, 1998; 46:495–504.

13. Eddleston M. Patterns and problems of deliberate self-poisoning in the developing world. Q J Med, 2000; 93:715–31.

14. Muldoon SR, Hodgson MJ. Risk factors for nonoccupational organophosphate pesticide poisoning. J Occup Med, 1992; 34:38–41.

15. Jaga K, Dharmani C. Sources of exposure to and public health implications of organophosphate pesticides. Pan Am J Public Health , 2003: 14(3): 171-185.

16. Eddleston M. Patterns and problems of deliberate self-poisoning in the developing world. Q J Med, 2000; 93:715–31.

17. Eddleston M, Eyer P, Worek F, Rezvi Sheriff MH, Buckley NA. Predicting outcome using butyrylcholinesterase activity in organophosphorus pesticide self-poisoning. Q J Med , 2008; 101:467–474.

18. Mahadeshwara P, Gouda HS, Hallikeri VR. Plasma cholinesterase: double-edged parameter in the diagnosis of acute organophosphorus poisoning. Med Sci Law, 2010; 50(3):159-60.

19. El-Naggar Ael-R, Abdalla MS, El-Sebaey AS, Badawy SM. Clinical fi ndings and cholinesterase levels in children of organophosphates and carbamates poisoning. Eur J Pediat. 2009; 168(8):951-956.

20. Chen S, Zhang J, Lumley L, Cashman JR. Immunodetection of serum albumin adducts as biomarkers for organophosphorus exposure. J Pharmacol Exp Ther, 2013;344(2):531-541.

21. Sudakin DL, Stone DL. Dialkyl phosphates as biomarkers of organophosphates: the current divide between epidemiology and clinical toxicology. Clin Toxicol (Phila), 2011; 49(9):771-781.

22. Zhang JW, Lv GC, Zhao Y. The signifi cance of the measurement of serum xanthine oxidase and oxidation markers in patients with acute organophosphorus pesticide poisoning. J Int Med Re, 2010;38(2):458-65.

23. Aurbek N, Thiermann H, Eyer F, Eyer P, Worek F. Suitability of human butyrylcholinesterase as therapeutic marker and pseudo catalytic scavenger in organophosphate poisoning: a kinetic analysis. Toxicology, 2009; 259(3):133-139.

24. Soltaninejad K, Shadnia S, Afkhami-Taghipour M, Saljooghi R, Mohammadirad A, Abdollahi M. Blood beta-glucuronidase as a suitable biomarker at acute exposure of severe organophosphorus poisoning in human. Hum Exp Toxicol, 2007; 26(12):963-966.

25. Nigg HN, Knaak JB. Blood cholinesterases as human biomarkers of organophosphorus pesticide exposure. Rev Environ Contam Toxicol, 2000; 163: 29-111.

26. Chatonnet A, Lockridge O.Comparison of butyrylcholinesterase and acetylcholinesterase.Biochem J, 1989: 260, 625-634.

27. Lotti M. Clinical toxicology of anticholinesterase agents in humans. In: Handbook of Pesticide Toxicology. Volume 2. Agents. 2nd edn. San Diego, Academic Press, 2001: 1043–85.

28. Namba T, Nolte C, Jackrel J, Grob D. Poisoning due to organophosphate insecticides. Am J Med, 1971; 50:475–492.

29. Bardin PG, van Eeden SF, Moolman JA, Foden AP, Joubert JR. Organophosphate and carbamate poisoning. Arch Intern Med, 1994; 154:1433–1441.

Lotti M. Cholinesterase inhibition: complexities in interpretation. Clin Chem, 1995; 41:1814–1818.30. Worek F, Diepold C, Eyer P. Dimethylphosphoryl-inhibited human cholinesterases: inhibition, reactivation,

and aging kinetics. Arch Toxicol , 1999; 73:7–14.

Page 404: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

402

31. Kwong TC. Organophosphate pesticides: biochemistry and clinical toxicology. Ther Drug Monit, 2002; 24:144–149.

32. Abdullat IM, Battah AH, Hadidi KA. The use of serial measurement of plasma cholinesterase in the management of acute poisoning with organophosphates and carbamates. Forensic Sci Int, 2006; 162:126–130.

33. Khan S, Hemalatha R, Jeyaseelan L, Oommen A, Zachariah A. Neuroparalysis and oxime effi cacy in organophosphate poisoning: a study of butyrylcholinesterase. Hum Exp Toxicol, 2001; 20:169–174.

34. Lee P, Tai DYH. Clinical features of patients with organophosphate poisoning requiring intensive care. Intensive Care Med, 2001; 27:694–699.

35. Chugh SN, Aggarwal N, Dabla S, Chhabra B. Comparative evaluation of atropine alone and atropine with pralidoxime (PAM) in the management of organophosphorus poisoning. J Indian Acad Clin Med, 2005; 6:33–37.

36. Shivakumar S, Raghavan K, Ishaq RM, Geetha S. Organophosphorus poisoning: a study on the effectiveness of therapy with oximes. J Assoc Physicians India, 2006; 54:250–251.

37. Stefanidou M, Athanaselis S, Spiliopoulou H. Butyrylcholinesterase: biomarker for exposure to organophosphorus insecticides. Intern Med J, 2009; 39(1):57-60.

38. Lotti M. Cholinesterase inhibition: complexities in interpretation. Clin Chem, 1995; 41(120: 1814-1818.39. Brahmi N., mailto:[email protected] A., Kouraichi N., Ghorbel H., Blel Y., Thabet

H., Hedhili A, Amamou M. Prognostic value of human erythrocyte acetyl cholinesterase in acute organophosphate poisoning. Am J Eme Med, 2006; 24(7): 822–827.

40. Thiermann H., Kehe K., Steinritz D., Mikler J., Hill I., Zilker T., Eyer P., Worek F. Red blood cell acetylcholinesterase and plasma butyrylcholinesterase status: important indicators for the treatment of patients poisoned by organophosphorus compounds. Arh Hig Rada Toksikol , 2007; 58:359-366.

41. Fujikawa Y, Satoh T, Suganuma A, Suzuki S, Niikura Y, Yui S, Yamaura Y. Extremely sensitive biomarker of acute organophosphorus insecticide exposure. Hum Exp Toxico,. 2005; 24(6):333-336.

42. Satoh T, Suzuki S, Kawai N, Nakamura T, Hosokawa M. Toxicological signifi cance in the cleavage of esterase-beta-glucuronidase complex in liver microsomes by organophosphorus compounds. Chem Biol Interact 1999;119-120:471-478.

43. Satoh T. Release of liver microsomal beta-glucuronidase from hepatocytes in vitro and in vivo by organophosphates and hepatotoxic agents. J Toxicol Sci, 1991; Suppl 1:133-1342.

44. Medda S, Stevens AM, Swank RT. Involvement of the esterase active site of egasyn in compartmentalization of beta-glucuronidase within the endoplasmic reticulum. Cell, 1987; 50(2):301-310.

45. Medda S, Swank RT. Egasyn, a protein which determines the subcellular distribution of beta-glucuronidase, has esterase activity.J Biol Chem, 1985; 260(29):15802-15808.

46. Islam MR, Waheed A, Shah GN, Tomatsu S, Sly WS. Human egasyn binds beta-glucuronidase but neither the esterase active site of egasyn nor the C terminus of beta-glucuronidase is involved in their interaction. Arch Biochem Biophy,. 1999; 372(1):53-61.

47. Ueyam J., Satohhttp://www.sciencedirect.com/science/article/pii/S037842740901546X - aff2 T, Kondo T., Takagihttp://www.sciencedirect.com/science/article/pii/S037842740901546X - aff1 K., Shibata E., Goto M., Kimata A., Saitohttp://www.sciencedirect.com/science/article/pii/S037842740901546X - aff5 I., Hasegawa T., Wakusawahttp://www.sciencedirect.com/science/article/pii/S037842740901546X - aff1 S. β-Glucuronidase activity is a sensitive biomarker to assess low-level organophosphorus insecticide exposure. Toxicol Let, 193(1); 2010, 115–119.

48. Sabbe MB, Desruelles D, Lissens W. Is beta-glucuronidase a clinical useful biomarker for an acute organophosphorus poisoning? Hum Exp Toxicol,2008; 27(5):431-433.

Furlong CE, Suzuki SM, Stevens RC, Marsillach J, Richter RJ, Jarvik GP, Checkoway H, Samii A, Costa

49. LG, Griffi th A, RobertsJW, Yearout D, Zabetiand CP. Human PON1, a biomarker of risk of disease and exposure. Chem Biol Interact, 2010; 187(1-3): 355–361.

50. Costa LG, Cole TB, Furlong CE. Polymorphisms of Paraoxonase ( PON1) and Their Signifi cance in Clinical Toxicology of Organophosphates. Clin Toxicol, 2003; 41(1): 37-45.

51. Yeung DT, Lenz DE, Cerasoli DM. Human Paraoxonase I: A Potential Bioscavenger of Organophosphorus Nerve Agents. Proteins Cell Regulation, 2008; 6: 151-170.

Page 405: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

403

52. Mackness B, Durrington P, Povey A, Thomson S, Dippnall M, Mackness M, Smith T, Cherry N. 52. Paraoxonase and susceptibility to organophosphorus poisoning in farmers dipping sheep. , 2003; 13(2):81-88.

53. Akgur SA, Ozturk P, Sozmen EY, Delen Y, Tanyalcin T, Ege B. Paraoxonase and acetylcholinesterase activities in humans exposed to organophosphorous compounds. J Toxicol Envir Health, 1999; 58(8): 469-474.

54. Furlong C E, Holland N, Richter R J, Bradman A, Ho A, Eskenazi B. PON1 status of farmworker mothers and children as a predictor of organophosphate sensitivity. Pharmacogenet Genomics, 2006;16:183–190

55. Possamai FP, Fortunato JJ, Feier G, Agostinho FR, Quevedo J, Wilhelm Filho D, Dal-Pizzol F. Oxidative stress after acute and sub-chronic malathion intoxication in Wistar rats.Environ Toxicol Pharmacol. 200; 23(2):198-204.

56. Ranjbar A., Pasalar P., Abdollahi M. Induction of oxidative stress and acetylcholinesterase inhibition in organophosphorous pesticide manufacturing workers. Hum Exp Toxicol, 2002; 21(4):179-182.

57. Ogut S, Gultekin F, Kisioglu AN, Kucukoner E. Oxidative stress in the blood of farm workers following intensive pesticide exposure. Toxicol Ind Health, 2011; 27(9): 820-825.

58. Shadnia S, Azizi E, Hosseini R, Khoei S, Fouladdel S,Pajoumand A, Jalali N, Abdollahi M. Evaluation of oxidative stress and genotoxicity in organophosphorus insecticide formulators. Hum Exp Toxicol, 2005; 24(9): 439-445.

59. Abdollahi M, Mostafalou S, Pournourmohammadi S, Shadnia S. Oxidative stress and cholinesterase inhibition in saliva and plasma of rats following subchronic exposure to malathion. Comp Biochem Physiol Part C: Toxicol Pharmacol, 2004; 137( 1): 29–34.

60. Zhang JW, Lv GC, Zhao Y. The signifi cance of the measurement of serum xanthine oxidase and oxidation markers in patients with acute organophosphorus pesticide poisoning. J Int Med Res, 2010;38(2):458-465.

61. Sudakin DL, Stone DL. Dialkyl phosphates as biomarkers of organophosphates: the current divide between epidemiology and clinical toxicology. Clin Toxicol (Phila). 2011; 49(9):771-781.

62. Krieger RI, Chen L, Ginevan M, Watkins D, Cochran RC, Driver JH, Ross JH. Implications of estimates of residential organophosphate exposure from dialkylphosphates (DAPs) and their relevance to risk. Regul Toxicol Pharmacol, 2012; 64(2):263-266.

63. Peeples ES, Lawrence M, Schopfer LM, Duysen EG, Spaulding R, Volker T, Thompson CM, Lockridge O. Albumin, a New Biomarker of Organophosphorus Toxicant Exposure, Identifi ed by Mass Spectrometry. Toxicol Sci, 2005; 83, 303–312.

64. Williams NH, Harrison JM, Read RW, Black RM. Phosphylated tyrosine in albumin

Page 406: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

404

Chapter 55

Development of an In-Vitro Assay of Apparent Permeability Across Artifi cial Membranes

for Antidotal Oximes

Victor VOICUa, Andrei Valentin MEDVEDOVICIb, Dalia Simona MIRONc, Flavian Ștefan RĂDULESCUc,1

aUniversity of Medicine and Pharmacy Carol Davila, Faculty of Medicine, Department of Clinical Pharmacology, Toxicology and

Psychopharmacology, Scoala Floreasca Street #8, Bucharest-011643, Romania.

bUniversity of Bucharest, Faculty of Chemistry, Department of Analytical Chemistry, Bucharest, Romania.

cUniversity of Medicine and Pharmacy Carol Davila, Faculty of Pharmacy, Bucharest, Romania.

Abstract. The paper presents the development of an in-vitro assay of the apparent permeability, adapted for compounds belonging to highly important group of acetylcholiesterase reactivators. The transfer of oxime compounds with different chemical structures (tertiary and quaternary oximes, entities with aliphatic or aromatic side chains and potentially increased lipophilicity) was assessed using artifi cial hydrophobic membranes impregnated in aqueous phosphate buffer or in biorelevant organic solvents. The apparent permeability values were correlated with the estimated n-octanol water partition coeffi cient. The experimental data indicated that lipophilic oximes have an increased potential of transfer across biological membranes and could represent promising candidates for a new approach in the development of new centrally acting antidotes.

Keywords. oximes, artifi cial membranes, in-vitro, permeability, ADME.

Introduction

The reactivators of human acetylcholinesterase inhibited by organophosphorous compounds have mainly a tertiary or quaternary oxime structure [1]. The mechanism of action has been described as nucleophilic attack on the phosphate segment, at the level of the inhibited active site of the enzyme. Both active forms of the antidotal entity are polar.

1 Corresponding Author (e-mail address: fl [email protected]).

Page 407: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

405

The cationic structure is necessary for the distribution at the site of action, whereas the anionic structure is responsible for the intimate mechanism of reactivation. Therefore, the profi le of the antidote is dominated by a moderate to high polarity, critically altering its biopharmaceutical profi le. The therapeutic effi cacy is rather limited, the available reports revealing the lack of a universal reactivator, able to signifi cantly improve the survival rate after exposure to various organophosphorous (OP) compounds, especially nerve agents [2]. This fact can be explained to a considerable extent by the complexity of the mechanism of action of the toxicants. There are signifi cant differences between the patterns of the OP-induced ageing processes or the stability and toxicological profi le of the OP-oxime complexes [3]. Moreover, the currently in-vitro and in-vivo experimental protocols applied for in-depth assessment of the toxic and antidotal mechanisms are not standardized. Therefore, the available data cannot be globally analyzed in order to get accurate insights. The mentioned aspects explain only partially the current status in the research and development of new compounds with antidotal properties. A key issue is to consider the existing difference between the physico-chemical characteristics of the two entities co-existing in the context of acute exposure and emergency therapeutic approaches [4]. Those obvious differences are the basic elements generating various in-vivo absorption and distribution profi les, which must be considered in order to initiate an optimal use, even for the existing reactivating structures having a long experience. The toxicant and the reactivating oxime entities belong to two distinct, even opposed chemical groups. The OP are highly lipophilic, with a rapid distribution to the lipidic compartments (especially the central nervous system), whereas the quaternary ammonium compounds such as the pyridinic oximes have a marked hydrosolubility, triggered by its notable polarity [5,6].

It must be pointed out that central role of the permeability across the biological, phospholipidic membranes has been recognized for the biological fate of xenobiotics [7]. Besides solubility, this biopharmaceutical parameter has been used for interpretation and prediction of several key pharmacokinetic phases [8]. Traditionally, it has been applied for the characterization of orally administered drug entities, emphasizing on the impact of the intestinal barrier. Various in-vitro screening tools have been developed, based on cellular layers or artifi cial membranes. The group of Camenisch G et al [9] has developed a specifi c in-vitro assay for the evaluation of apparent permeability, involving horizontal diffusion cells and artifi cial interfaces impregnated with an organic, biorelevant solvent. The experimental data has been correlated with the intestinal absorption and permeability across Caco-2 cells monolayer, suggesting that this protocol can generate information on the potential distribution at the level of various biological barriers. It seems that role of various molecular descriptors, e.g. molecular size, polar surface area and lipophilicity, could be globally assessed.

The aim of the current paper was to develop an adapted in-vitro protocol for the evaluation of the apparent permeability for oximes with proven or prospected antidotal potential. The selected compounds were representative for three main subgroups of relevant chemical entities, with tertiary, quaternary or enhanced lipophilicity structures. The experimental data were correlated with values of the biorelevant molecular descriptors and use for the evaluation of potential consequence on their in-vivo distribution.

Page 408: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

406

Materials and MethodsThe evaluations of the apparent permeability were conducted on a group of 12

compounds with oxime structures, their structure being presented in fi gure 1. Obidoxime chloride, pralidoxime chloride and HI6 chloride are quaternary ammonium compounds and are frequently used as reference entities in the screening of reactivating capacity. Diacetylmonoxime (DAM) and monoisonitrosoacetone (MINA) are tertiary oximes, with proven ability to reach the brain regions and consequently, to counteract the central effects of OP. 4PAPE, PAO, PAL, PAD, 2PATB, 3PATB and 4 PATB were previously synthesized and evaluated by Okuno S et al [10] and represent alternative structure with enhanced lipophilicity by addition of various side chains, linear or branched, aliphatic or aromatic. Notably, those compounds are quaternary (pyridinium) oximes, similar to the fi rst group.

Figure 1. The chemical structure of the evaluated oximes.

In-vitro evaluation protocol on side-by-side diffusion cells

The experimental protocol included a system with three horizontal diffusion cells (PermeGear, United States). The nominal volume of donor and receptor compartments was 3 ml, with a diameter of 10 mm of the transfer section, individual upper sampling ports and magnetic stirrers. The assembly was sealed with a metal clamp and 2 tefl on gaskets. Due to the prospected hydrophilic characters of the compounds, a hydrophobic membrane was selected (polycarbonate, 0.2 μm mean pore size, batch 202119, GE Water&Process Technologies). The membranes were soaked at room temperature for 12 hours in saline phosphate buffer (PBS, pH=7.4) or n-octanol saturated with PBS (in the last case, the membranes were washed superfi cially with PBS). The cell compartments were fi lled with 2,7, respectively 3 ml of aqueous buffer and the system was maintained at 37±0.5°C for equilibration for 10 min, under stirring at 500 rpm. A volume of 300 μl from stock solution (400 μg/ml in PBS) of each oxime was added in the donor compartment. Samples of 300 μl were collected from the receptor compartment at 30, 60, 90, 120, 150 and 180 min. The removed volume was replaced with fresh media. Each evaluation was performed in triplicate.

Page 409: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

407

Quantitative evaluation of the oximesThe amount of oxime in the collected samples was quantifi ed after appropriate

dilution, using spectrophotometric methods (8453 UV-Visible Spectrophotometer with Multicell Transport and G1116AA Advanced Software, Rev.B.04.01, Agilent Technologies, Germany).

Determination of apparent permeabilityThe rate of delivery across the membrane at steady state was calculated using the linear

segments of the concentration dependence on time (dCR/dt). The apparent permeability (Papp) for both types of treatment of the membrane was estimated as previously reported [9], using to the formula:

(1)

where VA is the volume of the receptor compartment, CD and CR the initial concentration of the oxime in the donor (40 μg/ml) and, respectively, in the receptor (0 μg/ml), and S is the surface area of the membrane (0.7854 cm2).

Results and discussionsThe two types of treatment applied to the hydrophobic interface represented an adaptation

of model used by Camenisch G [9]. A hydrophilic membrane could not be feasible barrier for the evaluation, especially for PBS treatment. In this case, a rapid equilibration could have generated between the two compartments of the cell, without accurate estimation of the existing differences in the hydrophilic character of the compounds and of their impact on the transfer phenomenon.

In case of membranes with hydrophilic pores, the maximum apparent permeability was obtained for DAM (1.6·106 cm/sec; table 1), probably due to the low molecular weight and volume.

Table 1. The mean values of the apparent permeability (Papp, 106 cm/sec; n=3) across polycarbonate membrane soaked in n-octanol or PBS

Compound Papp, n-octanol Papp, PBS

OBI 0.06 1.10PAM 0.06 0.98

HI6 0.06 1.01DAM 0.13 1.61

MINA 0.11 0.89PAO 0.09 0.53PAD 0.13 0.43PAL 0.21 0.29

PAPE 0.08 1.152PATB 0.08 0.973PATB 0.09 0.904PATB 0.09 1.12

dtdCCCSVP RRDAapp /)(/ ⋅−⋅=

Page 410: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

408

A similar thermodynamic activity was observed for the majority of quaternary ammonium compounds, correlated with the considerable positive charge. The lauryl, dodecyl and octadecyl derivative of pralidoxime (PAL, PAD and PAO) presented a minimum value of the apparent permeability in those conditions (0.29 - 0.53·106 cm/sec). A possible explanation is that the molecular structure of these compounds, composed of a long, linear hydrocarbon side chain and a polar, charged head, confers tensioactive properties. Therefore, it is reasonable to assume that a superfi cial, lipophilic fi lm is generated on the surface of the membrane, decreasing the transfer rate. The position of tert-butyl radical on the pyridine nucleus has little if any infl uence on the apparent permeability across the hydrophilic pores. The treatment with n-octanol changed the rank order within this sub-group, the conformation with lowest fl exibility (ortho-isomer) presenting the highest apparent permeability (0.077 ·106 cm/sec).

Previous reports suggested a sigmoid relationship between the apparent permeability using artifi cial membranes and the distribution coeffi cient n-octanol / aqueous buffer system at physiological pH, logD7.4 [9,11]. This fact confi rmed the utility and biorelevance of the molecular descriptor, overrated in some instances. To the best of our knowledge, the current in-vitro model has not been applied extensively to hydrophilic xenobiotics, corresponding to one of the plateau regions of the correlations (low permeability, high solubility). For hydrophobic pores assay, the presence of long linear side chains in PAL, PAO and PAD oximes triggered deviations from the apparent linear dependence between the logarithmic values of Papp and logD7.4. The higher lipophilicity determined almost an exponential increase in the apparent permeability through the membranes treated with n-octanol (fi gure 2). The region described by the corresponding molecular descriptors (e.g. estimated logD7.4 values) seems to be critical for the partition processes.

Figure 2. The correlation between the experimental values of the apparent permeability and the estimated partition coeffi cient at pH=7.4 (ADMET Predictor, Simulation Plus Inc). Note: ▲ – tertiary oximes; ● – quaternary oximes; ■ – long chain alkyl derivatives of 2PAM.

The results seem to confi rm the current limitation of the classical quaternary oxime in terms of delivery through the biological barriers at the critical sites for reactivation. Moreover, the tertiary structures seems to have an adequate hydro-lipophilic character, which, together with the existing clinical proofs, explain their superiority in the induced

-3,5

-3

-2,5

-2

-1,5

-3 -2 -1 0 1 2 3

Log

Papp

,n-o

ctan

ol

LogD (pH=7.4)

-3,5

-3

-2,5

-2

-1,5

-3 -2 -1 0 1 2 3

Log

Papp

, PB

S

LogD (pH=7.4)

Page 411: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

409

survival rate and counteracting seizures induced by sarin [12]. The drug design procedures focused on the increased permeability of oximes are promising approaches, but two critical aspects need to be concomitantly screened. On one hand, increasing the rate and extent of delivery in the central compartment could generate the expression of the toxicological profi le of the antidote. On the other hand, one should consider to what extent the intrinsic mechanism of action is affected. For example, PAO and PAD have superior penetration profi le across lipid barrier, but in the same time the available data indicate that have almost have of the reactivating activity of 2PAM (46 and, respectively, 59% [10]). As previously mentioned, the polarity is mandatory for an adequate antidotal profi le.

ConclusionsAn adapted in-vitro assay for the evaluation the apparent permeability across artifi cial,

hydrophobic membranes was developed and applied to a series of compound with oxime structure, reactivators of OP-inhibited AChE. The results confi rmed the biopharmaceutical limitations of quaternary ammonium compounds, due to their decreased lipophilicity. The tertiary oximes displayed an optimal hydro-lipophilic character, favorable to the permeability across biological barriers. The addition of long linear aliphatic side chain to the pyridinium group seems to be a promising approach in generating antidotal tools with improved penetration at the level of the central nervous system, but toxicological and reactivating patterns need to be carefully screened.

AcknowledgementThis work was supported by a grant of the Ministry of National Education, CNCS –

UEFISCDI, project number PN-II-ID-PCE-2012-4-0651. The authors acknowledge the support of Prof.Koichi Sakurada and Dr.Hikoto Ohta, who kindly provided PAO, PAL, PAD, 4PAPE, 2PATB, 3PATB and 4PATB compounds. Parts of these results were generated by ADMET Predictor™ software provided by Simulations Plus, Inc., Lancaster, California, USA.

References

K. Kuca, D. Juna, K. Musilek, Structural requirements of acetylcholinesterase reactivators, Mini Rev Med Chem6 (2006), 269-277.

J. Bajgar, Optimal choice of acetylcholinesterase reactivators for antidotal treatment of nerve agent intoxication, Acta Medica (Hradec Kralove) 53 (2006), 207-211.

F. Worek, T. Wille, M. Koller, H. Thiermann, Structural requirements for effective oximes - evaluation of kinetic in vitro data with phosphylated human AChE and structurally different oximes, Chem Biol Interact, 203 (2013), 125-128.

V. Voicu, F. Rădulescu, A. Medvedovici, Toxicological considerations of acetylcholinesterase reactivators, Expert Opin Drug Metab Toxicol, 9 (2013), 31-50.

V. Voicu, J. Bajgar, A. Medvedovici, F. Radulescu, D. Miron, Pharmacokinetics and pharmacodynamics of some oximes and associated therapeutic consequences: a critical review, J Appl Toxicol, 30 (2010), 719-729.

V. Voicu, H. Thiermann, F. Rădulescu, C. Mircioiu, D. Miron, The toxicokinetics and toxicodynamics of organophosphonates versus the pharmacokinetics and pharmacodynamics of oxime antidotes: biological consequences, Basic Clin Pharmacol Toxicol, 106 (2010), 73-85.

H. van de Waterbeemd, Which in vitro screens guide the prediction of oral absorption and volume of distribution? Basic Clin Pharmacol Toxicol, 96 (2005), 162-166.

Page 412: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

410

U. Fagerholm, The role of permeability in drug ADME/PK, interactions and toxicity - presentation of a permeability-based classifi cation system (PCS) for prediction of ADME/PK in humans, Pharm Res, 25 (2008), 625-638.

G. Camenisch, G. Folkers, H. van de Waterbeemd, Comparison of passive drug transport through Caco-2 cells and artifi cial membranes, Int J Pharm, 147 (1997), 61-70.

S. Okuno, K. Sakurada, H. Ohta, H. Ikegaya, Y. Kazui, T. Akutsu, T. Takatori, K. Iwadate, Blood-brain barrier penetration of novel pyridinealdoxime methiodide (PAM)-type oximes examined by brain microdialysis with LC-MS/MS, Toxicol Appl Pharmacol., 227 (2008), 8-15.

G. Camenisch, G. Folkers, H. van de Waterbeemd , Shapes of membrane permeability-lipophilicity curves: extension of theoretical models with an aqueous pore pathway, Eur J Pharm Sci, 6 (1998), 325-329.

T. Shih, J. Skovira, J. O’Donnell, J. McDonough, Treatment with tertiary oximes prevents seizures and improves survival following sarin intoxication, J Mol Neurosci, 40 (2010), 63-69.

Page 413: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

411

Chapter 56

Assessment of the Reactivating Potency of Different Oximes in Tabun Poisoned Rats

Ivan SAMNALIEV, Iskra PETROVAMilitaryMedicalAcademy, Sofi a, 1606, Bulgaria

Department “Disaster Medicine and Toxicology”, MMALaboratory of Military Toxicology

Abstract. Currently available antidote therapy of nerve agent poisoning consists of a triple medical approach – atropine, oxime reactivators and diazepam as an anticonvulsant. Commonly used oximes (Obidoxime, 2-PAM, TMB-4 and HI-6) possess limited effi cacy against tabun because of steric and structural changes of tabun-inhibited acetylcholinesterase (AChE) that hinder oximate anione successfully to attack enzyme-inhibitor conjugate. The insuffi cient reactivating potency of currently used oximes is the main reason for searching of new compounds with different chemical structure and increase affi nity to the tabun-inhibited enzyme. The aim of this study was to assess the reactivating effectiveness of some oximes synthesized in our laboratory, as analogous of well known reactivators HLÖ-7,K-048 and K-074, in tabun poisoned rats. The results obtained showed that HLÖ-7is the most effective and ensured reactivation of brain AChE inhibited by 0.5 LD50 tabun.

Key words: tabun, brain acetylcholinesterase, oxime reactivators.

1. Introduction

Highly toxic organophosphorus compounds such as soman, tabun, sarin, cyclosarin and VX, known as nerve agents, are potent inhibitors of enzyme acetylcholinesterase in central and peripheral nervous systems and this the main mechanism of their toxicity in mammals. Severe inhibition of AChE in the brain initiates seizures and epileptiform activity which is contributed to neuropathology caused by these agents (McDonough, Shih, 1997; Koplovitz et al., 2001; Shih et al., 2003). The pronounced inhibition of enzyme activity in blood and peripheral tissues (skeletal muscles, diaphragm) lead to development of severe intoxication (Mars, 2007). Because of this reason the reactivation of both brain and peripheral AChE activity plays a very important role in reducing nerve agent induced toxicity and has life-saving effect. Unfortunately currently used oxime reactivators don’t possess enough effi cacy against all representatives of nerve agents. There is no so called wide-spectrum oxime and the great challenge are poisonings caused by soman and tabun (Kassa, 2002, Kassa et al., 2006). Partially active against tabun are Obidoxime and TMB-

Page 414: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

412

4 while HI-6 is assumed to be most effective against soman (Jokanovic, 2006).Numerous compounds have been synthesized and tested as potential reactivators of tabun-inhibited AChE but only few of them demonstrated such activity (Jokanovic, 2009; Съмналиев, Петрова, 2010). In Chezh Republic were synthesized so called “K- oximes” and among them K-027, K-048 and K-074 possess reactivating potency against tabun (Kassa et al., 2006). In Germany was synthesized HLÖ-7 which is considered to be the only wide spectrum oxime till now (Eyer et al., 1992).

2. Material and methods.Animals. Experiments were carried out on male albino “Wistar” rats (180-220 g)

obtained from Military Medical Academy. Prior to the experiments they were housed with 6 animals per cage. Temperature was kept at 18-22 0C, humidity was maintained at 50-65% and 12 hr light-dark cycle was available. Rats were allowed to standard rodent food and tap water ad libitum. Experiments were carried out in accordance with the Regulation № 20/ 2012 “Minimal requirements for protection and humanely attitude to the experimental animals”.

Nerve agents: The native Tabun (GD, 86%) obtained from the stocks of the Laboratory of Military Toxicology, MMA, Sofi a was used in the current study. Tabun was injected i.m. in a fi nal concentration 0.1 g/ml. and at a dose 0.5 LD50.

Oxime reactivators. Three newly synthesized oximes – BT-10, BT-07 (4 M) and BT-03 were tested for reactivating potency against tabun. These compounds are analogous of the original oximes known as HLÖ-7, K074 and K027, respectively. BT-10 was synthesized by a modifying method (Съмналиев и съавт., 2012).The fi nal purity of oximes was > 98%. Oximes combined with atropine (10 mg/kg) were applied i.m. 1 minute after the challenge. Obidoxime was used as a conventional oxime at the same conditions of experiment. All oximes were applied at

Fig. 1. Structural formulas of tested oxime reactivators.

Page 415: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

413

Biochemical investigations. Enzyme activity of brain, erythrocyte and skeletal acetylcholinesterases and BuChE was determined 24 hr after the challenge by using Ellman’s methods (1961) and “HospitxDiagnostics”.

Date analysis: Statistical signifi cance was determined by using Student’s t-test and differences were considered signifi cant when p<0.05. Statistical evaluation was determined with the relevant computer programs (Basic Statistics, Version 6).

3. Results and discussion.The results obtained from the current study are presented on table 1. Poisoning with 0.5

LD50 tabun caused the most profound inhibition of the brain AChE – approximately 50% in comparison to the control group. This fi ndings is according to expectation because of previously studies have shown that G-agents, and especially tabun, cause rapid inhibition of the enzyme activity in central nervous system (Shih et al., 2005). From the tested oximes only BT-10 demonstrated reactivating potency in this case and ensured nearly 80% reactivation of the brain AChE. Surprisingly obidoxime did not show any effi cacy against tabun-inhibited brain AChE. Our results do not confi rm previously published data concerning reactivating activity of K027 and K074 in case of tabun-inhibited brain AChE (Kassa et al., 2006, 2007, 2008). In the current study BT-03 and BT-07 (4M) were not able to reactivate enzyme activity in CNS. The possible reason of this inconsistency probably is due to the different doses of oximes used in the current and previous experiments. The effi cacy of BT-10 (HLÖ-7) to reactivate brain AChE supports the fi ndings of other investigations that have shown this oxime to be very effective against nerve agents (Clement et al, 1992; Eyer et al., 1992; Worek et al., 1998). Presently published results obtained from a theoretical study clearly showed that HLÖ-7 is able to reactivate tabun-inhibited AChE (Yanwei et al., 2012). Our experiments were performed by using in vivo model and the fi ndings that HLÖ-7 reactivated brain enzyme activity gives an important information concerning the possibility of this compound to penetrate blood brain barrier in the current experimental conditions. In this way our results confi rm the admission that the benefi cial effect of oximes in treatment of nerve agent poisoning may result from reactivation of AChE activity both in peripheral and central

Page 416: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

414

compartments (Shrot et al., 2009). The changes of erythrocyte and skeletal AChE in this study were not signifi cant in comparison to the control group, may be, due to the relatively low doses of tabun. In some extent BT-03 demonstrated reactivating potency for both these sources of the enzyme activity.

Conclusion: In conclusion on the basis of the result obtained could be accept that BT-10 (HLÖ pt-7) possesses high reactivating activity for tabun-inhibited rat brain AChE and probably this is the reason for the antidote effi cacy of this promising oxime.

Table 1

Changes of the enzyme activity 24 hr after the poisoning with 0.5 LD50 tabun followed by treatment with tested oxime reactivators

ExperimentalGroups

Erythrocite AChE (U/L)

BrainAChE (U/L)

SkeletalAChE (U/L)

SerumBuChE (U/L)

Тabun 0.5 LD50

(% inhibition)

3.76 ± 0.58 (†††)

(9.0)

0.088 ± 0.027(***) (†††)

(43.6)

0.251 ± 0.11(†††)(11.4)

1121.5 ± 115.6(†)

(3.0)

Tabun 0.5 LD50+ ВТ-10(% reactivation)

2.46 ± 0.85(+++) (†††)

(0.0)

0.142 ± 0.016(+++) (†††)

(79.5)

0.418 ± 0.03(+++) (†††)

(321.8)

1045.8 ± 188.9

(0.0)

Таbun 0.5 LD50+ ВТ-04 (7М)(% reactivation)

1.80 ± 0.70(†††)

(0.0)

0.082 ± 0.011(+++) (0.0)

0.406 ± 0.04(†††)

(321.0)

974.0 ± 323.9

(0.0)Тabun 0.5 LD50+ ВТ-03(% reactivation)

4.19 ± 0.57(+++)(16.2)

0.088± 0.010 (+++) (0.0)

0.292 ± 0.07(+++)(71.9)

865.5 ± 215.5(†)

(0.0)Таbun 0.5 LD50+ Оbidoxime(% reactivation)

3.250 ± 0.85(†)

(0.0)

0.065 ± 0.015 (+++) (†)

(0.0)

0.245 ± 0.07(+++)(0.0)

1104.7 ± 159.4

(0.0)

Remarks: 1. * - comparison between control group (NaCl 0.9% only) and poisoned group (0.5 tabun only); † - comparison between poisoned group (0.5 tabun only) and groups treated with tested oximes; + - comparison between group treated with BT-10 and groups treated with other two oximes; 2. ** - p < 0.01, *** - p < 0.001; 3. Control values of the different biochemical parameters: ErAChE – 4.13 ± 0.62; BrAChE – 0.156 ± 0.059; skeletal AChE – 0.283 ± 0.07; BuChE – 1155.8 ± 587.7.

References

1. Съмналиев, И., Петрова, И. Съвременно състояние на антидотната терапия на отравянията с нервни агенти. Реактиватори на холинестеразата. // Военна медицина, 2010, № 3, 3-10.

2. Съмналиев, И., Петрова, Свиняровр И. Оценяване на новосинтезиран биспиридиниев диалдоксим като реактиватор на холинестеразата при интоксикация със зоман на модела на плъховел // Военна Медицина, 2012 (под печат).

3. Clement, J. G. et al. Effi cacy of HLo-7 and pyrimidoxime as antidotes of nerve agent poisoning in mice. // Arch. Toxicol., 66, 1992, 216-219.

4. Eyer, P., Hagedorn, I., Klimmek, R., Lippstreu, P., Loffl er, M., Oldiges, H. et al., Hlo-7 dimetansulphonate

Page 417: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

415

a potent bispyridinium-dioxime against anticholinesterases .// Arch. Toxicol., 1992, 66, 603 – 621. 5. McDonough, J. H., Shih, T-M. Neuropharmacological mechanisms of nervr-agents induced seizure abd

neuropathology. // Neurosci. Biochev. Rev., 21, 1997, 559-579.6. Jokanovic M., Stojiljkovic M. Current understanding of the application of pyridinium oximes as

cholinesterase reactivators in treatment of organophosphate poisoning. // Eur. J. Pharmacol., 553, 2006, 10-17.

7. Jokanovic, M. Medical treatment of acute poisoning with organophosphorus and carbamate pesicides. // Yoxicol. Let., 2009, doi:10.1016/j.toxlet.2009.07.025

8. Kassa J. Review of oximes in the antidotal treatment of poisoning by organophosphorus nerve agents. // L. Toxicol. Clin. Toxicol., 2002, 40, 803-816.

9. Kassa, J., Kuca, K., Cabal, J., Paar, M. A comparison of the effi cacy of new asymmetric bispyridinium oximes (K027, K048) with currently available oximes against tabun by in vivo methods. // J. Toxicol. Environ. Health. Part A, 2006, 69, 1875 – 1882.

10. Kassa, J., Kuca, K., Karashova, J., Musilek, J., Jun, D., Bajgar, J., Cabal, J., Stetina, R., Fusek, J. Development of New Reactivators of Tabun Inhibited Acetylcholinesterase and the Evaluation of Their Effi cacy by in Vitro and in Vivo Methods.In: Defence against Effects of Chemical Hazards: Toxicology, Diagnosis and Medical Countermeasures. Meeting Proceedings RTO-MP, 2007, 17-1 – 17-12.

11. Kassa, J., Jun, D., Karasova, J., Bajgar, J., Kuca, K. A comparison of reactivating effi cacy of newly developed oximes (K074, K075) and currently available oximes (obidoxime, HI-6) in soman, cyclosarin and tabun-poisoned rats. // Chem. Biol. Interact., 2008b, Sep 25, 175 (1-3): 425-7. Doi: 10.1016/j.cbi.2008.05.001

12. Koplovitz, I.,Schulz, S., Shutz, M., Railer, R., Macalalag, R., Schons, M., McDonough, J. Combination anticonvulsant treatment of soman-induced seizures. // J. Appl. Toxicol., 21, 2001, Suppl 1, S53-55.

13. Shih, T-M., Duniho, S. M., McDonough, J. H. Control on nerve agent-induced seizures is critical for neuroprotection and survival.// Toxicoll. Appl. Pharmacol., 2003, 188, 69-80.

14. Marrs, T. Toxicology of organophosphate nerve agents. // Chemical Warfare Agents. Toxicology and Treatment. Edit. T.M. Marrs, R.L. Maynard, F.R. Sidell, 2nd Edition, John Wiley&Sons, England, 2007, 191-221.

15. Shih, T-M., Kan, R. K., McDonough, J. H. In vivo cholinesterase inhibitory specifi city of organophosphorus nerve agents. Chem. Biol. Interact, 2005, 157-158, 293-303.

16. Shrot, S., Markel, G., Dushnitsky, T., Krivoy, A. The possible use of oximes as antidotal therapy in organophosphate-induced brain damage. // Neurotoxicology, 30, 2009,

17. Worek F., Widmann R., Knopff O., Szinicz L.. Reactivating potency of obidoxime, pralidoxime, HI-6 and HlЦ-7 in human erythrocyte acetylcholinesterase inhibited by highly toxic organophosphorus compounds.// Arch Toxicol, 1998, 72, 237-243.

18. Yanwei, L., Likai, D., Yueming, H.,Xiaomin, S., Jingtian, H. Theoritical study on the aging and reactivation mechanisms of tabun-inhibited acetylcholinesterase by using the qantum mechanical/molecular mechanical method. // Can. J. Chem., vol 90, 4, 2012, 376-383.

Page 418: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

416

Chapter 57

Investigation of Prophylactic Effi cacy of Drug Mixture Consisting of Physostigmine and Procyclidine Applied Alone or Followed

by Antidote Therapy in Soman Poisoning

Ivan SAMNALIEV, Trifon IVANOVMilitaryMedicalAcademy, Sofi a, 1606, Bulgaria

Department “Disaster Medicine and Toxicology”, MMALaboratory of Military Toxicology

Abstract. The main goal of a successful prophylaxis is to minimize or alleviate the toxic signs caused by nerve agents ( tabun, sarin, soman, cyclosarin, VX) and to ensure life saving effect. Such an effect could be achieved by using different drug mixtures consisting of usually 1. Reversible cholinesterase inhibitor and 2. Antimuscarinic compounds or compounds possess ability to affect not only cholinergic but other neuromediators in central nervous system. The purpose of the current study was to investigate prophylactic effi cacy of a drug mixtures consisting of physostigmine and procyclidine applied before poisoning with lethal doses of 2 nerve agent soman with or without additional antidote therapy ( HI-6, an oxime reactivator + atropine + midazolam, as a benzodiazepine). The results obtained showed that a prophylactic mixture consisting of physostigmine (1.0 mg/kg) and procyclidine (3.0 mg/kg) applied 30 and 60 min prior to intoxication is very effective against lethal effects of 1.5 and 2.0 LD50 soman when poisoning was followed by antidotal treatment with HI-6, atropine and midazolam.

Key words. Nerve agents, prophylaxis, physostigmime, procyclidine, midasolam, antidotal treatment

1. Introduction

The acute toxicity of organophosphorus (OP) compounds, including nerve agents ( sarin, tabun, soman, cyclosarin, VX), in mammals is primarily due to their irreversible binding to the esteric site of acetylcholinesterase (AChE, EC 3.1.1.7.) in the nervous system and a subsequent accumulation of acetylcholine in the synaptic cleft and at the myoneural junctions. The inhibition of AChE in the diaphragm and brain is important due to the

1 Corresponding Author: Prof. Christophor Dishovsky, Military Medical Academy, 3,St.G.Sofi iski Str., Sofi a 1606, Bulgaria, E-mail: [email protected]

Page 419: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

417

fact that death is attributed to respiratory failure (Tuovinen et al., 1999; Mars, 2007). The currently available medical approach aimed to counteract nerve agent’s toxicity consists of two steps. The fi rst one is pretreatment with pyridostigmine, a reversible inhibitor of AChE with peripheral action and the second step is post exposure antidote treatment. The commonly accepted treatment comprises atropine, an oxime reactivators and diazepam as an anticovulsant (Bajgar, 2004; Layish et al., 2005).

Many investigations have shown that physostigmine is much more effective, than pyridostimine, in counteracting nerve agent’s toxicity because of its ability to penetrate blood brain barrier (Philippens et al., 2000, 2006; Bajgar, 2004; Bajgar et al., 2009; Myrher, 2009). Additionally when physostigmine is combined with a central acting antimuscarinic compound, such as scopolamine, trihexyphenidyl, biperidene, a high level of protection against lethal toxic effects of nerve agents has achieved (McDonough, Shih, 1997;Weherell et al., 2002; Muggleton et al., 2003;Samnaliev, 2012).

The purpose of the current study was to investigate prophylactic effi cacy of a drug mixtures consisting of physostigmine and procyclidine applied before poisoning with lethal doses of 2 nerve agent soman with or without additional antidote therapy ( HI-6, an oxime reactivator + atropine + midazolam, asa benzodiazepine)

2. Material and methods

Animals. Experiments were carried out on male albino “Wistar” rats (180-220 g) obtained from Military Medical Academy. Prior to the experiments they were housed with 6 animals per cage. Temperature was kept at 18-22 0C, humidity was maintained at 50-65% and 12 hr light-dark cycle was available. Rats were allowed to standard rodent food and tap water ad libitum. Experiments were carried out in accordance with the Regulation № 20/ 2012 “Minimal requirements for protection and humanely attitude to the experimental animals”.

Nerve agents: The native Soman (GD, 96%) obtained from the stocks of the Laboratory of Military Toxicology, MMA, Sofi a was used in the current study.

Drugs and drugs combinations. A drug mixture consisted of physostigmine (0.1 mg/kg) and procyclidine (3.0 and 6.0 mg/kg) was applied 30 and 60 min prior to intoxication with soman (1.0, 1.5 and 2.0 LD50 i.m.). An antidotal treatment consisting of cholinesterase reactivator HI-6 (15.6 mg/kg), atropine (10.0 mg/kg) and midazolam (benzodiazepine, 1.0 mg/kg) was used as well. The prophylactic effi cacy was assessed by 24 hours rate of survival after intoxication.

Design of experiments. The current experiment included two parts. In the fi rst part of the study physostigmine combined with procyclidine (3.0 and 6.0 mg/kg) was applied 30 and 60 min before 1.5 LD50 soman, without or with post exposure antidote treatment. As a treatment were used two drug combinations: 1. Atropine (10 mg/kg) + oxime reactivator HI-6 (15.6 mg/kg) and 2. Atropine (10 mg/kg) + oxime reactivator HI-6 (15.6 mg/kg) + midazolam (1 mg/kg).

The rate of survival at different time points and total 24 hours survival after poisoning were registered.

Page 420: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

418

Date analysis: Statistical signifi cance was determined by using Student’s t-test and differences were considered signifi cant when p<0.05. Statistical evaluation was determined with the relevant computer programs (Basic Statistics, Version 6).

3. Results and discussion

In the fi rst part of the study physostigmine combined with procyclidine (3.0 and 6.0 mg/kg), applied 30 and 60 min before 1.5 LD50 soman showed insuffi cient effi cacy and several rats died within fi rst 4 hr of observation. When poisoning was followed by an antidotal treatment ( HI-6+ atropine) all of animals survived for 24 hr. Only one rat died in case of 60 min prophylactic regimen. When midazolam was added to HI-6 and atropine treatment survived 100% of animals.

Table 1

Rate of survival in experimental groups pretreated with physostigmine andProcyclidine 30 and 60 min. prior to intoxication with 1.0 and 1.5 LD 50 soman

without or with post exposure antidote treatment

Time (min)Groups

5 15 30 60 120 240 24 hr

soman 1.0 LD50

100 83.3 66.6 66.6 66.6 50.0 50.0

Physost. – 30’ 100 100 100 100 100 100 100

Physost. – 60’ 100 100 100 100 100 100 100

soman 1.5 LD50

66.6 16.6 16.6 16.6 16.6 16.6 16.6

Physost.+Procycl.3mg/kg - 30’ 100 66.6 66.6 50.0 50.0 50.0 50.0

Physost.+Procycl.3mg/kg - 60’ 100 33.3 33.3 33.3 3.33 33.3 33.3

Physost.+Procycl.6mg/kg - 30’ 100 83.3 83.3 83.3 83.3 83.3 83.3

Physost.+Procycl.6mg/kg - 60’ 100 50.0 50.0 50.0 50.0 50.0 50.0

Physost.+Procycl 30’ + HI-6+A 100 100 100 100.0 100 100 100

Physost.+Procycl 60’ + HI-6+A 100 83.3 83.3 83.3 83.3 83.3 83.3

Physost.+Procycl 30’ + HI-6+A + midazolam

100 100 100 100 100 100 100

Physost.+Procycl 60’ + HI-6+A+ midazolam

100 100 100 100 100 100 100

In the second part of the study poisoning was caused by 2.0 LD50 soman. In this case 30 min prophylactic regimen followed by HI- 6 and atropine treatment ensured 100% survival. When the prophylaxis was postponed to 60 min one of animal died. Again antidotal treatment with HI- 6 , atropine and midazolam protected all animals up to 24 hr. The signifi cance of prophylaxis as medical approach was observed in case of poisoning with 2.0 LD50 soman without pretreatment and followed by treatment with HI- 6 , atropine and midazolam. In this experimental group 4 of 6 rats died to the end of observation.

Page 421: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

419

Table 2

Rate of survival in experimental groups pretreated with physostigmine andProcyclidine 30 and 60 min. prior to intoxication 2.0 LD 50 soman followed by post exposure

antidote treatment

Time (min)Groups

5 15 30 60 120 240 24 hr

soman 2 LD50

100 16.6 0.0 0.0 0.0 0.0 0.0

Physost.+Procycl 30 + HI-6+A

100 100 100 100.0 100 100 100

Physost.+Procycl 60 + HI-6+A

100 83.3 83.3 83.3 83.3 83.3 83.3

Physost.+Procycl 30 + HI-6+A+midazolam

100 100 100 100 100 100 100

Physost.+Procycl 60 + HI-6+A+midazolam

100 100 100 100 100 100 100

HI-6+A+midazolam 100 83.3 83.3 83.3 83.3 66.6 33.3

In the current study we used two drugs as a prophylaxis against poisoning with soman –physostigmine and procyclidine. Our previous study have shown that physostigmine applied intramuscular at a dose of 0.07 mg/kg caused time-dependent inhibition of brain AChE as follow: 43% 30 min after injection and 27% at 60 min. (Samnaliev, Ivanov, 2008). A partial protection of brain AChE from the irreversible inhibition by soman could explain the prophylactic effi cacy demonstrated by physostigmine when is applied in case of poisoning with 1.0 LD50 soman. Procyclidine possesses broad spectrum of pharmacological properties – antimuscarinic, anti-NMDA receptors and antiglutamatergic activity. Many studies have shown that procyclidine, combined with reversible inhibitor of AChE, is very effective in reducing subconvulsions and convulsions produced by lethal doses of nerve agents (Kim et al., 2002; Myhrer et al., 2004a, 2004b; Philipens et al., 2006; Haug et al., 2007). At the same time a relationship between convulsions and the rate of lethality in nerve agent poisoned animals have proved (Shih et al., 2003; Haug et al., 2007).

Our results clearly showed that a drug combination of physostigmine and procyclidine demonstrated only limited protective effi cacy, independently of the time of prophylaxis, against lethal effects in case of poisoning with 1.5 LD50 soman. The rate of survival in experimental groups was approximately 50%. Adding of a post exposure antidotal treatment, consisting of atropine and HI-6, ensured 100% and 83.3% survival in groups pretreated 30 and 60 min., respectively prior to intoxication with 1.5 and 2.0 LD50 soman. When midazolam was used as a third component of the antidote combination all experimental animals survived to the end of observation (24 hr). our results confi rmed two aspects of antidote treatment. The fi rst one is the importance of oxime HI-6 in case of poisoning with soman. Previously studies have shown that HI-6 is the only oxime that demonstrated some activity against soman (Dishovsky, 1989, 1990; Kassa, 2002; Eyer, Work, 2007; Съмналиев, Петрова, 2010). The second one is the effi cacy of midazolam, as a antidote against nerve agents and its positively infl uence on the course of intoxication.

Page 422: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

420

Conclusions. A prophylactic mixture consisting of physostigmine (1.0 mg/kg) and procyclidine (3.0 mg/kg) applied 30 and 60 min prior to intoxication is very effective against lethal effects of 1.5 and 2.0 LD50 soman when poisoning was followed by antidotal treatment with HI-6, atropine and midazolam.

References

1. Съмналиев И., Иванов, Т. Изследване на влиянието на физостигмин и лекарствена комбинациявърху конвулсивната псимптоматика и промените на мозъчната ацетилхолинестераза на плъхове при интоксикация със зоман. Военна Медицина, 2, 2008, 12-18.

2. Съмналиев И., Петрова, И. Съвременно състояние на антидотната терапия на отравянията с нервни агенти. Реактиватори на холинестеразата. Военна Медицинар 3р 2010, 3-10.

3. Dishovsky C., Doctor of Science Work, MMA, Sofi a, 1989 (in Bulgarian).4. Dishovsky C., Reactivators of Cholinestrase, SSA, Sofi a, 1990 (in Bulgarian).5. Bajgar J. Organophosphates nerve agent poisoning: Mechanism of Action, Diagnosis, Prognosis,

Prophylaxis and Treatment. Advancing in Clinical Chemistry, 38 (2004), 151-216.6. Haug, K.H., Myhrer, T., Fonnum, F. The combination of donepezil and procyclidine protects against

soman-induced seizures in rats. Toxicol. Appl. Pharmacol, 220, 2007, 156-163.7. Kassa, J. Review of oximes in the antidotal treatment of poisoning by organophosphorous nerve agents. J.

Toxicol. Clinical Toxicology, Vol. 4, 6, 2002, 803-816.8. Kim Y-B, Cheon, K-C, Hur, G-H, Phi T-S, Choi, S-J, Hong, D., Kang, J-G. Effects of combinational

prophylactics composed of physostigmine and procyclidine on soman-induced lethality seizures and brain injuries.Environ. Toxicol. Pharmacol. 11 (1), 2002, 15-21.

9. Laysh, I., Krivoy, A., Rotman, E., Finkelstein, A., Tashma, Z., Yehezkelli Y. Pharmacologic Prophylaxis against Nerve Agent Poisoning. IMAJ, Vol. 7 March 2005, 182-187.

10. Marrs, T. C. Toxicology of organophosphate nerve agent.// Chemical Warfare Agents. Toxicology and Treatment. Edit. T.M. Marrs, R.L. Maynard, F.R. Sidell, 2nd Edition, John Wiley&Sons2nd Edition, John Wiley&Sons, England, 2007, 191-222.

11. McDonough, JH., Shih, T-M. Neuropharmacological mechanisms of nervr-agents induced seizure abd neuropathology. // Neurosci. Biochev. Rev., 21, 1997, 559-579.

12. Muggleton NG, Bowditch, AP, Grofts, HS, Pearce, PC. Assessment of a combination of physostigmine and scopolamine as pretreatment against the behavioural effects of organophosphates in the common marmoset (Callithrix jacchus). // Psychopharmacology (Berl), 166, 2003 Mar, 212-220.

13. Philippens, I., Melchers, B. P. C., Oliver, B., Brijzeel P.L.B. Scopolamine augments the effi cacy of physostigmine against soman poisoning in guine pigs. Pharmacol. Bioccem. Bechv. 65, 2000, 175-182.

14. Philippens, I., Jongsma, M. J., Vanwersch R. A. P. Effi cacy of pretreatment and treatment against soman intoxication. // Medical Trearment of Intoxication and Decontamination of Chemical Agents in the Area of Terrorist Attack. Edit. C. Dishovsky et al., Springer, Netherlands, 2006, 113-121.

15. Shih, T. M., Duniho, S. M., McDonough, J. H. Control on nerve agent-induced seizures is critical for neuroprotection and survival. Toxicoll. Appl. Pharmacol., 2003, 188, 69-80.

16. Tuovinen, K., Kaliste-Korhonen, E., Raushel, FM, Hanninen, O. Success of pyridostigmine, physostigmine, epistigmine and phosphotriesterase treatments in acute sarin intoxication.Toxicology, Vol. 134, Issues 2-3, 15 June, 1999, 169-178.

17. Wetherell, J., hall, T., Passingham, S. Physostigmine and hyosvcine improves protection against the lethal

Page 423: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

421

Chapter 58

Opportunities for Optimization of the Antidotal Treatment of Acute Poisoning Caused by Soman by Means of Antidote Combinations Containing HI-6

and Centrally Acting Cholynolitics

Chistophor DISCHOVSKI1, Ivan SAMNALIEV1, Dragomir DRAGANOV2

1 Military Medical Academy,Sofi a, 1606, BulgariaDepartment “Disaster Medicine and Toxicology”,

Laboratory of Military Toxicology, Military Medical Academy2 F. Hoffmann-La Roche, Ltd., Pharmaceuticals Division

Global Pharmacokinetic and Toxicokinetic Data

AbstractThe main aim of an antidote therapy of nerve agent poisoning is to ensure protection against lethal effects. At the same time the successful treatment has to minimize severe toxic signs such as convulsions and subconvulsions and related to them so called delayed toxic effects. Currently adopted antidote approach consists of an oxime cholinesterase reactivator (Obidoxime, 2-PAM, HI-6), atropine and diazepam as an anticonvulsant. Atropine is a classic antimuscarinic drug which does not infl uence N-cholinergic receptors. In addition the current hypothesis concerning nerve agent induced brain seizures accepts engaging of excitatory amino acids (glutamate, aspartate) as an important biochemical event leading to status epilepticus and possible brain damages. There are some drugs, for example, anti-Parkinson (Biperiden, Trihexyphenidil) which possess anticonvulsive activity used in a prophylactic regimen in case of nerve agent intoxication. The goal of this study was to assess the therapeutic and anticonvulsive effi cacy of two- and three component drug mixtures ( HI-6 and Atropine or HI-6 and Biperiden or HI-6, Atropine and Biperiden ) in soman poisoned rats.

Key words: soman, antidote treatment, anticonvulsant activity, HI-6, atropine, Biperiden.

1. Introduction.Highly toxic organophosphorus compounds known as nerve agents ( tabun, sarin,

soman, cyclosarin, VX) are still considered as a potential threat for terroristic act. The events in Japan (1995 in Tokyo and 1994 in Matsumoto) where gas sarin was used with terroristic aim strongly increase these apprehensions (Okomura et al., 1997, 2002). Currently adopted antidote approach consists of an oxime cholinesterase reactivator (Obidoxime, 2-PAM, HI-6), atropine and diazepam as an anticonvulsant (Mars, 2007). The quickly reducing

Page 424: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

422

of the nerve agent produced convulsions plays an important role in a successful antidote treatment (Shich et al., 2003, Myhrer et al., 2005,2006, 2007a,b). Antidotal treatment could be optimized through inclusion of centrally acting compounds which possess both antimuscarinic and antiglutamatergic activity. There are some very well known medicines (Benactizine, Procyclidine, Biperiden, Trihexyphenidil) used for treatment of Parkinson’s disease because of their ability to affect different systems, not only muscarinic, in central nervous system. Pharmacological all of them act as antagonists of the excitatory amino acids – glutamate and aspartate. Many studies have shown that used as prophylaxis of nerve agent poisoning these compounds are able to minimize suconvulsions and convulsions (Shih, McDonough, 1997, 2000; Myhrer et al., 2005; Съмналиев, 2012). In this reason it is of interest to assess their antidote and anticonvulsive effi cacy applied as a treatment after the challenge with nerve agents.

2. Material and methods.

Animals. Experiments were carried out on male albino “Wistar” rats (180-220 g) obtained from Military Medical Academy. Prior to the experiments they were housed with 6 animals per cage. Temperature was kept at 18-22 0C, humidity was maintained at 50-65% and 12 hr light-dark cycle was available. Rats were allowed to standard rodent food and tap water ad libitum. Experiments were carried out in accordance with the Regulation № 20/ 2012 “Minimal requirements for protection and humanely attitude to the experimental animals”.

Nerve agents: The native soman (GD, 96%) obtained from the stocks of the Laboratory of Military Toxicology, MMA, Sofi a was used in the current study.

Drugs. The following compounds were usede in the current study:• Cholinesterase reactivator. We used НІ-6 hydrochloride, (4-carbamoyl-pyridinium-

1-methyl) (2-hydroxyiminomethyl-pyridinium-1-methyl) ether dichloride synthesized in Laboratory of Military Toxicology, MMA with >98% purity ( HPLC method);

• Cholinolytics. Atropine sulfate (SOPHARMA, Bulgaria) and Biperiden lactate (Akineton, аmp. 1 ml 5 mg, Desma GmbH, Germany).

Drug combinations tested: НІ-6 ( 0 и 36.0 mg/kg), Biperiden lactate ( (0, 1.25, 2.5и 5.0 mg/kg) and Аtropinesulfate (0, 10 mg/kg). Control group (n = 12) was treated with NaCl (0.9%) and after that poisoned with 2.0 LD50 soman

All compounds were applied i.m. at a volume 0.1 ml/100 gb.w..• Soman. Acute intoxication was caused by 2.0 LD50 soman, i.m. at a volume0.1

ml/100 gb.wDesign of experiments. For the assessment of the therapeutic and antidote effi cacy of the drug combinations

tested the next groups were used:

GroupsАtropine sulfate

(mg/kg)НI-6

(mg/kg)Biperiden(mg/kg)

1 10.0 0.0 1.25 2 10.0 0.0 2.5 3 10.0 0.0 5.0

Page 425: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

423

4 10.0 36.0 0.0 5 10.0 36.0 1.25 6 10.0 36.0 2.5 7 10.0 36.0 5.08 0.0 36.0 1.25 9 0.0 36.0 2.5 10 0.0 36.0 5.011 10.0 0.0 0.0

Antidotal and anticonvulsive effi cacy. During the experiments we assessed two general parameters – rate of survival and anticonvulsive activity. The rate of survival was recorded at different time points up to 24 hr after the challenge.

Evaluation of anticonvulsive effi cacy. The anticonvulsant effi cacy of the drug mixtures was determined by using a seven degree scale for assessment of the toxic signs observed after the challenge by soman: 0 - none toxic signs, 1 - hyperactivity, 2 - shewing and/or salivation, 3 - fasciculations and/or tremor, 4 - subconvulsions, 5 - convulsions and 6 - death. Observations were done at 5, 10, 15, 30 min, 1, 1.5, 2, 2.5, 3, 4 and 24 hours after the intoxication. The grades for a given animal were summed across the observation time to obtain a severity score.

Date analysis: Statistical signifi cance was determined by using Student’s t-test and differences were considered signifi cant when p<0.05. Statistical evaluation was determined with the relevant computer programs (Basic Statistics, Version 6).

3. Results and discussion.Rate of survival. The results obtained for the effi cacy of the drug mixtures tested to

reduce the lethal effects of 2.- LD50 soman are presented on the table 1. It is obviously that HI-6 plays a very important role for antidote activity, generally. In the absence of oxime the rate of lethality increased progressively irrespective the dose of Biperiden and atropine. When HI-6 was applied, as a component of antidote treatment, the rate of lethality sharply decreased. In group treated with HI-6 and atropine (without Biperiden) 75% of rats survived to the end of observation. At the same time an antidote treatment with a drug mixture consisted of HI-6 and Biperiden (without atropine) ensured 100% survival of experimental animals. All doses of Biperiden tested in the current study, combined with HI-6, gave approximately the same antidote effi cacy. There were no signifi cant differences between antidote activity of three- and two-component drug mixtures that defi ned a combination of Biperiden and HI-6 as a very effective treatment against supra-lethal doses of soman.

Table 1

Rate of survival in experimental groups treated with drug combinations tested

HI-6 mg/kg 0 0 0 36.0 36.0 36.0 36.0 36.0 36.0 36.0 0

Bip. mg/kg 1.25 2.5 5.0 0 1.25 2.5 5.0 1.25 2.5 5.0 0

Atr. mg/kg 10.0 10.0 10.0 10.0 10.0 10.0 10.0 0 0 0 10.0

Page 426: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

424

GroupsTime (min) 1 2 3 4 5 6 7 8 9 10 11

5 100 100 100 100 100 100 100 100 100 100 75.0

10 75.0 100 100 100 100 100 100 100 100 100 50.0

20 50.0 25.0 50.0 100 100 100 100 100 100 100 030 50.0 25.0 50.0 75.0 100 100 100 100 100 100 060 25.0 25.0 25.0 75.0 100 100 100 100 100 100 0

90 25.0 25.0 25.0 75.0 100 100 100 100 100 100 0

120 25.0 25.0 25.0 75.0 100 100 100 100 100 100 0

180 25.0 25.0 25.0 75.0 100 100 100 100 100 100 0

210 25.0 25.0 25.0 75.0 100 100 100 100 100 100 0240 25.0 25.0 25.0 75.0 100 100 100 100 100 100 0

24 hr 0 25.0 25.0 75.0 100 100 100 100 100 100 0

Anticonvulsant effi cacy. By using seven degree scale for assessment of the severe of poisoning we calculated the mean values for each experimental group at different time points up to 24 hours after the challenge (table 2). The results obtained showed two important things..

Table 2Mean (± SD) values of the severe of intoxication in experimental groups determined

at different time points after poisoning with 2.0 LD50 soman and followed by antidote treatment with drug combinations tested

HI-6 mg/kg 0 0 0 36.0 36.0 36.0 36.0 36.0 36.0 36.0 0

Bip. mg/kg 1.25 2.5 5.0 0 1.25 2.5 5.0 1.25 2.5 5.0 0

Atr. mg/kg 10.0 10.0 10.0 10.0 10.0 10.0 10.0 0 0 0 10.0

GroupsTime 1 2 3 4 5 6 7 8 9 10 11

20 min.

3.7

± 1.5

***

3.6

± 1.5

***

3.8

± 1.3

***

3.5

± 1.2

***

3.3

± 1.1

***

3.1

± 0.5

***

3.1

± 0.7

***

2.3

± 0.8

***

2.9

± 0.7

***

2.6

± 0.5

***

5.0

± 1.3

60 min..

5.3

± 1.5

5.4

± 1.0

5.4

± 1.0

4.7

± 0.9

***

3.2

±0.5

***

3.0

± 0.2

***

3.1

± 0.5

***

2.9

± 0.2

***

2.8

± 0.3

***

2.5

± 0.3

***

6.0

± 0.0

120 min.

5.3

± 1.0

5.5

0.9

5.4

± 0.8

4.7

± 0.7

***

3.2

0.4

***

3.0

± 0.1

***

2.9

± 0.4

***

2.9

±0.2

***

2.7

± 0.4

***

2.8

± 0.3

***

6.0

± 0.0

180 min.

5.3

+ 1.5

5.4

+ 1.3

5.3

+ 1.5

4.9

+ 0.9

*

3.1

+ 0.5

***

3.1

+ 0.3

***

2.9

+ 0.4

***

2.9

+ 0.2

***

2.6

+ 0.3

***

3.1

+ 0.3

***

6.0

+ 0.0

Page 427: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

425

240 min.

5.3

± 1.1

5.4

± 0.9

5.4

± 0.9

4.8

± 0.7

***

3.1

± 0.4

***

3.1

± 0.3

***

2.9

± 0.3

***

2.9

± 0.2

***

2.8

± 0.3

***

3.1

± 0.2

***

6.0

± 0.0

24 hr.

6.0

± 0.0

5.1

± 1.3

5.5

± 0.8

3.0

± 1.5

***

1.3

± 0.7

***

0.8

± 0.7

***

0.0

-

***

0.5

± 0.5

***

0.0

-

***

0.0

-

***

6.0

± 0.0

The fi rst one is the signifi cant role of oxime HI -6 in reducing soman induced subconvulsions and convulsions. In groups treated only with cholinolytics ( atropine and Biperiden) toxic sings were much more demonstrative than in groups treated with drug combination consisted of HI -6 and cholinolytic. The second one is that combination of HI -6 plus Biperiden, regardless the dose of cholinolytic (1.25, 2.5 or 5.0 mg/kg) were very effective to minimize quickly severe toxic signs produced by somsn. There were no signifi cant differences between anticonvulsive effi cacy of drug combination of HI -6, atropine and Biperidene and two-component combination of Biperiden and HI -6. This fi ndings means that a mixture of HI -6 and Biperiden, applied after the challenge, is quite enough to protect animals from the acute toxicity of lethal dose of soman. Currently adopted treatment with a combination of HI -6 and atropine is considered to be the most appropriate in case of soman poisoning. Results obtained from our study confi rmed that this drug combination really demonstrated antidote effi cacy against both lethality and subconvulsions/convulsions produced by soman. At the same time a comparison of protective and anticonvulsive activity of the two-component drug mixtures consisted of HI -6 plus atropine and HI -6 plus Biperieden clearly showed priority of the second one.

During the last two decades the biochemical processes in central nervous system in case of acute poisoning with nerve agents have been extensively studying (Съмналиев, 2003; Fosbraey et al., 1900; Shih et. al., 1991, 1999, 2003; Shih, McDonough, 1997, 2000; Samnaliev, 2000; McDonough, Shih, 1997; Jacobson et al., 1997; de Groot et al., 2002; Myhrer et al., 2005, 2007a, 2007b, 2008, 2010; Myhrer, 2009; Carpentier, 2008; Kimberly et al., 2010; O’Donnel et al., 2010a, 2010b). Currently adopted hypothesis divides the biochemical events in CNS in to three stages: a/ a cholinergic stage that continues during the fi rst 5 min. after the intoxication; b/ a mixed stage that comprises both cholinergic stimulation and activation of excitatory amino acids (glutamate and aspartate) with a duration between 5 min. and 40 min. after the challenge and c/ non-cholinergic stage that is controlled completely by the excitatory amino acids. As a consequences of these biochemical changes in CNS occur brain seizures and if there is no pharmacological treatment fi nally a status epilepticus is developed (Shih, McDonough, 1997, 2000; McDonough, Shih, 1997;Shihetal., 1999). The results obtained from many studies have shown a connection between brain seizures and brain damages. Abnormally high levels of glutamate and related to this activation of AMPA (amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) and NMDA (N-methyl D-aspartate) receptors in CNS is considered to bethe main reason for this fi ndings (Carpentieretal., 2008; Myhrer, 2009).

As a anticonvulsants, in case of nerve agent poisoning, many natural and synthetic cholinolytics, such as Atropine, Scopolamine, Biperiden, Procyclidine, Benactizine and Trihexyphenidil, have been studied (Capacio, Shih, 1991; McDonoug, Shih, 1993;

Page 428: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

426

Anderson et al., 1994; McDonough et al., 1989, 1995; Shih et al., 1999; Shih, McDonough, 2000; Harisson et al., 2004; Myhrer et al., 2006, 2009; Shih et al., 2007; McDonouh, Shih, 2007; Weissman, Raveh, 2008). The results obtained from these works confi rmed the hypothesis mentioned above and clearly showed that drugs which possess a combined pharmacological action – both anticholinergic and antiglutamatergic activity are potent anticonvulsants.

In the current study we used three doses of Biperiden -1.25, 2.5 and 5.0 mg/kg combined with HI -6 and applied 1 min. after poisoning with supralethal dose of somаn (2.0 LD50). Our data strongly confi rm previously observations that Bipriden has properties of an effi cacious antidote against soman. Its activity is expressed in completely protection from lethal effects as well as ability to minimize subconvulsions and convulsions produced by soman. With respect to protective and anticonvulsant activity the drug mixture consisting of HI -6 and Bipriden is more effective than HI -6 and atropine in case of acute soman poisoning.

References

1. Съмналиев, И. Изследване на профилактичната ефективност на лекарствена комбинация, съдържаща физостигмин и проциклидин, въведена самостоятелно или с последваща антидотна терапия при интоксикация със зоман. // Военна Медицина, 2012а (под печат).

2. Съмналиев, И. Оценка на антиконвулсивната ефективност на лекарствена комбинация, съдържаща физостигмин и проциклидин, въведена самостоятелно или с последваща антидотна терапия при интоксикация със зоман. // Военна Медицина, 2012б (под печат).

3. Anderson D. R., Gennings, C., Carter, W. H., Harris, L. W., Lennox, W. J., Bowesox, S. L.Effi cacy comparison of scopolamine and diazepam against soman-induced debilition in guinea pigs. Fund. // Appl. Toxicol.,22, 1994, 588-593.

4. Capacio, B. R., Shih T-M. Anticonvulsant actions of anticholinergic drugs in soman poisoning. // Epilepsia, 32, 1991, 604-615.

5. Carpentier, P., Testylier, G., Baille, V., Pernot, F., Foquin, A., Delacour, C. Soman-induced seizures and related brain demages. // J. Med. CBR Def., Vol. 6, 2008, 1-25.

6. De Groot, D., Bierman, E., Brujinzeel, P., Carpentier, B., Lallement, G., Melchers, B., Phillippens, I. Benefi cial effect of TCP on soman intoxication in guinea pigs: seizures, brain damage and learning behaviour. // J Appl Toxicol, 2002, Vol. 21 (S1), 57-65.

7. Kimberly, D. S., Lumley, L. A., Robison, Ch. L., Meyerhoff J. L., Dillman, J. F.Transcritional analysis of rat pyriform cortex following exposure to the organophosphate anticholinesterase sarin and induction of seizures. J. Neuroinfl am., 2011, 8,8, 83.21.doi: 10.1186/1742-2094-8.83.

8. Harisson P., Sheridan, R., Green, A., Scott, L., Tattersall, J. A guinea pig hippocampal slice model of organophosphate-induced seizure activity. // J Pharmacol. Exp. Ther, 310, 2004 Aug, 678-686.

9. O’Donnell, J. C., Acon-Chen, C., McDonough, J. H., Shih, T-M. Comparison of extracellular striatal acetylcholine and brain seizures activity following acute exposure to the nerve agents cyclosarin and tabun in freely moving guinea pigs. Toxicol. Mech. Meth., 20 (9), 2010b, 600-608.

10. O’Donnell, J. C., McDonough, J. H., Shih, T-M. In vivo microdialysis and EEG activity in freely moving guinea pigs exposed to organophosphorus nerve agents sarin and VX : analysis of acetylcholine and glutamate. // Arch. Toxicol.,2011: DOI 10.1007/s00204-011-0724-z

11. Okumura, T., Takasu, N., Ishimatsu, S., Mijnoki, S., Mitsuhashi, A.,Кumada K., Tanaka, K., Hinohara, S. Report on 640 victims on the Tokyo subway sarin attack. // Ann. Emerg. Med.,28, 1996, 129-135.

12. Okumura, T., Nomura, T., Suzuki, T., Sugita, M.,Takeuchi et al. The dark morning: the experiences and lessons learned from the Tokyo subway sarin attack. In: Chemical Warfare Agents: Toxicology and Treatment, 2nd Ed., Wiley and Sons, Chichester, UK, 2007, 277-303.

13. Marrs, T. Toxicology of organophosphate nerve agents. // Chemical Warfare Agents. Toxicology and Treatment. Edit. T.M. Marrs, R.L. Maynard, F.R. Sidell, 2nd Edition, John Wiley&Sons, England, 2007, 191-221.

Page 429: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

427

14. McDonough JH, Shih, T-M. Pharmacological moduladion of soman-induced seizures. // Neurosci Biobehav Rev, 17 (2), 1993, 203-215.

15. McDonough J. H., Dohterman, L.W., Smith, C.D., Shih, T-M. Protection against nerve agent-induced neuropathology, but not cardiac pathology, is associated with the anticonvulsant action of drug treatment. // Neurotoxicol., 16, 1995a, 123-132.

16. McDonough J. H., Shih, T-M. A study of the NMDA antagonist properties of anticholinergic drugs. // Pharmacol Biochem Behav, 51, 1995b, 249-253.

17. McDonough, J. H., Shih, T-M. Neuropharmacological mechanisms of nervr-agents induced seizure abd neuropathology. // Neurosci. Biochev. Rev., 21, 1997, 559-579.

18. McDonough J. H., Shih TM. Atropine and other anticholinergic drugs. // In: Chemical Warfare Agents. T.C. Marrs, R.L. Maynard, F.R. Sidell . Edd., Wiley, 2007, pp 288-303.

19. Myhrer, T., Andersen, J. M., Nguyen, N. H., Aas, P. Soman-induced convulsions in rats terminated with pharmacological agents aftre 45 min.: Neuropathology and cognitive performance. // Neurotoxicol., Jan. 26, (1), 2005, 15527872.

20. Myhrer T., Enger, S., Aas, P. Pharmacological therapies against soman-induced seizures in rats 30 min following onset and anticonvulsant impact. Eur. J. Pharmacol., 548, 2006, 83-89.

21. Myhrer, T. Neuronal structures involved in the induction and propagation of seizures caused by nerve agents: Implications for medical treatment. // Toxicology, Jul. 1, 2007a, 17689166.

22. Myhrer T., Enger, S., Aas, P. Anticonvulsant effects of damage to structures involved in seizures induction in rats exposed to soman. // Neurotoxicology, 2007b Apr 24.

23. Myhrer, T., Enger, S., Aas, P. Anticonvulsant Effi cacy of drugs with Cholinergic and/or Glutamatergic Antagonism Microinfused into Area Tempestas of Rats Exposed to Soman. // Neurochem. Res., Aug. 21 2008, 17710542.

24. Myhrer, T., Enger, S., Aas, P. Modulators of metabotropic glutamate receptors microinfused into perirhinal cortex: anticonvulsant effects in rate challenged with soman. // E. J. Pharmacol., 03/2010; DOI: 10.1016/j.ejphar.2010.02.047

25. Myhrer, T. Prophylactic and Therapeutic Measures in Nerve Agent Poisoning // Handbook of Toxicology of Chemical Warfare Agents. Edit. R. Gupta, Elsevier Inc. U.K., 2009, 965-975.

26. Samnaliev I. Assessment of the drug combinations containing biperiden and cholinesterase reactivators as a prophylaxis against soman induced poisoning. Vojenske Zdavotnicke Listy, 2002, LXXI 2, 86-92.

27. Shih T-M., McDonough, J. H. Neurochemical mechanisms in soman-induced seizures. // J Appl Toxicol, 1997, 17 (4), 255-264; 1999, 147-153.

28. Shih T-M., McDonough, J. H. Effi cacy of biperiden and atropine as anticonvulsant treatment for organophosphorus nerve agent intoxication. // Arch. Toxicol., 74, 2000, 165-172.

29. Shih, T-M., Duniho, S. M., McDonough, J. H. Control on nerve agent-induced seizures is critical for neuroprotection and survival.// Toxicoll. Appl. Pharmacol., 2003, 188, 69-80.

30. Weissman, B.A., Raveh, L., Therapy agaainst organophosphate poisoning: The importatnt of anticholinergic drugs with antiglutamatergic properties. Toxicol. Appl. Pharmacol., 232, 2008, 351-358.

Page 430: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

428

Chapter 59

Toxidin - Bulgarian Ampule Form of Reactivator of Cholinesterse HI-6

Christophor DISHOVSKY, Silvia STOIKOVA, Iskra PETROVA Department of Medicine of Disasters and Toxicology, Department of Toxicology,

MilitaryMedivalAcademy, Sofi a, Bulgaria

Abstract. Acetylcholinesterase reactivator HI-6 belongs to the mostpromising antidotes used at present time in case of OPC poisonings. In Bulgaria the ampule form Toxidin of this compound was developed in 1986. The new HPLC method was described. The stability and purity of HI-6 from old ampules were tested. The results demonstrated that in this lyophilic form, HI-6 is very stable.

Keywords. Cholinesterase reactivators, synthesis, HI-6, Toxidine, HPLC

IntroductionReactivators of cholinesterase (ChE) are pharmacological drugs used asantidotes

in intoxications with organophosphorous compounds (OPC).The major oximes such as 2PAM, 2PAS, TMB-4 and toxogonin are being applied on a wide range and have been examined in detail.The diffi culty in reactivation of the ChE activity and slight antidote effect concerning intoxication with some OPC, are some of the reasons for continuing the examinations for the creation of new reactivators of ChE. The H-Oximes, called after the person, who for the fi rst time synthesized them with her team, Prof. Ilze Hagedorn, open a new era in this direction [1, 2]. The most well-knowna mong them is HI-6 ((1-(4-imino-carbonylpyridinium) 1-(2hydroxyimino-methyl-pyridinium) dimethylether dichloride).The research so far shows that at the moment it is one of the best reactivators of the inhibited from Soman acetylcholinesterase(AChE). This reactivator as two salts –dichloride and dimethanesulphonate was introduced in several armies.

Reactivator of ChE ToxidinIn Military Medical Academy – Department of Toxicology, HI-6, has been synthe-

sized with the help of a modifi ed method described in our Bg Patent No 52193 [3 ]. The scheme of the synthesis is presented on the fi g.1.

Corresponding Author: Christophor Dishovsky, MMA, Sofi a, e-mail address: [email protected]

Page 431: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

429

Our HI-6 has good antidote activity. That was confi rmed by pharmacolo-gical and toxicological investigations. [5,6,7,8].

First stage:

Second stage:

Fig. 1. Scheme of the synthesis of HI-6. Were :- Intermediate = 1- chloromethoxy - methyl- ( 2-hydroxyiminomethyl-pyridinium) chloride- Toxidine = (1-(4-imino-carbonylpyridinium) 1-(2-hydroxyiminomethyl-pyridinium) dimethylether dichloride

Full investigations have been made as part of the preclinical pharmacological toxicological studies of the HI-6 and its ampoule form - Toxidine (1986) [5,6].Biochemical and hematological research in dogs does not show statistically reliable reactions. This is a proof that HI-6 does not cause toxic and unwanted reactions toward the biological and hematological indexes [5,6,7,8].The following body organs were analyzed histomorphologicaly: brain,miocardium, lung, v. cava, liver, kidney and skeleton mussels in the place of injection.The method of hematoxilin-eozin was used. During the histo-morphologic investigation changes of the investigated organs due to the use of HI-6 were not detected .

The HI-6 – dichloride salt, have a low stability in water solutions. Having in mind this disadvantage we created a lyophilic injection form. She was described in BG Patent No 62589 [4].

The Toxidine was permitted for human use from Bulgarian Drug Agency (No 481 / 17/03/1988). In Bulgarian Army he was accepted for use in 1990. Unfortunately this antidote is not produced at the moment.

We preserve some ampules of Toxicidine produced in 1997 year (batch No 0396 /1997).

N O ClNN

OHO ClCl

N

OH

Cldichloro dimethylether

intermediate

pyridine-2-aldoxime

OH

N

H2N O

N O

2Cl

N O ClN

OH

Cl N

O NH2

N

iso-nicotineamide

intermediatetoxidine

Page 432: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

430

The aim of this study was to investigate the stability and purity of HI-6 from old ampules. For this purpose to develope a new HPLC method.

Materials and methods

ChemistryAcetonitrile gradient-grade and all other chemicals and solvents (analytical grade)

were purchased from Sigma Chemical Co. (USA) or Merck (Germany).In all experiments deionized water was used (18.2 МΩ.cm).

HPLC analysis HPLC analysis was carried on Gilson (Villers-le-Bel, France) consisting of a 305 master

pump, a 306 slave pump, an 805 manometric module and an 811C dynamic mixer with Spectrofl ow 757 UV detector (Applied Bio-systems, Ramsey, NJ). Toxidine (20μL, 100 μg/mL in mobile phase) was analyzed on Alltech RP C18 column (10 μm, 250 х 4.6 mm, Alltech, USA) using 5 mM sodium 1-octanesulfonate in triethylamine-phosphate buffer (50 mM, pH 3.0):acetonitrile (80:20) as mobile phase and UV detection at 215 nm[9,10]. The column fl ow was set to 1.0 mL/min. The quantifi cation of purity of compound toxidine was calculated as a percentage of the total amount of the peaks area.

Results and discussion

HPLC analysis Toxidine was dissolved immediately prior to analysis due to its low stability in solution.

Initial reagents (pyridine-2-aldoxime, intermediate and iso-nicotine amide) used in the synthetic stages were characterized as previously described. The purity of the intermediate obtained in the fi rst stage was 80%. The percent purity of the fi nal product (toxidine) was 98%. The chromatogram is shown on Fig. 2.

Under the same chromatographic conditions lyophilized toxidine was analyzed in 20-years old samples stored at different conditions (+4 oC and room temperature). The chromatograms obtained are presented on Fig. 3. There is practically no difference in the purity of the toxidine ampules stored at different temperatures. The HPLC results are summarized in Table 1.

ConclusionNew modifi ed HPLC method for determination of HI-6 was developed. Investigation

of the old ampules (20 years) of Toxidin, showed that the compound is the same as newly synthesized one. Our results demonstrated that in this lyophilic form, HI-6 is very stable.

Table 1. The purity of toxidine, newly synthesized, and from ampules stored at different temperatures.

Compound and storage condition Purity (%) toxidine Impurities Toxidine, newly synthesized 98 2% intermediate

Toxidine, lyophilized, ampule, stored at room temperature 96

1% iso-nicotine amide1% pyridine-2-aldoxime

2% intermediate Toxidine, lyophilized, ampule, stored at +4 oC 98 2% intermediate

Page 433: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

431

Fig. 2. Chromatogram of newly synthesized toxidine.

A.

Page 434: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

432

B.

Fig. 3. Chromatogram of: A. lyophilized toxidine in ampule stored at +4 °C,

B. lyophilized toxidine in ampule stored at room temperature.

References:

1. Hagedorn, L., Bis-quaternary pyridinsalze, Deut.Pat. 773 775, 10702. Oldiges, H. and Schoene, K., Pyrydinium und Imidozolium-salze als antidote gegenuber soman und

Paraoxon vegiftungen bei Mausen, Arch. Toxicol., 26, 293,1070.3. Mishova N., I. Petrova, E. Manolov, C. Dishovsky, Bg Patent № 52193, Reg. № 65392/08.05.1984.4. Dishovsky C., I. Petrova, E. Slavova, D. Popova, Bg Patent № 62589, Reg. №101150/20.01.1997.5. Dishovsky C., Doctor of Science Work, MMA, Sofi a, 1989, (in Bulgarian).6. Dishovsky C., “Reactivators of Cholinesterase”, SA, Sofi a, 1990 (in Bulgarian).7. Dishovsky, C., Pharmacology and Toxicology of HI-6, Proceedings of the Symposium on Defensive

Aspects of Chemical and Biological Warfare, 3-6 September 2002, NATO Studies, Analysis and Simulation Panel, Delft, The Neterlands, 2002.

8. Dishovsky, C., Pharmacology and Toxicology of Oxime Reactivators of Cholinesterase, Toxicology Letters, v., 144, Suppl., 1, 2003, s 90.

9. I. Petrova, I. Samnaliev, C. Dishovsky, Synthesis of new reactivators of cholinesterase, In: Medical Management of Chemical and Biological Casualties, (Eds: St. Tonev, K. Kanev, Chr. Dishovsky), IRITA, Sofi a, 2009, 115 – 120.

10. H.Singh, D. Moorad-Doctor, R. H. Ratcliffe, K.Wachtel, A.Castillo and G.E. Garcia, A rapid cation-exchange HPLC method for detection and quantifi cation of pyridinium oximes in plasma and tissue, Journal of Analytical Toxicology, vol. 31, 2007, 69 – 74.

Page 435: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

433

Chapter 60

Mycotoxins: Novel Approaches for Biological Threat Mitigation

Prerna AGARWAL1, Rajesh ARORA1, 2*, R. CHAWLA1, D. GUPTA 1, A. ZHELEVA3, V. GADJEVA3, Stoycho STOEV4

1Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization (DRDO), New Delhi, India,

2Offi ce of the Director General-Life Sciences, Defence Research and Development Organization Hqrs, Room No. 339, DRDO Bhawan, New Delhi-110011, India

3Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria

4Department of General and Clinical Pathology, Faculty of Veterinary Medicine, Trakia University, Stara Zagora, Bulgaria (currently Visiting Professor in t

he Faculty of Science, University of Johannesburg, South Africa)*Corresponding Author’s email: [email protected]

Abstract. This chapter presents an overview of the natural toxins with special reference to the toxins produced by fungi called “ mycotoxins”. Mycotoxins are biologically originated toxins, produced by fungi that infect agricultural crops during their growth, drying, storage and transportation. When taken in diet, they are capable of producing toxic effects in both human and animals. Due to their potency, their devastating biological effects (such as carcinomas), and their production by a wide number of different fungal species, the possibility of their use as biological warfare agents exists. The main mycotoxins of interest at present are afl atoxins and ochratoxins. Various techniques have been employed for the detection and purifi cation of these mycotoxins and biosensors developed for their detection. In consequence to the food safety issue by these mycotoxins, EU has established various regulations regarding their control in food and feedstuffs. The chemical diversity of mycotoxins and the diversity of substrates in which they occur, pose challenges to formulation of detoxifi cation agents (chemical and herbal). The present review defi nes precisely subcategories of mycotoxin-adsorbing and biotransforming herbal agents, based on their functional properties, their mode of action and their biological or physical and chemical characteristics, mainly for protection against these mycotoxins.

Keywords. Mycotoxins, biological warfare, biosensors, masked mycotoxins, health risk assessment, chemical and herbal antidotes.

1. IntroductionScientifi c interest in natural toxins is progressively increasing as they often have a

strong cytotoxic, carcinogenic or mutagenic potential and can have devastating effects on human health. Mycotoxins are usually discovered in the cases of sudden occurrence of

Page 436: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

434

liver toxicities or carcinomas in poultry, animals or humans of a particular location. On the basis of their origin, the natural toxins can be classifi ed as (Figure1):

Figure 1. Types of natural toxins

According to WHO, mycotoxins pose major food safety issues followed by bacterial toxins, phytotoxins and other toxins. Mycotoxins are an important group of naturally occurring chemical substances and can be found in fairly large concentrations in human diet consisted from contaminated food. The mycotoxin content in various foods and feeds presents a serious health problem for animal and humans in many developing- as well as developed countries in all over the world (Stoev, 2013). According to a report of FAO (Food Agricultural Organization), 25% of the world’s annual crop production is contaminated by mycotoxins (Rice and Ross 1994). Fungi can either cause mycoses (due to exposure to various fungal species) or mycotoxicoses (due to exposure to the secondary metabolites or toxins of the same fungi).

1.1 What are Mycotoxins?

Mycotoxins are secondary metabolites produced by certain moulds such as Aspergillus, Fusarium and Penicillium etc. in agricultural products at specifi c environmental conditions and can be a major health hazard. Due to its unavoidable and unpredictable nature, mycotoxin contamination presents a unique challenge to food safety. The term “ Mycotoxins” describes pharmacologically active mould metabolites, characterized by vertebrate toxicity. More than 300 secondary metabolites have been identifi ed, but only about 30 have been found to be toxic (Figure 2).9

Mycotoxins fall into several chemically unrelated classes that are produced in a strain-specifi c manner and depict some complicated toxic activities in sensitive species including carcinogenicity, inhibition of protein synthesis, immunosuppression, dermal irritation and

Page 437: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

435

other metabolic perturbations. Entry routes for mycotoxins generally include ingestion of contaminated food, inhalation of toxic spores and direct dermal contact.

Figure 2. Levels of Mycotoxin Accumulation

1.2. Historical Perspective

The evolution of mycotoxins dates back to 1962, when during a veterinary crises in England, London about 100,000 Turkey poultry died (Money 2002). This disease was named as Turkey X disease. It was observed that the Turkey X disease is caused by one of the secondary metabolites today known as afl atoxin presented in contaminated peanut meal. Later on, other mycotoxins were also discovered, eg. ergot alkaloids, patulin (originally discovered as an antibiotic), Ochratoxins, etc. The golden period in mycotoxicological research was the period between 1960 and 1975 and is known as the Mycotoxin Gold Rush (Maggon et al. 1977) because several groups of scientists joined together to discover other mycotoxins as well. Table 1 shows various mycotoxins and the diseases associated with them.

Table 1. Epidemiology of diseases associated with different Mycotoxins

S.NO Mycotoxin Disease Causative Organism Affected Organism Year Location

1 Ergot alkaloid Ergotism Claviceps pupurea Human, Cattles

17621966

Europe, France

2 T-2Diaceto-xyscirpeno

Aleykiya Fusarium sporotrichioidesFusarium poae

Human, cattle, horse, pig, dog

19131942

Former U.S.S.R

Page 438: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

436

3 Fumonisin B1 Leukoencephalomalacia and porcine pulmonary oedema

Fusariummoniliforme (F. verticillioides)

Swine, horse,

1928 United States

4 Luteoskyrin Cyclochlorotine

Yellowed rice Toxicosis Pyrobaculum islandicum

Chicken 1694 USA, Europe,Japan

5 Afl atoxins Turkey X Aspergillus fl avus Turkey, trout, dog

1960 UK, USA

6 Patulin Hyperkeratosis Aspergillus clavatusPenicillium urticae

Calf 19531961

S.Africa, USA

7 Penicillic acid Hepatitis X P. aurantiogriseum complex Propionibacterium rubrumAspergillus ochraceus

Dog, swine

1952 USAJapan,S.Africa

8 Ochratoxin AFumonisin B1

MycotoxicNephropathy

P. verrucosum,A. ochraceus,F. verticilioides

Pigs, chicks

1968 2010

Scandi-navianCountriesBalkans

9 Fumonisin B1 Equine leukoencephalomalacia, Porcine Pulmonary Edema, Oesophageal cancer, Neural tube defects in progeny

Fusarium moniliforme/. Verticillioides, F. proliferatum

Horse, Pigs, Human

1976 1988

USA

10 Zearalenone Vulvovaginitis, Vaginal/rectal prolapse, Swelling of mammary glands, Testicular / ovary atrophy

Fusarium graminearum/. Fusarium culmorum

Pigs of 3-5 months

1998 USA

11 Satratoxin, Verrucarin, Roridin

Mouth and gastrointestinal damages, symmetrical trophic necroses, haemorrhages

Stachybotris alternans/altra

Horse, Ruminants, Pigs, Chicks

1998 USA

1.3. Importance of Mycotoxin ResearchTwo aspects of mycotoxin research that have received the extensive coverage in

the recent past are: (i) their possible use as agents of biological warfare and (ii) their potential involvement in various health hazards, recent one includes building related disease conditions (viz. Sick- Building Syndrome) (Dales 1991 and Miller 1992 ). Biological weapons pose a great threat from military and civilian prospective in today’s scenario as the non-state actions are fast acquiring techniques to manufacture bioweapons, several mycotoxins pose great concern. Hence, it is becoming increasingly important to consider different strategies for preventing and preparing for attacks by enemies. The most important toxins are Afl atoxins and Ochratoxins produced mainly by Aspergillus and Penicillium species. During 1980, was reported that the Iraqi scientists used afl atoxin as a part of their prepared bioweapons (Stone 2001). The use of Trichothecens in Biowar by the Soviet Union on attacking Hmong Tribesman in Laos and Kampuchea has been reported in 1981. Haemorrhaging, blistering of skin and other clinical responses were observed in the victims. Economic losses of hundreds of millions of dollars each year have been reported. Masked mycotoxins, the derivatives of mycotoxins, are undetectable by conventional

Page 439: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

437

analytical techniques (their structure changes in due to internal characteristics of plants) and they pose a great threat.

2. Classifi cation of mycotoxins and their health effectsThe main mycotoxins include:� Afl atoxins� Ochratoxins� Fumonisins� Citrinin� Ergot alkaloids� Patulin� Zearalenone� TricothecenesThe presence of mycotoxins in grain and other staple foods and feedstuffs has serious

implications for human and animal health. The risk of mycotoxins varies in accordance with continent, country, region, age, sex etc (Rodricks et al. 1977). Exposure to mycotoxins can produce both acute and chronic toxicities ranging from death to various deleterious effects. Mycotoxicoses in animals is more common than in humans. The most common mycotoxicoses in animals include ergotism, various kinds of fusariotoxicoses, afl atoxicosis, mycotoxic nephropathy, stachybotryotoxicosis, equine leukoencephalomalacia, and many others (Stoev, 2007). The commonly encountered diseases include nephropathies in human and swine. It has been reported that Ochratoxin A, in synergism with other mycotoxins, is the major cause of these nephropathies. Their complicated and overlapping toxigenic activities results in serious health concern including carcinogenicity, teratogenicity, neurotoxicity, nephrotoxicity, hepatotoxicity, strong immunosuppression, etc. It is reported that mycotoxicoses or exposure of toxins in humans can be of two types acute or chronic. Mycotoxins cause toxicity at various levels like molecular, subcellular, cellular and extracellular level. At molecular level, they interact with macromolecules like proteins, DNA, RNA. At sub cellular and cellular level they interact with enzymatic reactions, various cellular organelles, and alter cell metabolism. Whereas at the extracellular level their effect on various tissues and organs have been reported. Some of the health effects of various types of mycotoxins are elaborated in Table 2.

3. Screening Techniques

3.1. Methods for Detection of MycotoxinsAs the numbers of mycotoxins present in food varies up to 100, it is virtually very

diffi cult to detect accurately each one of them, hence rapid and reliable methods for recognizing fungal growth in food or plants are needed. Earlier viable counting methods were used like but these have drawbacks that through them only viable mycelium can be fi ltered and the mycotoxins are still retained. Hence non- microbiological methods are more promising these days. Some of the methods used for the detection of fungi-producing mycotoxins are summarized in Table 3.

Page 440: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

438

Table 1. Mycotoxins: Types and Characteristics

S.No. Mycotoxin Type Acronym Species Producing Susceptible substrates

Possible health effects

1. Afl atoxin BI AFB1 Aspergillus fl avus, Aspergillus arachidicola, A. bombycis, A. minisclerotigenes, A. nomius, A. ochraceoroseus, A. parasiticus.

Cereals, bakery, feed, fruits and vegetables, herbs and spices, nuts, seed and others

Mutagenic, carcinogenic and heptotoxic

B2 AFB2 A. fl avus, A. arachidicola, A. bombycis, A. minisclerotigenes, A. nomius, A. ochraceoroseus.

GI A. fl avus, A. arachidicola, A. bombycis, A. minisclerotigenes, A. nomius, A. ochraceoroseus

Cereals, bakery, feed, fruits and vegetables, herbs and spices, nuts, seed and others

Mutagenic, carcinogenic and heptotoxic

G2 A. parasiticus, A. pseudotamarii, A. rambellii, Emericella venezuelensis

2. Tenuazonic acid TeA Alternaria alternata Crops Neurotoxic, Nephrotoxic, Hemorrhagic

4. Fumonisins B1 FB1 Fusarium section liseola

Cereals and bakery

Neurotoxic, Nephrotoxic and Carcinogenic

B2 FB2

5. Ochratoxin A OTA Aspergillus section Circumdati and many Penicillium species

Cereals, bakery, feed, fruits and vegetables, herbs and spices and others

Nephrotoxic, teratogenic, Heptotoxic, Carcinogenic, immunogenic, causes nephropathies and urinary tumours both in animals and humans.

6. Patulin PAT Penicillium expansum, Bysochlamis nivea, Aspergillus clavatus

Unfermented foods products

Acute toxicity

7. HT-2 and T-2 toxins

HT-2 Fusarium acuminatum, F.poae

Cereals Vomiting, Nausea, Immunosuppression

T-2 F. sporotrichioides, F. langsethiae

8. Deoxynivanlenol DON Fusarium graminarearum, F.culmorum, F.cerealis

Grains Vomiting, Nausea, Diarrhoea

9. Zearalenone ZEN Fusarium graminarearum (F. roseum), F. culmorum, F. equiseti, F. cerealis, F. vertcilloides, F. incaratum

Cereals, bakery, feed, fruits and vegetables

Hyper estrogenic

Page 441: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

439

Table 2. Methods for detection of Mycotoxins (Rai et al 2012)

S.No. Methods Techniques1. Immunological • Chemical e.g. Chitrin assay, Ergosterol assay

• Enzymatic e.g. ATP assay• Metabolic e.g. Impediometry• Immunochemical–e.g. EPS antibodies, MLA, Sandwich ELISA• Electrochemical immunoassays

2. Analytical • Thin layer chromatography (TLC)• High Performance Liquid Chromatography ( HPLC)• Gas Chromatography (GC)• Capillary Electrophoresis (CE)• Mass Spectroscopy (MS)• Ambient Ionization Mass Spectrometry• Thin layer chromatography (TLC)• High Performance Liquid Chromatography

3. Molecular • cDNA approach• Polymerase chain reaction • Molecular Imprinting

4. Recent • Lateral Flow Immunochromatography

• Fluorescence Polarization Immunoassays• Electronic noses• Biosensors• Piezoelectric Sensors

The analysis of mycotoxins in different foodstuff is an issue of serious concern. The analysis of mycotoxins in different foodstuffs depends on the matrix in which they are present. Chromatographic columns are widely employed for the purifi cation of different mycotoxins. The latest purifi cation technique is based on immunoaffi nity clean-up. These columns are made up of monoclonal antibodies, which are specifi c for the toxins to be extracted: the antibodies are immobilised on Sepharose. Recently, immunological and molecular detectors have also been developed like recombinant fragments of antibodies, DNA Aptamers, or molecular imprinted polymers (MIPs), for high performance analysis system. Various analytical methods for Mycotoxins have been recently published (van Egmond 2004).

3.2. Biosensors for Detection of MycotoxinsBiosensors are an effi cient method for the detection of mycotoxins because of their

characteristics such as (i) simplicity (ii) easy use (iii) high selectivity (iv) low cost (v) their integration with automated and portable devices. Since some of the mycotoxins are electroactive, therefore, they can be effectively employed for rapid detection. For mycotoxins like ZEA, direct amperometric determination can be made (Duca et al. 2010). But in some cases like Afl atoxin B1, Afl atoxin B2 and DON, adsorptive accumulation by stripping voltammetry procedure is effective in the detection in trace quantities. Enzyme sensors to mycotoxins have also been designed because of their catalytic and inhibitory characteristics e.g. for detection of AFB1 levels in food, AChE- based sensors have been

Page 442: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

440

developed. Similarly for the detection of Ochratoxin A (OTA), horseradish peroxidase enzyme sensor-based systems have been developed and for both Afl atoxins and Ochratoxins Quartz Crystal Microbalance (QCM) has been used. Surface Plasmon Resonance (SPR)-based biosensors, Total Internal Refl ection Ellipsometry (TIRE) and FLD detection are some of the recently emerged biosensors (Camp et al. 2012).

4. Detoxifi cation of mycotoxins

Preventive measures have been taken in the US and other countires to prevent the penetration of fungus in growing crops and hence subsequent toxin formation (Council for Agricultural Science & Technology 1989).

• Production of fungal resistant breeds.• Adaptation of proper agricultural methods (eg. Irrigation, crop rotation etc.).• Harvesting at maturity• Proper drying after harvesting• Use of chemicals to control fungal growth Such chemicals should be used which does not affects the nutritional value of treated

crops

4.1. Physical and Chemical DegradationMany measures have been adopted by different countries to prevent the production

of mycotoxins on the food commodities, but they fail to provide complete protection from mycotoxins. Therefore various physical and chemical methods can be used for the degradation of mycotoxins. Physical methods which have been used involves heating, irradiation, solvent extraction and absorption whereas, chemicals which have been used for mycotoxins degradation in crops includes ammoniation, acid-base method, oxidizing agents, bi-sulphite and vitamin-C treatments. Ammoniation has extensively been studied. The products of ammoniated crops are only restricted to animal feed.

4.2. Biological Detoxifi cation of Mycotoxins in CropsThis approach have been widely studied but is not practical acceptable. Several

microorganisms have been used to degrade mycotoxins (Ciegler et al 1966).e.g. Flavobacterium aurantiacum, is able to degrade Afl atoxins B1, G1 & M1. Aspergillus species effectively degrades afl atoxins B1.

5. Risk Assessment and food safety issues with special reference to EU

There are two main compounds taken into account in risk assessment: the toxicologic effect of particular mycotoxin and an estimate of the mycotoxin exposure of the consumer. If the Risk Assessment of a particular mycotoxin suggests a signifi cant risk to the consumer, setting maximum permissible limits presents a powerful tool for its effective control in the food chain (Rosner, 1998). As far as the effective control of these Mycotoxins is concerned, developed countries have administered better monitoring facilities than the developing ones. An integrated approach to food safety, known as Hazard Analysis and Critical Control Point (HACCP) can be applied for proper management of the hazards present in

Page 443: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

441

food and feedstuff (Stoev, 2007). Various organizations like World Health Organisation (WHO) and Health Organisation’s Joint Expert Committee on Food Additives (JECFA) and the European Community’s Scientifi c Committee for Food (SCF), have assessed the risk from mycotoxins and have advocated controls to reduce consumer exposure. JECFA has given a mechanism for assessing the risk associated with contaminants ( mycotoxins). The toxicological evaluation carried out by JECFA normally results in the estimation of a Provisional Tolerable Weekly Intake (PTWI). The evaluation is based on the determination of no-observed-effect-level (NOEL) in toxicological studies, and the subsequent application of a safety factor. It is important to know, that this approach for establishing maximum tolerated levels of mycotoxins in foods does not apply for toxins where carcinogenicity is the basis for concern as is the case with afl atoxins (Stoev, 2013). In these cases the necessary approach must be “as low as reasonably achievable” (ALARA principle) or as low as possible but technologically feasible and analytically detectable in the food ready for consumption (Rosner, 1998). Therefore, International Agency for Research on Cancer (IARC) has a programme for the evaluation of carcinogenic effects of some mycotoxins on humans and according to it, mycotoxins can be distributed in group 1 (carcinogenic to humans as afl atoxins), group 2A (probably carcinogenic to humans), group 2B (possibly carcinogenic to humans as fumonisins or ochratoxin A), group 3 (not classifi able as to its carcinogenicity to humans) (Castegnaro and McGregor, 1998). Periodically, some revisions of the criteria for this classifi cation and reclassifi cation of some mycotoxins are made by IARC. The report of JECFA has in mind the mycotoxin absorption, toxicity, health hazards and control (World Health Organization 2002). There are some organizations in Europe which are involved in risk assessment of mycotoxins (EC 2003), listed in Table 4.

Table 3. Some of the European organisations associated with risk assessment

S.No.. Head body Committee Object1. European Committee Scientifi c Committee on Food

(SCF) Scientifi c Committee on Animal Nutrition (SCAN)

Occurrence of mycotoxins in food or animal feeds.Risk associated with the occurrence.

2. European Food Safety Authority (EFSA)

Scientifi c Panel on Contaminants (SPC)

Other issues related to mycotoxins

3. European Committee Scientifi c Co-operation on Questions Relating to Food Projects

Risk associated with dietary mycotoxins

4. European Committee Quality of Life Program (QLP) Risk associated Ochratoxins

5. --- European Branch of the International Life Science Institute

Associated with nutrition, food safety, toxicology and environmental risk

On the other hand, the question of establishing maximum levels of mycotoxins and other chemical contaminants in foods and various commodities is considered by the Codex Committee on Food Additives and Contaminants (CCFAC) in consultation with the Codex Commodity Committee, but the fi nal decision is usually taken by the Codex Alimentarius Commission (CAC). The conclusions are based on the scientifi c evaluation made by JECFA and other relevant expert meetings (Boutrif and Canet, 1998).

Page 444: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

442

The EU has set some values to limit the daily intake of each mycotoxin. The values are known as TDI or tolerated daily intake. Different mycotoxins have different TDI values. As Afl atoxin B1 is the most toxic among all so no limit is set for it. The TDI values for different mycotoxins are given in Table 5.

Table 4. TDI values of mycotoxins (EC act 2006)

S.No. Mycotoxins TDI values per kg body wt per day (kg/bw/d)

1. Afl atoxin 0.01-0.1 μg/kg2. Ochratoxin 14 ng/kg 3. Patulin 0.4 μg/kg 4. Fumonisins 2μg/kg 5. Trichothecenes (DON, T-2, HT-2 toxins) 1 μg/kg6. Zearalenone 0.2 μg/kg

6. Development of Antidotes to mycotoxins

The foregoing discussion highlights the need to develop specifi c antidotes against mycotoxins that minimize the health impacts on both animals and humans. Approaches -detoxifi cation of mycotoxins contamination have included chemical and biological treatments, depending on their mode of action. The development of antidotes for detoxifi cation of mycotoxins faces many challenges: (i) no specifi c antidotes have been developed for the different mycotoxins (ii) synergistic effects of other mycotoxins present in the matrices (iii) complex molecular structures of toxins and their respective lethal concentrations (iv) masked- mycotoxins are an emerging concern and one of the greatest challenge being faced during development of an antidote. (Gareis et al. 1990). Mycotoxin detoxifi cation could be of two types: (i) Chemical and (ii) Natural. Herbal antidotes have an advantage over chemical ones as they nourish the body and augment immunity also by supporting various body functions (Treadway 1998).

6.1. Mycotoxin-adsorbing agents/ Chemical AntidotesMycotoxin-adsorbing agents are large molecular weight compounds that bind to the

mycotoxins in contaminated feed without dissociating in the gastrointestinal tract. In this way the toxin-adsorbing agent complex passes through the body and is eliminated via the faeces. This prevents or minimizes exposure of animals to mycotoxins. The chemical adsorbents/ agents used for the detoxifi cation of mycotoxins are highlighted in Figure 3

6.2. Natural Antidote/ Herbal DrugsAround 3000 plant species have been reported are used as therapeutic agents and

hence, can be formulated as an antidote against mycotoxins. Different preparations made from a plant or plants can be used to prevent and treat diseases and ailments or to promote health and healing. These biological ingredients extracted from natural substances result in multiple effective actions on different biological molecular targets; hence various plants can be used against different targets. As reported, some “herbal drugs” are well known for their effects against hepatocellular damages and are used for treating different liver diseases,

Page 445: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

443

and can possibly be used for the protection against the toxic effect of different mycotoxins. Some plants e.g. Andrographis paniculata, Annona atemoya, Picsoshiza kurroa, Silybum marianum Phyllanthus niruri/amarus, Piper longum, Podophyllum hexandrum, Tinospora cordifolia, Semecarpus anacardium etc. hold promise in this regard. Regarding the parameters to be used for assessing the effi cacy of herbal antidotes, there is much that needs to be done and endeavours are being made in this direction to bioprospect such agents from indigenous sources, based on literature search, ethnopharmacological leads and bioinformatics approaches. The herbal formulations being tested against mycotoxins work on different biochemical pathways and affect several organ systems simultaneously, yielding holistic effects.

7. ConclusionThe effi cacy of the antidotes is unknown due to complexity in their development process

and incomplete pre- clinical trials. Hence, future studies and research in the described fi eld can help to formulate herbal antidotes against various mycotoxins. The herbal antidotes are safe and effective. Both in vivo and in vitro studies are underway to develop chemical adsorbent/herbal agent-based formulations for detoxifi cation of mycotoxins. Further studies are also on-going to provide effi cient and reliable drugs to reduce the toxicity of mycotoxins in vivo at minimal cost. Such antidotes would be a boon for boosting both animal and human health, particularly from a biothreat mitigation perspective.

8. AcknowledgmentsThe authors would like to thank the European Union for grant of the project

entitled “Studies on some herbal additives giving partial protection against toxic or immunosuppressive effects of some mycotoxins and improving wound granulation” (Grant Agreement Number: PIRSES-GA-2012-316067). Under the aegis of Marie Curie Actions People International Research Staff Exchange Scheme (Seventh Framework Programme). The authors also wish to express their gratitude to Dr. Manas K. Mandal, Director General-Life Sciences, DRDO and Director, INMAS for providing research facilities and support.

Figure 3. Agents which may possibly reduce the impact of mycotoxins in feedstuffs

• AFM1

• Fumonising

• OTA

• FB1, FB2

• T-2 toxin and ZEA

Page 446: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

444

REFERENCES:

Boutrif E., Canet C. Mycotoxin prevention and control: FAO programes. // Revue Med Vet., 149, 1998,p.681-694.Camp M., Gariboa D., Prieto-Simlonb B. Novel nanobiotechnological concepts in electrochemical biosensors for the analysis of toxins. // Analyst., 137, 2012, p. 1055-1067.Castegnaro M., Gregor M. . Carcinogenic risk assessment of mycotoxins. // Revue Med Vet., 149, 1998,p. 671-678.Ciegler A., Lillehoj EB., Paterson RE., Hall HH. Microbial detoxifi cation of afl atoxin.// Applied Microbiology, 14, 1966, p .934-9.Commission Regulation (EC) 1881. Setting maximum levels for certain contaminants in foodstuffs. // Offi cial Journal of the European Union, L364, 5e24, 19 December 2006.Dales RE., Burnett R., Zwaneburg H. Adverse health effects among adult exposed to home dampness and molds. // Am. Rev. Respir. Dis., 143, 1991, p. 505–509.European Commission. Mycotoxins In: Opinion of the Scientifi c Committee on Animal Nutrition on undesirable substances in feed. // European Commission, Health& Consumer Protection Directorate-General, Brussels, 2013, p 6.Food and Agricultural Organization of the United Nations food and nutrition. Worldwide regulations for mycotoxins, A compendium. // Food and Agricultural Organization, Rome, Italy., paper 4, 1997.Food and Agriculture Organization. Safety evaluation of certain mycotoxins in food. // Fifty-sixth meeting of the Joint FAO/WHO Expert Committee on Food Additives (JECFA), FAO Food and Nutrition., Food and Agriculture Organization of the United Nations, Rome., paper74, 2001.Gareis M., Bauer J., Thiem J., Plank G., Grabley S., Gedek B. Cleavage of Zearalenone-Glycoside, a “Masked” Mycotoxin, during Digestion in Swine. // Journal of Veterinary Medicine, Series B37, 1990, p. 236–240.Krogh P., Hasselager E. Studies on fungal nephrotoxicity. // R. Vet. Agric. Coll. Yearbook, 1968, p. 198-214.Maggon KK., Gupta SK,. Venkitasubramanian TA. Biosynthesis of afl atoxins. // Bacteriol. Rev., 41, 1977, p. 822–855.Marasas, W.F.O., Kellerman, J.S., Pienaar, J.G., Naude, T.W. Leukoencephalomalacia: A mycotoxicosis of Equidae caused by Fusarium moniliforme Sheldon. Onderstepoort J Vet Res. 1976,43: 113-122.Marasas, W.F.O., Jaskiewics, K., Venter, F.S., van Schalkwyk, D.J. Fusarium moniliforme contamination of maize in oesophageal cancer areas in Transkei. S Afr Med J. 1988, 74: 110-114.Miller JD. Fungi as contaminants in indoor air. // Atmosph. Environ., 26, 1992, p. 2163–2172.Money N. The mysterious world of mushrooms, molds and mycologists. // Mr. Bloomfi eld’s orchard, Oxford University Press, New York, N.Y, 2002.Rai MK., Bonde SR., Ingle AP., Gade AK. Mycotoxin: Rapid detection, Differentiation and Safety. // J Pharm Educ Res., 3(1), 2012, p.22-34.Rice LG., Ross FB. Methods for detection and quantitation of fumonisins in corn, cereal products and animal excreta. // J. Food Protect., 57 (5), 1994, p. 36–40. Rodricks JV., Hesseltine CW., Mehlman MA. Mycotoxins in human and animal health. // Proceedings of a conference convened at University of Maryland, October 4-8, 1976.Rosner H. Mycotoxin Regulations: an Update. // Revue Med Vet., 149, 1998,p. 679-680.Shier WT. Estrogenic mycotoxins. // Revue Med Vet., 149, 1998, p. 599-604.Stoev SD. Food safety and increasing hazard of mycotoxin occurrence in foods and feeds, // Critical Reviews in Food Science and Nutrition, 53 (9), 2013, p. 887-901.Stoev, S. Food Safety and Some Foodborne Mycotoxicoses.// Vet Africa 2007 Congress, 27–28 July, 2007, Johannesburg, South Africa.Treadway S. An Ayurvedic herbal approach to a healthy liver. // Clin Nutr Insights, 6, 1998, p. 1–3.Van Egmond HP. 2004. Natural toxins: risks, regulations and the analytical situation in Europe. Analytical and bioanalytical chemistry 378: 1152–1160.Weiss R., Fintelmann V., Weiss RF. Herbal medicine. // Georg Thieme Verlag, 2002.World Health Organization. Evaluation of certain mycotoxins in food. // Fifty-sixth report of the Joint FAO/WHO Expert Committee on Food Additives, WHO Technical Report Series 906. World Health Organization, Geneva, 2002.

Page 447: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

445

Chapter 61

Signifi cance of Butyrylcholinesterase Variants for Military Personnel

Dimo DIMOV, Kamen KANEV,Disaster medicine and toxicology department, Military Medical Academy – Sofi a, BULGARIA

Abstract. There arenumerous chemicals and drugs that interact with the cholinergic nervous system.: nerve agents, prophylactic antidotes, myorelaxants and other therapeutic drugs. The clinical signs of organophosphorus intoxication and effective treatment depend on the activity of the enzyme butyrylcholinesterase (BCHE). The variations in BCHE gene alter the enzyme activity and in this way effective drug treatment and prognosis of intoxications may therefore be infl uenced by the genotype of the individual. During the mission abroad military personnel may be exposed to a wide range of chemicals and drugs. In a military setting, genetic variation in BCHE might result in altered response to deployment related drugs such as prophylactic antidotes: pyridostigmine, physostigmine, heptyl physostigmine. This study has identifi ed the frequency of genetic variation in BCHE among Bulgarian military personnel. We suggest that our results will determine the servicemanwith increased sensitivity to a range of chemicals and drugs of interest to Defence.

Key words: Butyrylcholinesterase, military personnel, organophosphorus compounds

Introduction

Human butyrylcholinesterase (BCHE) is an enzyme found in plasma, liver, lungs, heart, brain and many other parts of the body. The enzyme hydrolyses drugs containing ester bonds such as drugs acting at the neuromuscular junction (succinylcholine), local anaesthetics (procaine, chloroprocaine and cocaine) and heroin [1].It is also a biological scavenger for organophosphorus and carbamate compounds.[2]. Organophosphorus compounds (OPs) are triesters of phosphoric acid and their major use is as insecticides. Chemical warfare agents that affect the nervous system and nerve agents (eg. soman and sarin) also belong to this class of chemicals.

In human, the butyrylcholinesterasegene (BCHE) is located on chromosome 3 (3q26.1–q26.2).[3]. The enzyme has normal BChE activity (8440±1780 IU/L for butyrylthiocholine used as substrate) in serum is called as usual BChE[4]. However, more than 40 genetic variants of BCHE have been described.[5]. These mutations in BChE gene cause the different BChE genotypes that have different levels of enzyme activity.

Page 448: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

446

Some of genetic polymorphisms are responsible for development of prolonged apnea. The rapid hydrolysis of succinylcholine by BCHE accounts for its short duration of action. Mutations in the BCHE gene affecting the function of this enzyme can lead to prolonged neuromuscular blockade by succinylcholine. The clinical signifi cance of genetic variants in butyrylcholinesterase gene (BCHE) in patients with a suspected prolonged duration of action of succinylcholine was demonstrated inrecent study. An eleven of 13 patients with a prolonged duration of action of succinylcholine had mutations in BCHE, indicating that this is the possible reason for aprolonged period of apnea.[6]. Military personnel have increased risk of traumatic injury. In these cases they might receive of succinylcholine as a myorelaxant for surgical procedure. Episodes of prolonged apnea can be avoided by preoperative BCHE phenotyping. Furthermore, military personnel are deployed to various locations in the worldand receive medications and vaccines different from the civilian populace or they might be exposed to organophosphate (OP) and/or carbamate cholinesterase inhibitors (AChEi). Individuals with low BuChE activity would be more sensitive to these compounds and drugs. This assumption prompted us to investigate the frequency of BChE phenotypes among Bulgarian military personnel and identify serviceman expected to have a problem in case of a surgical procedure (after application short muscle relaxants) or exposure of anticholinesterase agents.

Aim: The aim of the present study is to assess the frequency of BChE phenotypes among Bulgarian military personnel and identify individuals expected to have a problem in case of a surgical procedure (after application short muscle relaxants) or exposure of anticholinesterase agents.

Material and Methods:

SubjectsParticipants were Bulgarian Defence Force personnel who provided 10 ml of whole

blood as part of a larger medical screening process. The selected individuals were asymptomatic, with serum activity of hepatic enzymes ALT and AST in reference range. The survey was conducted on 542 blood samples obtained from voluntary donors. The serum was separated after centrifugation and stored at – 20 0 C before further use.

BChE activity measurement and determination of BChE phenotypeMeasurements were performed according to standard method using propionylthiocholne

as substrate. This method was introduced by Dietz and it is based on hydrolysis of thioesters by BChE derived from human serum. (7) The concentration of products of the hydrolysis is directly proportional to the BChE activity (UI/L at 370C) and can be detected by spectrophotometer. In the presence of inhibitors dibucaine and sodium fl uoride the intensity of produced color is changed and by this way can be distinguish phenotypes of BChE.

We used spectrophotometer Screen master with fl ow cuvette. Reagents for determination of BChE activity were propionylthiocholine, DNTB, buffer (TECO diagnostics). These reagents were diluted with 6 ml distilled water and the fi nal concentration of DNTB and propionylthiocholine were 0.4 mM and 4 mM, respectively. To 1 ml of this solution was added 0.01 ml undiluted human serum. The absorbance was measured by Screen Master at 405 nm for 90 sec and it corresponds to BChE activity in the international unit (UI).

Page 449: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

447

For determination of dibucaine number (DN) and fl uoride number (FN) we used the same reagents (propionylthiocholine, buffer and DNTB) diluted with 0.3 mmol/l dibucaine and 40 mmol/l sodium fl uoride, respectively. Then we added 0.01 ml undiluted serum to 1 ml of working solution and measured BChE activity. From the degree of inhibition DN and FN were calculated. The BChE phenotypes were determined by classifi cation based on Dietz et all. [7]. Phenotype frequencies were calculated using statistical methods as STDEV and t-test.

Results/ DiscussionIn our study we found the mean value of serum activity of normal BCHE phenotype

was 9040 ±2011 UI. The reference range of enzyme activity and percent of inhibition for other BCHE variants are shown in table 1.

Table 1. Reference value of BCHE activity and percent of inhibition (Dibucaine and Sodium Fluoride) in Bulgarian serviceman

BChE phenotype Enzyme activity UI/L DN % FN %

UU 9040 ±2011 81 ± 1.49 79.6 ±1.08

UA 5710 ±845 71.8 ± 1.78 80.5 ±1.47

UF 6675± 715 78.9 ±1.44 71.8 ±1.62

US 5245± 636 84.1 ±0.74 78.9 ±0.98

AA 2684 18.9 82.4

AK 3105± 346 58.7 ±1.95 80 ±0.3

AS 1381 20.7 83.1

In the survey we established that the frequency of normal BCHE phenotype among Bulgarian serviceman is 94.6% and it is comparable to results of civilian populace and other country in Balkan region. [8]. Other allele frequencies were calculated and were compared to published data.(table 2). Overall, results demonstrated phenotype frequencies similar to our previous study for Bulgarian population and other European countries. [9]. We did not detected homozygote variants BCHE FF and BCHE SS due to their extremely low frequency. But hypersensitive variants BCHE AA, BCHEAK and BCHEAS to succinylcholine were found in 1/185 of military personnel. Based on results obtained from this study, the frequency of abnormal BChE phenotypes was 5.4%.Also the data illustratethat 1/21 is the frequency of moderate sensitive individuals (BCHE UA, BCHE US and BCHE UA).

Characterization of inherited BChE variants by inhibitorbasedphenotyping is a well-described technique capable of identifying atypical, fl uoride, and silent alleles. The plasma enzyme butyrylcholinesterase (BChE) is of clinical interest because of the occurrence of genetic variants with decreased ability to hydrolyse, and therefore inactivate, muscle-relaxant drugs such as suxamethonium. Analysis of BChE involves the determination of both enzyme activity and biochemical phenotypes which are used to determine the risk of so-called ‘scoline apnoea’. The widespread use of organophosphorus compounds as

Page 450: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

448

insecticides and the frequent misuse of nerve agents in military confl icts or terrorist attacks emphasize the clinical relevance of BCHE variant in military personnel. Here, for the fi rst time, we present data for the distribution of BCHE phenotypes in members of Bulgarian Armed Forces. The results show that no signifi cant difference in allele frequencies between civilian citizens and military personnel. However, the differences with regard to military and civilian prophylaxis strategies and organization of medical support during mission abroad lead to necessity of Butyrylcholinesterase screening of military personnel.

Table 2. Frequency of BChE phenotypes in Bulgarian military personnel

BChE phenotype Number of individuals Frequency of BChE phenotype %UU 513 94.6

UA 13 2.4

UF 8 1.5

US 5 0.9

AA 1 0.18

AK 1 0.18

AS 1 0.18

ConclusionMilitary personnel with an abnormal genetic BCHE variant would potentially

experience prolonged action of succinylcholine. BChE phenotyping is a cheap and rapid method of obtaining prior knowledge of possible hypersensitivity in members of the Bulgarian Armed Forced to succinylcholine and various organophosphorus compounds.Determination of BChE activity and phenotype by the descrive above method is well suited to preliminary screening of military personnel before mission abroad.

References:

[1]. Girard, T. and C.H. Kindler, Pharmacogenetics and anaesthesiology. Current Pharmacogenomics,. 2(2): p. 119-135., 2004

[2]. Singh, S., M. Verma, C.O. Leelamma, K. Nain, R.C. Goel, N.K. Ganguly, and B.K. Sharma, Red cell acetyl cholinesterase and plasma cholinesterase activity and genetic variants of plasma cholinesterase in Northwest Indian adults. International Journal of Clinical Pharmacology and Therapeutics,. 35(9): p. 357-360., 1997

[3]. Gaughan G, Park H, Priddle J, Craig I, Craig S: Refi nement of the localizationof human butyrylcholinesterase to chromosome 3q26.1-q26.2 using a PCR-derivedprobe. Genomics; 11:455–8, 1991

[4]. Melih O. Babaoglu Ж Turgay Ocal Ж Banu Bayar S. Oguz Kayaalp Ж Atila Bozkurt, Frequency and enzyme activity of the butyrylcholinesteraseK-variant in a Turkish population, Eur J Clin Pharmacol 59: 875–877, 2004

[5]. Lockridge O, Masson P: Pesticides and susceptible populations: People withbutyrylcholinesterase genetic variants may be at risk. Neurotoxicology, 21:113–26; 2000

[6]. Mollerup HM, Gдtke MR., Butyrylcholinesterase gene mutations in patients with prolonged apnea after succinylcholine for electroconvulsive therapy, Acta Anaesthesiol Scand. Jan;55(1):82-6., 2011

[7]. Dietz A.A., Rubinstein H.M., Lubrano T., Colorimetric determination of serum cholinesterase and its genetic variants by the propionylthiocholine-dithiobis(nitrobenzoic acid)procedure, Clin Chem.;19(11):1309-13, 1973

[8]. Lajtman Z, Surina B, Bartolek D, Car D, Gasparović S, Kirincić N, Rudez J. “Warning card” as a prevention of prolonged apnea in children, Coll Antropol., 26 Suppl:129-37, 2002

[9]. Whittaker M., Cholinesterase monographs in human genetics, In Backman Karger, vol. 11, p. 1-90, 1986

Page 451: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

449

Chapter 62

Antidotes – the Role of the Pharmacy in Medical Response in Crisis

Kanev K., Paskalev K.,Chobanov N.,Kolev S.Department of Toxicology and Disaster Medicine,

Military Medical Academy-Sofi a,Bulgaria

Abstract: Antidotes are an important element of the medical support in disasters with chemical weapons (CW) involved in. The treatment of victims requires antidotes as lifesaving medicines especially caused by CW. Therefore it is necessary to know what kinds of antidotes are available in the Bulgarian Healthcare System, especially after the terrorist act in Bulgaria. At present are available only 15 % of the specifi c antidotes according to WHO’s nomenclature. With these resources the crisis will be diffi cult to solve. Therefore we fi nd useful the American model for delivery of medicinal products because of its practicality and easy reproduction. It was found that if the names of antidotes regulated by Law on Medicinal Products in Human Medicine are used accurately, they will be explicitly requested and if needed easily and quickly delivered.

Key words: antidotes, medical, crises

Introduction

In recent years, the antidotes together with the other two groups of medicinal products -antibiotics and vaccines have become important issue for the pharmaceutical support during crisis.

This has become especially actual after the terrorist attack in the Tokyo subway on March 20, 1995, and few years later in the theater “Dubrovka” in Moscow on the October 23, 2002, and most importantly, for Bulgaria in 18 July 2012, when the country became the scene of an act of terrorism. These facts confi rm that a terrorist act can occur at any time and any place.

Aim1. To present and compare the model of national health legislation for pharmaceutical

support, in this case antidotes for the healthcare system of the Republic of Bulgaria especially in disaster situation caused by the use of Chemical Weapons (CW), with the analogical American model.

2. To analyze the actual availability of antidotes in Bulgaria and to express our view on the pharmaceutical support during a crisis.

Page 452: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

450

MethodLogical analysis of national Bulgarian Health Laws, and the relevant topics published

in thenational and international literature.

ResultsIn the recent years, the use of chemical weapons (CW) as a weapon for terror has

increased. This threat requires that the healthcare system must be prepared to respond adequately in case of possible use of CW. In case like this the pharmaceutical support plays an important role. In this study we focused our attention only on specifi c antidotes. These are life saving medical products used in case of intoxication, especially in cases caused by chemical warfare agents.

In Republic Bulgaria the Law for Medical Products regulates the manufacture, registration, import and distribution of these products, including antidotes, and also defi nes the pharmaceutical support in disaster situations. So this, in fact is the current framework for the pharmaceutical support in the country.

According to the offi cial classifi cation of the medicalproducts adopted by the Law for Medical Products of the Republic of Bulgaria, which is based on the Anatomic Therapeutic Chemical classifi cation (ATC), the WHO, antidotes are pharmacological subgroup within the 14th group of “V Various”. These products are classifi ed with ATC code V03AB, which categorised this group of medicines. According to the Bulgarian Drug Agency (BDA) in the country are registered only 5 medicinal products in this group, while the recommended by WHO in this group are included 30 medicines.(Table1). This means that in Bulgaria are offi cially registered only 15% of all specifi c antidotes.(Table 2).

The health system in Bulgaria completelylacksof specifi c antidotes for some kindsof intoxication because they are not produced in the country, and also, there are only limited number of registered antidotes in Bulgaria.

The reason is entirely due to the limited indications for these products. Only specifi c intoxication can be treated with created exactly for this purpose antidote.

Therefore, it can remain unused and this can lead to signifi cant fi nancial losses. The fact is confi rmed by the list of antidotes included in the Diagnostic and therapeutic algorithm for Clinical pathway, № 293/294” Poisoning and toxic effects of medicinal products and household poisons in individuals over 18 / under 18 years.”

Obviously, with this limited resource, the management in case of crisis will be diffi cult, although the defi cit of these medicinal products could bepartially compensated by symptomatic treatment. In other words, the pharmaceutical support with specifi c antidotes of the health care system in the country in crisis is insuffi cient.

We surveyed the literature how other countries provide pharmaceutical support in crisis. Anthony Burda and Todd Sigg in “Pharmacy preparedness for incidents involving weapons of mass destruction” (WMD) describe workable and ease for introduction model.This is one element of the “Nunn-Lugar Cooperative Threat Reduction Program”.

Their paper is more directed to the responsible persons in the pharmaceutical sector and the aim is to provide a system with pharmaceutical products in the case of terrorism. The authors studied the measures that were taken in the U.S. after the attack in the

Page 453: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

451

Table 1

Tokyo subway. It was found that the health system has limited amounts of products for pharmaceutical support in crisis, no reliable guides with information about the hazards from the weapons of mass destructionand means of their elimination, etc.

The article briefl y describes the different types of WMD, their toxicity, the clinical picture of poisoning, and most importantly the medicins, that can be used to treat the victims. All these medicinal products including antidotes are specifi ed by trade name of medicinal products, its generic name, product formulation, selling price, shelf life of the product and the manufacturer and the distributor name, address and contact numbers.

Such detailed information ensures the pharmaceutical support in crisis. Moreover, it has been defi ned the time for which the units participating and responsible for the pharmaceutical support in crisis should act.

Page 454: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

452

Table 1

ConclusionsAntidotes are important part for the pharmaceutical support during crisis. They must

be provided according to the Chapter Three of the Law on Medicinal Products in Human Medicine. If these products are not registered in Bulgaria, they can be provided under the same act, Article 10 (1).

Another disturbing conclusion is that specifi c antidotes may not be utilized in their period of shelf life as pharmaceutical products, in which case the health system would suffers fi nancial losses. These losses, however, can not be legally justifi ed, but they are the

Page 455: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

453

reason for the lack of the availability of antidotes in clinical practice. As an essential part for the medical response during crisis, they should be available although, they may not be utilized during the period of their shelf life.

In other words, the health system must be fi nancially supported in advance to guarantee the availability of specifi c antidotes in case of crisis. The above mentioned American model besides being a good example is a part of sound fi nancial program supported by the Government of the United States, which annually provide funds to enable the population to be provided with everything in the event of a crisis.

We also found that still some representatives of the group of specifi c antidotes are quoted in the regulations, plans for actions in crisis with other names other than those requested by the Law for Medical Products of the Republic of Bulgaria. The use of the generic names or the names in accordance with the law would allow any antidote to be accurately and clearly named, and if needed, it could be easily and quickly supplied.

References:

[1] Law on Pharmaceuticals and Pharmacies in Human Medicine[2] Clinical Financing № 293 “poisoning and toxic effects of drugs and household poisons in individuals over

18 years”.[3] Clinical Financing № 294 “poisoning and toxic effects of drugs and household poisons in individuals less

than 18 years[4] Pharmacy preparedness for incidents involving weapons of mass destruction”.[5] Nunn-Lugar Cooperative Threat Reduction Program

Page 456: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

454

A

abstinence syndrome, 137acetylcholinesterase, 15, 16, 26, 27, 28, 37, 39, 40, 49,

50, 59, 60, 69, 82, 87, 88, 89, 90, 94, 384, 389, 390, 392, 393, 394, 401, 402, 403, 404, 409, 411, 415, 416, 428

acute intoxications, 111, 127, 208, 210, 216acute poisonings, 111, 113, 124, 225, 229ADME, 404, 410adverse drug reactions, 237, 263, 264, 265, 266, 267adverse effects, 129, 134, 148, 150, 151, 155, 207,

263, 277, 333, 338afl atoxins, 129, 131, 135, 433, 440, 441, 444alcain, 189, 191, 192alcohol, 102, 103, 104, 113, 114, 130, 132, 137, 138,

139, 140, 141, 167, 210, 211, 212, 213, 226, 229, 262, 268, 269, 275, 276, 277, 278, 279, 305, 306, 314, 340, 347

amlodipine, 201, 203, 204, 206, 207amphetamine, 102, 106, 109, 110, 238, 241, 242, 243,

244, 245, 246, 254, 255, 256, 257, 258, 259, 260, 261, 262, 300

amphetamine intoxication, 106anaphylaxis, 184, 188, 217anticonvulsant activity, 421, 426antidotal treatment, 98, 409, 415, 416, 417, 418, 419,

420antidotes, 126, 356, 384, 386, 404, 409, 415, 428, 433,

442, 443, 445, 449, 450, 451, 452, 453antioxidant, 81, 82, 148, 149, 154, 156, 157, 158, 160,

161, 165, 236, 237, 238, 260, 261, 262, 268, 271, 276, 277, 278, 295, 297, 298, 299, 311, 313, 317, 322, 325, 398, 399, 400

antioxidant effect, 292, 294antipsychotic medications, 167APOЕ, 345Aronia melanocarpa, 160, 161, 164, 165, 166artifi cial membranes, 404, 405, 408, 410atropine, 18, 19, 42, 93, 94, 203, 384, 385, 386, 388,

402, 411, 412, 416, 417, 418, 419, 420, 421, 423, 425, 426, 427

aum shinrikyo, 369, 370, 371, 372, 373

B

benzene, 119, 148, 321, 322, 324, 325, 326, 327

biogenic monoamines, 341biological warfare, 133, 135, 433, 436biomarker, 15, 16, 17, 24, 25, 27, 28, 35, 36, 37, 39,

46, 49, 69, 79, 93, 94, 98, 349, 393, 395, 396, 397, 398, 399, 401, 402, 403

biomonitoring, 25, 63, 65, 69, 78, 79, 172, 333, 338, 340

biosensor, 15, 16, 17, 19, 24, 25, 26, 28, 31, 35, 36, 38, 39, 43, 49, 50, 51, 52, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 70, 72, 74, 75, 76, 77, 78, 79, 80, 81, 433, 440, 444

biperiden, 427blood, 15, 16, 17, 18, 19, 21, 22, 23, 24, 25, 26, 27, 28,

29, 30, 31, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 73, 75, 76, 77, 78, 79, 80, 82, 89, 93, 94, 95, 96, 98, 102, 103, 104, 117, 118, 121, 122, 124, 126, 133, 137, 138, 154, 157, 158, 167, 168, 169, 170, 171, 172, 174, 186, 191, 201, 206, 238, 254, 255, 256, 269, 270, 271, 302, 303, 304, 305, 339, 361, 363, 365, 384, 386, 389, 390, 391, 392, 396, 397, 398, 399, 400, 402, 403, 411, 414, 417, 446

blood esterases, 15, 22, 23, 24, 26, 27, 28, 29, 36, 38, 44, 48, 52, 59, 67, 75, 78, 80, 82, 98

brain, 6, 7, 42, 47, 141, 147, 185, 241, 257, 260, 261, 262, 272, 279, 306, 414

brain acetylcholinesterase, 50, 69, 392, 411brain toxicity, 238, 241, 246, 254, 255, 257, 259, 260,

268bronchitis, 115, 116, 118, 170, 173, 174butyrylcholinesterase, 10, 15, 16, 24, 26, 27, 28, 36,

37, 60, 79, 81, 82, 83, 88, 89, 90, 92, 94, 98, 389, 390, 393, 394, 401, 402, 445, 446, 447, 448

C

cadmium, 170, 171, 172, 173, 174, 175, 176, 329CaE, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 27, 28, 29,

30, 31, 32, 33, 34, 35, 51, 59, 60, 61, 62, 64, 65, 66, 68, 69, 70, 71, 72, 73, 75, 76, 77, 78, 79, 93, 94, 95, 96, 97, 98

capsaicin, 189, 190, 192, 193carbamates, 336, 384, 385, 386, 387, 388, 389, 390,

391, 392, 401, 402carbon disulfi de, 8, 345, 346, 347, 348, 349, 352, 353carboxylesterase, 15, 16, 25, 26, 27, 28, 36, 37, 38, 66,

Subject Index

Page 457: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

455

68, 69, 70, 79, 80, 81, 88, 89, 91, 93, 94, 98, 396catechol, 51, 70, 321, 322, 323, 324, 325, 326chemical and herbal antidotes, 433chemical incident, 328, 354, 356, 357chemicals, 18, 41, 71, 93, 94, 116, 119, 121, 122, 161,

207, 228, 243, 249, 277, 292, 322, 333, 334, 337, 345, 346, 354, 355, 358, 359, 383, 430, 440, 445

chemical weapon destruction, 374cholinesterase reactivators, 93, 94, 415, 427, 428cholinesterases, 19, 26, 30, 37, 60, 66, 72, 75, 82, 301,

389, 390, 391, 392, 394, 401clinical case, 102, 118, 151, 189clinical trials, 148, 149, 151, 154, 263, 265, 266, 267,

443cocaine, 145, 228, 229, 238, 241, 242, 243, 244, 245,

246, 261, 262, 279, 301, 302, 303, 304, 305, 306, 313, 314, 315, 317, 318, 445

communication, 179, 208, 209, 210, 212, 213, 214, 215, 267

crises, 435, 449cytoprotection, 82, 307, 313cytotoxicity, 135, 182, 236, 248, 250, 251, 252, 253,

262, 269, 278, 280, 281, 282, 283, 285, 286, 292, 296, 297, 298, 307, 310, 315, 316, 317, 318, 323

D

D-amphetamine, 254, 255, 257, 258, 259, 260, 261dependence, 25, 30, 55, 56, 61, 74, 101, 102, 124, 127,

137, 140, 143, 145, 242, 243, 245, 254, 255, 259, 261, 268, 301, 302, 304, 305, 306, 352, 391, 407, 408

destruction of the chemical weapon, 374, 379, 383detection, 15, 19, 25, 28, 36, 43, 49, 51, 52, 53, 54, 56,

57, 59, 61, 68, 70, 74, 75, 78, 79, 82, 85, 86, 87, 93, 263, 264, 266, 267, 286, 304, 306, 336, 337, 338, 339, 340, 350, 389, 390, 393, 399, 400, 430, 432, 433, 437, 439, 440, 444

detoxication, 92, 102, 103, 104, 166, 237, 292, 356distribution, 35, 49, 105, 111, 112, 113, 121, 125, 142,

155, 194, 197, 202, 210, 212, 285, 307, 346, 347, 351, 353, 382, 402, 405, 408, 409, 448, 450

drug addiction, 127, 143, 144, 146drug users, 143, 301

E

e-health system, 328effi ciency, 37, 51, 66, 157, 179, 246, 330, 384, 385,

386, 387, 397enalapril, 201, 202, 203, 204, 206, 207envenomation, 187, 188, 213, 216, 217, 223environmental pollution, 328, 329, 330, 331enzyme-polyelectrolyte nanofi lms, 51enzyme interactions, 148

esterase assay, 26, 51esterase genes, 88, 89, 92esterase status, 15, 17, 24, 25, 26, 27, 28, 29, 33, 34,

35, 36, 80, 93, 94, 98ethanol, 103, 111, 113, 137, 138, 140, 166, 227, 228,

229, 238, 242, 246, 250, 268, 269, 270, 272, 273, 274, 275, 276, 277, 278, 279, 303

exogenic Intoxication, 167exposure, 15, 16, 17, 21, 22, 23, 24, 25, 26, 27, 28, 29,

35, 36, 38, 39, 40, 44, 46, 47, 48, 49, 63, 67, 69, 79, 80, 87, 90, 93, 94, 98, 115, 116, 118, 119, 120, 121, 122, 123, 129, 130, 133, 134, 136, 166, 170, 171, 172, 173, 174, 175, 176, 181, 182, 183, 189, 190, 191, 192, 276, 277, 279, 280, 282, 283, 299, 321, 324, 333, 334, 335, 338, 339, 340, 345, 346, 347, 348, 349, 351, 352, 353, 356, 357, 359, 362, 364, 365, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 405, 417, 418, 419, 426, 434, 437, 440, 441, 442, 446

F

factors, 29, 115, 116, 119, 120, 121, 122, 123, 130, 140, 156, 176, 177, 178, 179, 182, 183, 194, 195, 196, 197, 198, 199, 208, 209, 210, 226, 233, 235, 240, 263, 264, 266, 267, 280, 283, 293, 301, 302, 345, 347, 355, 384, 396, 401

Flavonoids, 148, 149, 155fruit juice, 129, 160, 161, 165, 166

G

genetic polymorphism, 37, 81, 296, 352gold nanoparticles, 280, 286Gypsophila trichotoma, 82, 313, 314, 315, 318

H

hair, 333, 334, 335, 336, 337, 338, 339, 340health effects, 27, 118, 119, 136, 174, 175, 177, 180,

182, 193, 330, 333, 334, 345, 346, 437, 438, 444health risk assessment, 4, 119, 338, 345, 354, 355, 357,

359, 433hepatocytes, 82, 236, 239, 240, 246, 254, 255, 256,

258, 259, 260, 261, 269, 278, 280, 281, 285, 293, 294, 296, 297, 298, 299, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 346, 402

hepatotoxicity, 132, 135, 160, 161, 163, 164, 165, 166, 202, 235, 236, 240, 254, 259, 260, 261, 262, 268, 275, 295, 297, 299, 300, 310, 312, 315, 318, 437

HEP G2, 280, 281, 282, 283, 284, 285hexobarbital, 287, 288HI-6, 93, 94, 95, 96, 97, 98, 384, 411, 412, 415, 416,

417, 418, 419, 420, 421, 423, 424, 425, 426, 428, 429, 430, 432

hippocampus, 144, 341, 342histamine, 137, 138, 139, 140, 141, 142, 185, 217

Page 458: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

456

HPLC, 166, 311, 339, 347, 422, 428, 430, 432, 439human, 15, 16, 18, 20, 24, 25, 26, 27, 28, 29, 30, 31,

33, 34, 35, 36, 37, 38, 49, 50, 58, 61, 62, 63, 64, 65, 68, 69, 70, 71, 75, 76, 77, 78, 79, 80, 81, 92, 93, 94, 98, 111, 127, 129, 135, 136, 141, 142, 143, 144, 145, 146, 147, 148, 149, 152, 153, 154, 155, 158, 175, 177, 180, 183, 193, 194, 195, 217, 222, 248, 249, 250, 251, 252, 253, 266, 269, 278, 280, 281, 282, 284, 286, 295, 296, 300, 302, 306, 308, 312, 321, 325, 326, 327, 329, 330, 333, 334, 335, 336, 337, 338, 339, 340, 353, 357, 358, 359, 360, 361, 363, 365, 385, 389, 390, 391, 393, 395, 396, 397, 398, 399, 400, 401, 402, 404, 409, 415, 429, 433, 434, 435, 436, 437, 443, 444, 445, 446, 448, 449, 452, 453

human rights, 143, 144, 145, 146, 147human toxicity, 360hypoxia, 101, 104, 105, 238, 276, 327

I

identifi cation of polymorphism genotypes, 88intoxication, 24, 47, 48, 93, 94, 95, 96, 97, 102, 103,

104, 106, 114, 115, 118, 124, 132, 170, 184, 185, 187, 201, 203, 205, 207, 208, 209, 210, 211, 212, 213, 214, 228, 236, 298, 354, 356, 364, 365, 387, 388, 393, 394, 403, 409, 410, 411, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 445, 450

isolated rat hepatocytes, 7, 82, 236, 240, 246, 254, 255, 261, 293, 294, 296, 297, 298, 299, 307, 308, 309, 310, 311, 312, 313, 315, 316, 317, 318

L

L-valine, 243, 256, 270, 341Layer-by-layer, 26, 38, 51, 52, 54, 66, 68, 70, 80LBL, 17, 19, 35, 39, 43, 51, 65, 67, 68, 70, 74, 77,

78, 80learning, 108, 302, 341, 426life-threatening, 106, 184localanesthetic, 189

M

manganese dioxide, 52, 53, 82, 83, 84, 85, 87manganese parkinsonism, 115medical Education and Training, 354medical education and training, 357medical intelligence, 360, 362, 363, 364medical Support and Management, 354memory, 234, 341, 342metal aerosols, 115methadone, 101, 102, 103, 104, 105, 107, 109, 110,

124, 125, 126, 127, 128, 145midasolam, 416

military personnel, 445, 446, 447, 448miners, 119, 120, 121, 122, 123morbidity, 111, 112, 113, 114, 118, 123, 167, 187, 209, 213,

218, 225, 226, 267, 328, 329, 330, 331, 361, 364mortality, 115, 123, 134, 167, 171, 174, 175, 176, 225,

226, 229, 267, 287, 289, 329, 330, 331, 343, 361multidisciplinary team, 124, 127mycotoxicoses, 129, 131, 134, 135, 434, 437, 444mycotoxins, 129, 130, 131, 132, 133, 134, 135, 136,

182, 433, 434, 435, 436, 437, 439, 440, 441, 442, 443, 444

myosmine, 7, 307, 308, 309, 310, 311, 312

N

7-Nitroindazole, 6, 241, 242, 244, 245, 246, 247nano toxicity, 280neonicotinoids, 336, 337, 360, 361, 362, 363, 364,

365nerve agents, 24, 69, 79, 87, 364, 387, 393, 397, 399,

400, 405, 411, 412, 413, 415, 416, 417, 419, 420, 421, 422, 425, 426, 427, 445, 448

neurobiological studies, 143neuroleptix, 167neuropathy target esterase, 15, 16, 25, 26, 27, 28, 36,

38, 39, 40, 49, 50, 66, 67, 68, 69, 79, 80, 88, 91, 92, 94, 394, 401

neuropeptides, 190, 287, 288, 289, 291nnos, 241, 242, 243, 244, 245, 246, 254, 257, 259, 260,

261, 268, 272, 275, 277nociception, 287, 289non-tumor cell lines, 248NTE, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 27, 28, 29,

31, 34, 35, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 59, 60, 61, 62, 63, 64, 65, 68, 69, 70, 73, 75, 76, 77, 78, 79, 92, 94, 394

O

ochratoxins, 129, 433, 435, 436, 437, 440, 441ocspray, 189oleoresin Capsicum, 189, 192olinical case, 115, 167OPCs, 15, 16, 17, 19, 20, 23, 24, 27, 28, 29, 39, 40, 42,

43, 44, 45, 48, 69, 88, 93, 94, 98OPRD1, 301, 302, 303, 305, 306organic pollutants, 333, 334, 336, 337, 338organophosphorus, 15, 16, 24, 25, 26, 27, 36, 37, 38,

39, 40, 49, 69, 79, 80, 84, 88, 93, 94, 98, 336, 339, 389, 390, 392, 393, 400, 401, 402, 403, 411, 415, 416, 421, 426, 427, 445, 447, 448

organophosphorus compounds, 15, 16, 24, 25, 26, 27, 36, 37, 38, 39, 49, 79, 80, 88, 98, 389, 390, 392, 393, 400, 402, 411, 415, 421, 445, 447, 448

organophosphorus compounds, 15, 16, 24, 25, 26, 27,

Page 459: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

457

36, 37, 38, 39, 49, 79, 80, 88, 389, 390, 392, 402, 411, 415, 421, 445, 447, 448

organopohosphates, 384oxime reactivators, 411, 413, 414, 417oximes, 384, 386, 387, 399, 402, 404, 405, 406, 407, 408,

409, 410, 411, 412, 413, 414, 415, 420, 428, 432

P

paracetamol, p160, 161, 162, 164, 237paraoxon, 18, 19, 31, 33, 37, 41, 42, 43, 60, 63, 70,

73, 75, 76, 81, 91, 92, 93, 94, 95, 96, 97, 98, 384, 385, 386, 391, 392, 397, 432

paraoxonase, 17, 25, 27, 28, 29, 30, 33, 36, 37, 68, 69, 70, 79, 81, 92, 93, 94, 98, 397, 402, 403

paraoxonase 1 gene, 88, 89, 91, 92PCR, 88, 89, 90, 91, 301, 303, 304, 305, 345, 348, 353, 448PCR technique, 88pepperspray, 189permeability, 238, 322, 324, 325, 327, 344, 386, 404,

405, 406, 407, 408, 409, 410persistent vegetative state, 101, 103, 104pharmacovigilance, 263, 264, 267phenol, 19, 31, 35, 43, 51, 57, 59, 60, 62, 63, 65, 66,

68, 69, 70, 72, 73, 74, 75, 76, 77, 78, 80, 89, 149, 261, 271, 278, 321, 322, 323

physostigmime, 416pneumoconiosis, 115, 116, 118, 120, 123pneumofi brosis, 118, 170, 172, 173, 174PON1, 17, 19, 25, 27, 28, 29, 30, 31, 33, 34, 35, 36, 37,

60, 68, 69, 70, 72, 73, 75, 76, 77, 78, 79, 81, 88, 89, 90, 91, 92, 93, 94, 95, 96, 397, 398, 402, 403

procyclidine, 416, 417, 418, 419, 420prophylaxis, 142, 216, 220, 387, 388, 416, 418, 419,

422, 427, 448protective effect, 160, 165, 236, 237, 260, 293, 299,

307, 384, 385protective factors, 194proteinuria, 170, 174, 343psychosocial aspects, 124

Q

Quercetin, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159

R

rats, 26, 27, 28, 29, 30, 31, 34, 36, 37, 38, 40, 41, 47, 49, 69, 80, 93, 94, 98, 136, 150, 156, 158, 159, 160, 161, 163, 164, 165, 166, 237, 238, 239, 241, 242, 243, 244, 245, 246, 247, 254, 255, 256, 257, 258, 259, 260, 261, 262, 268, 269, 270, 272, 273, 274, 275, 276, 277, 278, 279, 285, 291, 292, 293, 296, 299, 300, 306, 308, 314, 318, 341, 342, 343, 360, 361, 384, 385, 387, 396, 398, 403, 411, 412,

415, 417, 418, 420, 421, 422, 423, 427reactionary masses, 374, 375, 379, 380, 383resilience, 194, 195, 196, 198, 200restriction endonuclease cleavage, 88rodents, 27, 28, 29, 34, 35, 165, 275, 287, 341

S

saponarin, 313sarin, 38, 69, 91, 94, 369, 370, 371, 372, 373, 388, 389,

390, 391, 392, 399, 409, 410, 411, 416, 420, 421, 426, 445

schiff bases, 248scolopendra, 216, 223, 224screen printed electrodes, 51serotoninsyndrome, 106, 110SHR, 238, 239, 240, 254, 255, 257, 258, 259, 260, 261,

262, 268, 269, 270, 273, 274, 275, 276, 277, 279, 299

sick building syndrome, 91, 92, 131, 177, 178, 181, 182, 183

silver, 248, 252, 280, 283, 286soil pollution, 374soman, 69, 91, 94, 98, 371, 384, 385, 386, 387, 388,

389, 390, 391, 399, 400, 411, 412, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 432, 445

suicidal self-poisonings, 194synthesis, 18, 24, 41, 48, 52, 80, 83, 107, 132, 136,

155, 240, 249, 276, 278, 285, 311, 327, 345, 346, 353, 360, 370, 380, 381, 382, 428, 429, 434

T

tabun, 69, 94, 371, 384, 388, 399, 400, 411, 412, 413, 414, 415, 416, 421, 426

technogenic emissions of arsenic, 374thermal neutralization, 379, 383thick fi lm thiol sensitive sensor, 82toxicity, 16, 17, 20, 21, 22, 23, 24, 28, 29, 36, 37, 38,

39, 40, 41, 43, 44, 45, 47, 48, 69, 79, 90, 107, 108, 126, 131, 132, 135, 136, 148, 149, 150, 152, 154, 156, 160, 161, 165, 166, 201, 203, 204, 207, 209, 233, 234, 235, 236, 237, 238, 239, 240, 241, 246, 252, 254, 255, 257, 259, 260, 261, 268, 269, 275, 278, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 291, 292, 293, 294, 295, 297, 298, 299, 300, 307, 312, 313, 315, 318, 321, 326, 333, 341, 342, 344, 358, 360, 361, 362, 364, 374, 377, 383, 384, 385, 386, 387, 389, 393, 397, 398, 400, 410, 411, 416, 417, 425, 434, 437, 438, 441, 443, 451

toxicology, 24, 27, 36, 37, 49, 70, 101, 111, 112, 135, 155, 208, 209, 210, 212, 214, 225, 233, 234, 235, 240, 254, 262, 268, 278, 309, 327, 356, 364, 393, 399, 400, 401, 402, 403, 441, 445

Page 460: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

458

toxidine, 93, 94, 428, 429, 430toxoallergic reaction, 216treatment, 15, 17, 23, 24, 48, 65, 78, 88, 90, 91, 92, 94, 95,

96, 97, 98, 101, 102, 103, 104, 105, 109, 110, 111, 114, 124, 125, 126, 127, 128, 130, 132, 134, 141, 143, 144, 145, 146, 149, 150, 151, 152, 157, 161, 162, 167, 168, 179, 185, 189, 191, 193, 198, 201, 202, 203, 206, 208, 209, 210, 212, 213, 214, 216, 217, 218, 220, 221, 222, 223, 228, 237, 241, 252, 253, 255, 263, 265, 268, 275, 287, 289, 306, 324,

335, 341, 343, 356, 357, 361, 362, 364, 384, 398, 402, 407, 408, 409, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 427, 445, 449, 450

trichothecenes, 129, 131, 132, 135, 136, 442tyrosinase, 16, 17, 19, 25, 26, 31, 35, 38, 39, 43, 51,

52, 56, 57, 58, 61, 64, 65, 66, 68, 70, 71, 72, 74, 75, 76, 78, 80

Z

zinc, 175, 248, 252, 253

Page 461: Toxicilogical problems 1

TOXICOLOGICAL PROBLEMS

459

Page 462: Toxicilogical problems 1

C. Dishovsky, J. Radenkova-Saeva

460