Top Banner
This article was downloaded by: [122.172.193.255] On: 26 July 2013, At: 07:33 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK Journal of Biomolecular Structure and Dynamics Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/tbsd20 Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi, Vibrio cholerae, and Staphylococcus areus by potential phytoligands via in silico screening Sinosh Skariyachan a , Nisha Jayaprakash a , Navya Bharadwaj a & Rajeswari Narayanappa a a R&D Centre, Department of Biotechnology , Dayananda Sagar College of Engineering , Bangalore , 560 078 , Karnataka , India Published online: 23 Jul 2013. To cite this article: Journal of Biomolecular Structure and Dynamics (2013): Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi, Vibrio cholerae, and Staphylococcus areus by potential phytoligands via in silico screening, Journal of Biomolecular Structure and Dynamics, DOI: 10.1080/07391102.2013.819787 To link to this article: http://dx.doi.org/10.1080/07391102.2013.819787 PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions
18

Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Apr 01, 2023

Download

Documents

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: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

This article was downloaded by: [122.172.193.255]On: 26 July 2013, At: 07:33Publisher: Taylor & FrancisInforma Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,37-41 Mortimer Street, London W1T 3JH, UK

Journal of Biomolecular Structure and DynamicsPublication details, including instructions for authors and subscription information:http://www.tandfonline.com/loi/tbsd20

Exploring insights for virulent gene inhibition ofmultidrug resistant Salmonella typhi, Vibrio cholerae,and Staphylococcus areus by potential phytoligands viain silico screeningSinosh Skariyachan a , Nisha Jayaprakash a , Navya Bharadwaj a & Rajeswari Narayanappa aa R&D Centre, Department of Biotechnology , Dayananda Sagar College of Engineering ,Bangalore , 560 078 , Karnataka , IndiaPublished online: 23 Jul 2013.

To cite this article: Journal of Biomolecular Structure and Dynamics (2013): Exploring insights for virulent gene inhibitionof multidrug resistant Salmonella typhi, Vibrio cholerae, and Staphylococcus areus by potential phytoligands via in silicoscreening, Journal of Biomolecular Structure and Dynamics, DOI: 10.1080/07391102.2013.819787

To link to this article: http://dx.doi.org/10.1080/07391102.2013.819787

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) containedin the publications on our platform. However, Taylor & Francis, our agents, and our licensors make norepresentations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of theContent. Any opinions and views expressed in this publication are the opinions and views of the authors, andare not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon andshould be independently verified with primary sources of information. Taylor and Francis shall not be liable forany losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoeveror howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use ofthe Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematicreproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in anyform to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://www.tandfonline.com/page/terms-and-conditions

Page 2: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi, Vibriocholerae, and Staphylococcus areus by potential phytoligands via in silico screening

Sinosh Skariyachan, Nisha Jayaprakash, Navya Bharadwaj and Rajeswari Narayanappa*

R&D Centre, Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore 560 078, Karnataka, India

Communicated by Ramaswamy H. Sarma

(Received 22 April 2013; final version received 24 June 2013)

In our recent studies on prevalence of multidrug resistant pathogens in Byramangala reservoir, Karnataka, India, weidentified Salmonella typhi, Staphylococcus aureus, and Vibrio cholerae which had acquired multiple drug resistance(MDR) and emerged as superbugs. Hence, there is a pressing demand to identify alternative therapeutic remedies. Ourstudy focused on the screening of herbal leads by structure-based virtual screening. The virulent gene products of thesepathogens towards Kanamycin(aph), Trimethoprim(dfrA1), Methicillin (mecI), and Vancomycin (vanH) were identified asthe probable drug targets and their 3D structures were predicted by homology modeling. The predicted models showedgood stereochemical validity. By extensive literature survey, we selected 58 phytoligands and their drug likeliness andpharmacokinetic properties were computationally predicted. The inhibitory properties of these ligands against drug targetswere studied by molecular docking. Our studies revealed that Baicalein from S. baicalensis (baikal skullcap) and Luteolinfrom Taraxacum officinale (dandelion) were identified as potential inhibitors against aph of S. typhi. Resveratrol from Vitisvinifera (grape vine) and Wogonin from S. baicalensis were identified as potential inhibitors against dfrA1 of S. typhi.Herniarin from Herniaria glabra (rupture worts) and Pyrocide from Daucus carota (Carrot) were identified as the bestleads against dfrA1 of V. cholerae. Taraxacin of T. officinale (weber) and Luteolin were identified as potential inhibitorsagainst Mec1. Apigenin from Coffee arabica (coffee) and Luteolin were identified as the best leads against vanH ofS. aureus. Our findings pave crucial insights for exploring alternative therapeutics against MDR pathogens.

Keywords: multidrug resistant pathogens; virulent genes; homology modeling; virtual screening; potential inhibitors;herbal leads

1. Introduction

Antibiotics are the most common chemotherapeuticsubstances used against human illness, agricultural pests,and livestock (Bao, Maruya, Snyder, & Zeng, 2012;Hagedorn et al., 1999). Residues from many ecologicalniches may contain these drugs or drug resistance genesthat can contaminate natural ecosystems. One of themajor impacts of antibiotic release in natural environ-ments is the emergence of multidrug resistant (MDR)pathogens. Most of the bacteria acquired multipleresistances to routinely used antibiotics and emerged asMDR and extensively drug-resistant (XDR) pathogens.They not only require longer and more complextreatments, but are more exorbitant to diagnose and treat.The molecular mechanisms for antibiotics’ resistancesamong bacterial populations are quite complex anddiverse, which developed resistance to many convention-ally used antibiotics, including the strongest β- lactamgroups (Martínez, 2012; Walsh, Weeks, Livermore, &

Toleman, 2011). Hence, the therapeutic values of antimi-crobial drugs are constantly diminishing, which havesignificant global attention (Davies & Davies, 2010).

The mechanisms of drug resistance could be acquiredand transmitted horizontally through the conjugation of aplasmid or by acquisition of drug resistance genes andtransposable elements. Recently, new mechanisms ofresistances have resulted in the simultaneous developmentof resistance to several antibiotic classes creating verydangerous multidrug-resistant (MDR) pathogens knownas “superbugs” (Maclean, Hall, Perron, & Buckling,2010). The overuse and misuse of these chemotherapeuticagents in pharmaceutical and food industry are one ofthe main factors for drug resistance (Giedraitienė,Vitkauskienė, Naginienė, & Pavilonis, 2011).

By considering all these socioenvironmental issues,there is a pressing need for screening novel leadmolecules. The new approaches which have to be imple-mented include: identification of new molecular markers,

*Corresponding author. Email: [email protected]

Journal of Biomolecular Structure and Dynamics, 2013http://dx.doi.org/10.1080/07391102.2013.819787

Copyright � 2013 Taylor & Francis

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 3: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

identification of novel lead molecules for antibioticdevelopment, identification of novel treatment methods,and identification of a sample bacteria and its susceptibil-ity to antibiotic treatment. There is also a need for adeeper understanding of the mechanisms by whichbacteria gain resistance to antibiotics, which will aidin identifying novel targets for drugs or treatment(Daniels, 2011).

Our previous studies on characterization ofpathogenic bacteria from water samples collected fromByramangala reservoir, a major source of drinking waterfor urban and rural residents in Bangalore District,Karnataka, India showed that Salmonella typhi, Staphylo-coccus aureus, and Vibrio cholerae acquired highdrug resistance against conventional therapeutic drugs.S. aureus showed resistance to conventionally used drugssuch as Vancomycin, Methicillin, Oxacillin, Tetracycline,etc. Similarly, Salmonella sps showed resistance toTrimethoprim, Kanamycin, Cefoperazone, Cefotaxime,Cefixime, Moxifloxacin, Piperacillin/Tazobactam, Co-trimoxazole, Levofloxacin, Trimethoprim, and Ceftazi-dime (Skariyachan et al., 2012). These pathogens areresponsible for adverse health implications in humansand animals. The drug resistance seems mainly due tomutations, acquisition of drug resistance genes, transferof drug-resistant plasmids, and horizontal gene transfer(Lupo, Coyne, & Berendonk, 2012; Nazir, Abraham, &Islam, 2012). Hence, there is a necessity for alternativetherapeutic strategies against such kind of pathogens. Apromising approach is to inhibit the expression ofdrug-resistant genes by potential inhibitors. The inhibi-tions of virulent gene products by natural remedies haveprofound scope in health care management. Structure-based virtual screening offers a significant application toscreen such kind of natural substances, especially theactive compounds present in many medicinal herbs. Thevirulent functions of drug-resistant genes could beprevented by such kind of lead molecules. This couldgive an alternative therapeutic insight against MDR andXDR pathogens when most conventional therapeuticsubstance seems to have failed. Hence, we aimed toevaluate the inhibitory properties of various herbal leadmolecules against virulent gene products of these threepathogenic bacteria by structure-based virtual screening.The new lead molecules were chosen based ontraditional knowledge of medicine as their use is wellestablished and widely acknowledged to be safe andeffective.

2. Materials and methods

2.1. Identification of virulent gene products for drugresistance

The genes responsible for multidrug resistances ofS. typhi, S. aureus, and V. cholerae toward present

generation drugs such as Kanamycin, Trimethoprim,Methicillin, and Vancomycin were retrieved from EntrezGene database (Maglott, Ostell, Pruitt, & Tatusova,2011). The amino acid sequences of all gene productswere retrieved from Uniprot database (Apweiler et al.,2004). The proteins responsible for drug resistancetowards Kanamycin (UniProt: E2D0Y8), Trimethoprim(UniProt: A7DY50, G7TU76), Methicillin (UniProt:P68261), and Vancomycin (UniProt: 7BWD8) wereretrieved in FASTA format.

2.2. Computer aided molecular modeling andvalidation

The three-dimensional structure of the selected drugtargets is essential for structure-based virtual screeningand drug designing. The structure of Methicillin-resistantprotein is available in its native form (PDB: 1OKR)(García-Castellanos et al., 2003). However, the three-dimensional structures of drug-resistant gene productsfor Kanamycin, Trimethoprim, and Vancomycin are notavailable in their native form. Hence, the structures ofthese virulent proteins were predicted by computer-aidedhomology modeling.

The amino acid sequences of the virulent proteins forKanamycin of S. typhi, Trimethoprim of S. typhi andV. cholerae, and Vancomycin of Staphylococcus areuswere retrieved from UniProt database. The best templatestowards homology modeling of all selected genes wereretrieved by PSI-BLAST (Altschul et al., 1997). The besthomologous templates were selected based on thepercentage of identity, similarity, alignment scores, andE-value. The three-dimensional structural coordinates ofall the best templates were retrieved from Protein DataBank (Berman, Henrick, Nakamura, & Markley, 2007).The homologous relationship between the targets andtemplates were further confirmed by multiple sequencealignment and phylogenetic analysis (Supplementarymaterials, Figure 1) by T-COFFEE (Notredame, Higgins,& Heringa, 2000).

The three-dimensional structures of Kanamycin-,Trimethoprim-, and Vancomycin-resistant proteins werepredicted by comparative modeling by Modeller 9v11(Eswar, Eramian, Webb, Shen, & Sali, 2008). Thestructural alignment between the target and templatewere performed and the initial model was created. Themodeled proteins were visualized using UCSF-Chimera(Pettersen et al., 2004) and refined by various computa-tional biology tools. The stereochemistries of modeledproteins were validated by ProCheck (Laskowski,Rullmann, MacArthur, Kaptein, & Thornton, 1996). Theoverall quality factors of nonbonded interactions betweendifferent atoms were calculated by ERRAT (Colovos &Yeates, 1993). The backbone fragments of the modeledprotein were threaded against the template chains byDaliLite (Holm, Kääriäinen, Rosenström, & Schenkel,

2 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 4: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

2008) and root mean square deviation (RMSD) wascalculated. The hypothetical model was deposited to Pro-tein Model Data Base (Castrignanò, De Meo, Cozzetto,Talamo, & Tramontano, 2006). These modeled proteinswere used as probable drug targets for further analysis inour study.

2.3. Screening and selection of phytoligands

We had identified about 58 phytoligands after extensiveliterature survey and they were screened based on druglikeliness and pharmacokinetics prediction by PreAD-MET tool (Veber et al., 2002). The selected phytoligandsare shown in supplementary materials, Table 1. Thethree-dimensional structures of these phytoligands wereretrieved from ChemSpider (Hettne et al., 2010) data-base. The drug likeness properties of the phytoligandswere computationally predicted by Lipinski’s rule of five(Giménez, Santos, Ferrarini, & Fernandes, 2010),Comprehensive Medicinal Chemistry (CMC)-like rule(Ajay, Walters, & Murcko, 1998), MDDR (MDL DrugData Report)-like rule (Frimurer, Bywater, Naerum,Lauritsen, & Brunak, 2000), Lead-like Rule (Opreaet al., 2007) and World drug index (WDI)-like rule(Wagener & van Geerestein, 2000) available in PreAD-MET package. Those lead molecules that qualified therules were selected for docking studies. Subsequently,pharmacokinetic features such as human intestinalabsorption, Caco2 (heterogeneous human epithelial colo-rectal adenocarcinoma) cell permeability, Madin-Darbycanine kidney (MDCK) cell permeability, skin perme-ability, and blood brain barrier penetration were alsopredicted by PreADMET (Veber et al., 2000). In additionto ADME (adsorption, distribution, metabolism, andexcretion) prediction, toxicity was also predicted toanalyze carcinogenicity in mouse and rat models bycomputer-aided approach. Similarly, the mutagenecity ofthe selected lead molecules was predicted using Amestest by PreADMET.

2.4. Structure-based drug design

The study of receptor–ligand interactions is the mainconcept behind structure-based drug designing (Zhang &Lai, 2011). The inhibitory properties of the selectedligands against probable drug targets were studied bymolecular docking by AutoDock Vina (Trott & Olson,2010). This program was executed to simulate the realtime molecular interactions of the receptor and ligands.The active site of the target was identified based on thestructural and chemical properties by Lamarckian geneticalgorithm and AutoGrid program. Rigid body dockingstudies were carried out in which the function scoring,energy calculations, and ranking of the best conforma-tions were carried out. The electrostatic interactions weredetermined by interpolating the values of the electrostaticpotential and multiplying by the charge on the atom.

Around 2500,000 binding poses were identified by 10iterations and the best binding poses were selected basedon the lowest energy calculations and the number ofhydrogen bonds stabilizes the interaction.

3. Results and discussion

Our previous pilot studies on characterization ofpathogenic bacteria from water samples collected fromByramangala reservoir revealed that most isolates werefound to be drug resistances to conventionally used anti-biotics (Skariyachan et al., 2013). From our studies, itwas evident that S. typhi, S. aureus, and V. cholerae haveshown maximum drug resistances. Hence, we haveselected these organisms for present studies. S. typhi wasfound to be resistant against the conventionally used anti-biotics such as Trimethoprim and Kanamycin. V. choleraealso showed drug resistance to Trimethoprim. S. aureusshowed multidrug resistance to present generationdrugs such as Methicillin and Vancomycin (Skariyachanet al., 2012). Our previous results clearly indicate thatthese bacteria became resistant to conventionally useddrugs and emerged as superbugs (multidrug-resistantorganisms). Recent studies reported that many presentgeneration drugs seem to have failed against superbugswhich have created major health implications worldwide(Nazir, Abraham, & Islam, 2012). Hence, in the presentstudy, we have focused to inhibit the virulent functionsof the drug-resistant genes by novel phytoligands. Ourprevious studies proved the utility of computer-aidedvirtual screening toward many multidrug-resistantpathogens (Skariyachan et al., 2012). Several herbal sub-stances are known to have vital inhibitory propertiesagainst various drug targets of many gastrointestinalpathogens (Eumkeb & Chukrathok 2013; Eumkeb,Siriwong, & Thumanu, 2012; Rúa et al., 2012; Schrader,2010).

The genes responsible for multidrug properties of thementioned organisms were screened from Entrez Genedatabase and the protein sequences of these geneswere retrieved from Uniprot database. These geneswere screened based on literature studies, where the genesequence coding for the MDR properties of thesame strains were already reported (Chen et al., 2010;Hiramatsu, Asada, Suzuki, Okonogi, & Yokota, 1992;Martínez et al., 2007; Reimer et al., 2011; Weigel et al.,2003). Aminoglycoside phosphotransferase (aph; UniprotID: E2D0Y8) of 271 amino acids length, virulent proteinfor Kanamycin resistance (Chen et al., 2010), anddihydrofolate reductase (dhfr; Uniprot ID: A7DY50) ofsequence length 152 amino acids, responsible factor forTrimethoprim resistance of S. typhi (Martínez et al.,2007), were selected. Similarly, dihydrofolate reductasetype I (dfrA1; Uniprot ID: G7TU76), virulent factor forTrimethoprim resistance, of 157 amino acids from

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 3

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 5: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

V. cholerae (Reimer et al., 2011) were selected. Methicil-lin-resistant gene (Mec1; Uniprot ID: P68261) of 123amino acids (Hiramatsu et al., 1992) and Vancomycin-resistant gene (VanH; Uniprot ID: Q7BWD8) of 322amino acids from S. aureus (Weigel et al., 2003) were alsoselected. The three-dimensional structure of aminoglyco-side phosphotransferase, dihydrofolate reductase, andvancomycin-resistant proteins are not reported in any ofthe structural database. Hence, homology modeling wascarried out to predict the three-dimensional structure ofthese proteins by Modeller 9v11. The tool PSI-BLAST

was used to identify the best templates for modeling.Further, multiple sequence analysis was performed byT-COFFEE and the phylogenetic characterization wascarried out by NJ plot. This step is important to identifythe best templates for modeling as the templates sharedcommon evolutionary origin with the targets in a phyloge-netic tree. The features of the best homologous templatesare shown in Table 1.

The structural alignments between each of the targetwith their template showed that the qualities of allmodels are reliable. The structure-annotated pairwise

Table 1. Identification of the best homologous templates for homology modeling of virulent proteins.

TargetUniprotID Organism Template Organism

PDB IDandChain Length

Percentageof Identity

E-value

Secondarystructure

Resolutionfactor ofthe crystalstructure

(Å)

aph E2D0Y8 S. typhi Aminoglycosidephosphotransferase

Acinetobacterbaumannii

4EJ7_A 292 99 0 61%helices

2.29

11% sheetdhfr A7DY50 S. typhi Dihydrofolate

reductaseStreptococcuspneumoniae

3IX9_A 190 31 1e-13

20%helices

1.95

30% sheetvanH Q7BWD8 S. aureus Lactate

dehydrogenaseLactobacillushelveticus

2DLD_A 337 35 1.5e-48

38%helices

2.7

16% sheetdfrA1 G7TU76 V.

choleraeDihydrofolatereductase

S. aureus 3IX9_A 190 82 1e-21

20%helices

1.8

30% sheet

Figure 1. (a) Three-dimensional model of Aminoglycoside 3'-phosphotransferase type I (aph) of S. typhi. (b) Ramachandran plot of themodel showed that 94.9% residues are in most favored region [A, B, L] and 5.1% residues are in additional allowed region [a, b, l, p].

4 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 6: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

sequence identity between targets and best templatesgenerated by DaliLite revealed the good quality of themodel. (Supplementary materials, Figures 1–4). The rootmean square deviation (RMSD) values of structuralsuperposition between models, aminoglycoside phospho-transferase, dihydrofolate reductase (S. typhi), vancomy-cin resistance protein, and dihidrofolate reductase type I(V. cholerae) were calculated and the best templates werefound to be 2, 1.6, 1.6, and 1.1Å, respectively. RMSDstatistics are generally used to monitor the degree ofsimilarity between two optimally superposed protein

three-dimensional structures (Bennett, 2008) and assessthe quality of the model. The Z scores of each modelwere determined by DaliLite (Holm et al., 2008) werefound to be acceptable (Z score was 41.9 for aph, 44.7for dhfr, 44.7 for vanH and 27.7 for dfrA1) in compari-sons with experimental structures, which implies goodquality models. The Z scores of our models are reliable,which reveals the accuracy and native folds of proteins(Carugo, 2007).

The hypothetical models were further refined by vari-ous stereochemical analyses. The Ramachandran plot of

Figure 2. (a) Three-dimensional model of dihidrofolate reductase (dhfr) of S. typhi. (b) Ramachandrn plot of the model showed that91.7% residues are in most favored region [A, B, L] and 8.3% residues are in additional allowed region [a, b, l, p].

Figure 3. (a) Three-dimensional model of Dihydrofolate reductase type 1(dfrA1) of V. cholerae. (b) Ramachandrn plot of the modelshowed that 92.7% residues are in most favored region [A, B, L] and 7.3% residues are in additional allowed region [a, b, l, p].

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 5

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 7: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

aminoglycoside phosphotransferase (aph) of S. typhirevealed that 94.9% of residues in the most favoredregion and 5.1% residues are additional allowed regions(Figure 1). The overall quality factor of aph identified byERRAT was found to be 87.79%. The hypotheticalmodel of dihydrofolate reductase (dhfr) of S. typhi alsohas good stereochemical quality, which indicates 91.7%

of residues in the most allowed region and 8.3% residuesare additional allowed regions (Figure 2). However, wehave observed errors in ERRAT plot and the patterns ofnonbonded atomic interactions were found to be 40.97%.Similarly, Ramachandran plot of dihydrofolate reductasetype I (dfrA1), drug target of V. cholerae, indicated that92.7% of residues are in the most favored region and

Figure 4. (a) Three-dimensional model of Vancomycin resistance protein (vanH) of S. aureus. (b) Ramachandrn plot of the modelrevealed that 91.0% residues are in most favored region [A,B,L] and 8.3% residues are in additional allowed region [a, b, l, p].

Figure 5. Inhibitory properties of potential phytoligands toward aminoglycoside 3'-phosphotransferase type I (aph) of S. typhiidentified by molecular docking. Ligands and interacting residues are shown in stick figures (a) Phytoligand Baicalien interacted withmodeled aph protein at Thr 99 and Thr 98 by the formation of two hydrogen bonds (binding energy �6.39 Kcal/mol). (b) Interactionof Luteolin with aph is stabilized by two hydrogen bonds, which are shown in the figure. The interacting amino acids are found to beSer 10 and Asn 88 (binding energy �6.42 Kcal/ mol).

6 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 8: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Table 2. Docking interactions between aminoglycoside phosphotransferase (aph), virulent factor for Kanamycin resistance, anddihydrofolate reductase (dhfr), virulent factor for Trimethoprim resistance, of S. typhi, and the selected lead molecules. The bindingenergy (Kcal/mol) and interacting residues are shown in the table. Baicalin and Luteolin against aph and Resveratrol and Wogoninagainst dhfr were identified as the best inhibitors as these liagnds showed minimum binding energy and more number of hydrogenbonds compared to other lead molecules. The best ligands are highlighted.

Ligand

Interaction with Aminoglycoside phosphotransferase(aph) Interaction with dihydrofolate reductase (dhfr)

BindingEnergy (Kcal/

mol)

Number ofhydrogenbonds

Amino acidinteracting withligand

Binding energy(Kcal/mol)

Number ofhydrogenbonds

Amino acidinteracting withligand

Allicin �6.17 2 Ile101, Thr98 –3.43 1 Gly94Apigenin �5.14 2 Thr98, Thr99 –6.43 1 Glu23Baicalein �6.39 2 Thr 98, Thr 99 –5.29 2 Ser92, Ala3Borneol �5.1 1 Asp216 –4.34 2 Ser92, Tyr98Deoxyhexose �5.41 0 – –5.43 1 Glu23Diosphenol �3.49 0 � �4.29 2 Ser92; Asn68Esculetin �4.36 1 Gly218 �5.34 2 Ser92;Tyr98Eugenol �6.12 0 � �1.64 1 Glu67Fraxetin �4.57 0 � �0.87 1 Gly94Gallic Acid �3.15 2 Asp67, Asp216, �5.63 1 Ser92Geraniol �4.64 1 Arg148 �4.25 2 Tyr98, Ser92Geranyl Acetate �4.95 2 Asp198, Asp216 �5.46 1 Asn68Herniarin �5.07 1 Gly218 �5.07 1 Glu23Isohamnetin �4.7 1 Arg148 �4.77 0 0Kaempferol �5.54 1 Gln35 �5.08 1 Met1Ledol �2.88 2 Asp67, Arg148 �3.48 1 Met 46Limonene �4.25 1 Arg148 �3.51 2 His109, Val89Linalool �4.6 2 Asp67, Phe271 �4.01 2 Ser92, Ser92Linalyl Acetate �4.77 2 Asp198, Asp198 �5.48 1 Ile10Lutein �4.67 2 Asp216 �5.55 1 Asp106Luteolin �6.42 2 Asn 88, Ser 10 �5.03 2 Ala3, Gly94Melatonin �4.61 2 Asp67,Gln35 �5.79 1 Ser92Menthol �4.68 1 Arg71 �5.64 1 Ser92Methyl Benzoate �4.36 1 Asp67 �4.88 1 Ala3Methylanisole �4.69 1 Asp198 �5.74 1 Val69Methylisoeugenol �5.18 0 � �5.24 1 Tyr98Methylpyrrolidine �5.25 1 Arg148 �5.62 0 0Myrcene �6.2 1 Asp216 �3.58 1 Glu 23Myrtenol �5.05 1 Lys55 �5.25 1 Tyr98Neocnidilide �4.49 1 Arg219 �3.9 1 Glu23Neryl Acetate �3.46 1 Arg67 �3.37 1 Arg52Niacin �6.39 0 � �5.67 0 0Ocimene �5.3 1 Arg148 �3.48 1 Gly94Oleuropein �4.93 0 0 0 0 0Osthol �3.28 1 Arg148 �4.14 1 Val69Palmatine �5.21 1 Arg67 �1.44 1 Arg219Phellandrene �4.3 1 Arg71 �0.27 1 Arg71Pinene �4.76 2 Arg219, Arg219 �3.43 1 Glu23Pinocarvone �3.78 1 Asp216 �4.21 0 �Pulegone �3.49 0 � �3.46 1 Ser92Pyrocide �2.46 0 � �3.07 0 �Resveratrol �4.39 0 � �7.58 2 Glu 23, Ser 92Sabenene �5.3 0 � �2.08 0 �Scopoletin �4.93 0 � �5.43 0 �Senkyunolide �1.28 0 � �3.51 0 �Sotolon �4.21 0 � �1.01 0 �Taraxacin �4.3 0 � �4.46 1 Phe 271Terpinolene �4.66 1 Gln35 �4.55 0 �Thujone �3.38 0 � �3.03 0 �Thymol �4.09 0 � �4.69 1 Arg148Trans�Jasmone �4.3 0 � �3.64 1 Pro 82

(Continued)

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 7

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 9: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

7.3% residues are in additionally allowed region(Figure 3). The quality of the model was estimated as58.38%. The stereochemical analysis of vancomycinresistance protein (vanH), potential drug target ofS. aureus, showed that 91.2% of residues are in the mostfavored region (Figure 4) and 8.3% residues are inadditionally allowed region. The quality of the modelwas found to be 67. 84% by protein structure verificationalgorithm, ERRAT (Colovos & Yeates, 1993), whichevaluates the progress of crystallographic model buildingand refinement. This parogram analyzes the statistics ofnonbonded interactions between different atom types.From our results, it is evident that all the four predictedmodels have good stereochemical validity and highaccuracy. Our models satisfy most of the criteria andregulations required for judgment of good-quality models(Hollingsworth & Karplus, 2010; Hooft, Sander, &

Vriend, 1997; Kolaskar & Sawant, 1996; Zhang &Skolnick, 1998). The hypothetical models of all thefour drug targets were submitted to PMDB database(Castrignanò et al., 2006) and can be downloaded bytheir unique accession numbers; PM0078109 (aph),PM0078110 (dfrA1, S. typhi), PM0078111 (vanH), andPM0078112 (dfrA1, V. cholerae).

The drug likeness features of various phytoligandsare shown in Table 3. Most of the herbal ligands selectedwere obeying Lipinski’s rule of five, however, showedsmall variations in CMC-like rule and Lead-like rule.Similarly, most of the molecules are in the 90% cut-offrange for WDI-like rule and showed midstructure forMDDR-like rule. The predicted drug likenesses revealedthat some of the molecules have shown better druglikeliness and those qualified most of the rules werescreened for molecular docking studies. Similarly, the

Table 2. (Continued)

Ligand

Interaction with Aminoglycoside phosphotransferase(aph) Interaction with dihydrofolate reductase (dhfr)

BindingEnergy (Kcal/

mol)

Number ofhydrogenbonds

Amino acidinteracting withligand

Binding energy(Kcal/mol)

Number ofhydrogenbonds

Amino acidinteracting withligand

Trigonelline �4.93 0 � �2.81 0 �Umbelliferone �2.28 0 � �3.55 1 Met1Valeric Acid �4.21 1 Asp216 �2.03 0 �Verbenene �3.3 1 Asp67 �3.19 1Violaxanthin �3.76 1 Arg148 �4.64 0 �Wogonin �3.78 1 Asp216 �7.28 2 Ala 3, Gly 93Zingerone �3.49 0 Thr98 �2.98 0 �

Figure 6. Molecular interaction between dihydrofolate reductase (dhfr) of S. typhi and the best inhibitors are shown the figure asstick figures (a) Herbal ligand Resveratrol binding with the receptor at Glu 23 and Ser 92 by two hydrogen bonds (binding energy -7.58 Kcal/ mol). (b) Interaction between Wogonin and dfrA1 is stabilized by two hydrogen bonds and the interacting amino acids areAla 3 and Gly 94 (binding energy be �7.28 Kcal/mol).

8 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 10: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

pharmacokinetic features were also predicted (Supple-mentary materials, Table 3). From our results, it isevident that few molecules are suitable in terms of theirpharmacokinetic features such as human intestinalabsorption, Caco2 cell permeability, MDCK cell perme-ability, skin permeability, and blood barrier penetration.

Skin permeability rate is a crucial parameter for thetransdermal delivery of drugs and for the risk assessmentof all chemicals that come into contact with the skin

either accidentally or by design (Singh & Singh, 1993).PreADMET predicts in vitro skin permeability and theresults explained in terms of logKpKp (cm/h) is definedas

Kp ¼ km�D=h

where Km is the distribution coefficient between stratumcorneum and vehicle, and D is the average diffusioncoefficient (cm2/h), and h is the thickness of skin (cm)

Figure 7. Docked structures of dihydrofolate reductase type 1(dfrA1) of V. cholerae and the best inhibitors (a) Ligand Herniarinbinding with the receptor at Ser 97 and Gly 98 by two hydrogen bonds (binding energy -8.06 kcal/mol). (b) Interaction betweenPyrocide and dfrA1 is stabilized by a hydrogen bond formed at Tyr 103 (binding energy �8.93 kcal/mol).

Figure 8. Docked pose of methicillin resistance protein (mecI) of S. aureus and the best inhibitors (a) Taraxacin was found to bebest inhibitor, the ligand binds at Lys 89 and Trp with two hydrogen bonds (binding energy -7.28 Kcal/mol). (b) Luteolin identifiedas another best inhibitor which binds at Ala 101 and Tyr 102 by two hydrogen bonds (binding energy �7.58 Kcal/mol).

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 9

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 11: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

(Singh & Singh, 1993). The threshold value <1 indicateslow skin permeability, but the values between one andtwo, and >2 revealed middle and high transdemal perme-ability, respectively. In the present study, except resvera-trol, the skin permeability threshold values of the mostselected molecules were found to be low. The predictedtoxicity results of selected molecules revealed that mostof them are noncarcinogens in mouse models, however,predicted as mutagens. Similarly, the carcinogenicitypredictions for most of the molecules in rat models werepredicted as positive results (Supplementary material,Table 4). Most of the lead molecules described in thepresent study are naturally derived compounds; hence,spontaneous mutations are more prevalent. There arereports that revealed that examination of the mutagenic-ity of naturally occurring leads may indicate theirrelevance of a test compound's mutagenicity. However,many life-threatening or severe debilitating diseases mayjustify treatment with mutagenic or even carcinogenictherapeutic agents (Clive, 1985).

Out of the 58 molecules docked against the fiveproteins, Baicalein (5, 6, 7-trihydroxyflavone), a type offlavonoid, commonly present in the root of Scutellariabaicalensis, and Luteolin (2- (3, 4- Dihydroxyphenyl)-5,7-dihydroxy-4-chromenone), another flavanoid presentin Terminalia chebula showed the best interactions withaminoglycoside phosphotransferase (aph, virulent factorfor Kanamycin resistance) of S. typhi. These ligandsqualified all the rules for drug likeness (Supplementarymaterials, Table 2) and showed better pharmacokineticfeatures (Supplementary materials, Tables 3 and 4).Hence, these can be screened as promising drugcandidates. The binding energy of aph- Baicalein dockedcomplex was estimated to be �6.39 kcal/mol and the

interactions were stabilized by two hydrogen bonds (Thr98, Thr 99). The interacting residues present in thebinding cavity are Phe 56, Pro 82, Thr 98, Thr 99, Thr105, Ile 205, and Ile 215 (Figure 5(a)). Similarly, thebinding energy of aph- Luteolin was estimated to be-6.42 kcal/ mol and the interactions were stabilized bytwo hydrogen bonds (Asn 88 and Ser 10). The mainresidues associated with the interactions are Asn 88, Glu14, Ser 10, Lys 89, and Trp 13 (Figure 5(b)). From ourstudies, it is evident that these phytoligands have signifi-cant binding and inhibitory properties toward kanamy-cin-resistant protein (Table 2). Recent studies revealedthat these phytoligands have good inhibitory propertiestoward various MDR pathogens and their probable drugtargets (Cushnie & Lamb, 2005; Eumkeb et al., 2012).

Similarly, Resveratrol (3, 5, 4′-trihydroxystilbene), anatural phytoalexin commonly found in Vitis vinifera(grape) and Wogonin, an oxy-methylated flavone foundin S. baicalensis (baikal skullcap) showed significantinhibitory activities against dihydrofolate reductase (dhfr,gene product responsible for Trimethoprim resistance) ofS. typhi. From our preliminary screening, it is evidentthat these molecules are potential inhibitors as both thesewere well qualified for druggish and ADME features(Supplementary materials, Table 3). The binding energyof Resveratrol towards dfrA1 was identified as�7.58 kcal/ mol and the interaction was stabilized bytwo hydrogen bonds (Glu 23, Ser 92). The main interact-ing residues present in the binding cavity are Met 1, Val2, Glu 23, Leu 26, Phe 27, Ile 30, Ser 92, His 109, Ile110, Ser 111, and Tyr 146 (Figure 6(a)). The bindingenergy of Wogonin against dhfr was estimated to be�7.28 kcal/ mol. The interactions were stabilized by twohydrogen bonds (Ala 3, Gly 93) and the main residues

Figure 9. Inhibitory activity of herbal ligands against vancomycin resistance protein (vanH) of S. aureus (a) The interaction betweenApigenin and VanH protein is stabilized by two hydrogen bonds formed at Tyr 102, Leu 200 (binding energy �6.07 Kcal/mol). (b)Luteolin binds with the receptor at Gln 35, Asp 198, and Asp 64 by three hydrogen bonds. The binding energy of the interaction wasfound to be 6.32Kcal/mol.

10 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 12: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Table 3. Docking interaction between Vancomycin-resistant (vanH) protein, virulent factor for Vancomycin resistance, andMethicillin-resistant protein (mecI), virulent factor for methicillin resistance, of S. aureus, and the selected lead molecules. Apigeninand Luteolin against vanH, and Taraxacin and Luteolin against mecI were identified as the best inhibitors based on better bindingefficiency. The best ligands are highlighted.

Ligand

Vancomycin-resistant protein (vanH) Methicillin-resistant protein (mecI)

BindingEnergy (Kcal/

mol)

Number ofhydrogenbonds

Amino acidinteracting withligand

Bindingenergy (Kcal/

mol)

Number ofHydrogenbonds

Amino acidinteracting withligand

Allicin �2.61 1 Asn101 �4.07 1 Tyr102Apigenin �6.07 2 Tyr 102, Leu 200 �5.59 0 0Baicalein �4.58 0 0 �2.65 1 Leu200Borneol �5.63 1 Tyr6 �4.99 0 0Deoxyhexose �4.98 1 Thr82 �4.15 1 Tyr102Diosphenol �4.32 1 Asn88 �4.34 2 Val202, Arg 231Esculetin �4.97 1 Lys89 �3.81 1 Tyr102Eugenol �4.89 1 Asn85 �1.68 1 Val202Fraxetin �3.87 1 Lys89 �4.85 1 Leu200Gallic acid �4.49 1 Asn85 �3.75 1 Arg231Geraniol �3.85 1 Lys89 �4.62 1 Leu200Geranyl acetate �4.37 0 0 �6.58 0 0Herniarin �3.68 2 Trp13,Ile8 �4.65 0 0Isohamnetin �4.41 1 Lys89 �2.32 1 Thr 229Kaempferol �4.43 1 Ser10 �3.11 1 Arg 231Ledol �4.46 1 Lys89 �4.75 1 Asp 255Limonene �4.49 1 Lys89 �4.35 1 Ala 294Linalool �3.74 1 Glu7 �4.17 1 His 201Linalyl acetate �4.76 2 Thr82, Phe86 �3.03 1 Tyr 102Lutein �5.26 1 Glu104 �3.74 1 His 201Luteolin �6.32 3 Gln 35, Asp 198,

Asp 64�7.58 2 Ala 101, Tyr 102

Melatonin �3.96 1 Thr82 �5.91 2 Tyr102, His201Menthol �4.60 0 � �3.69 1 Asp255Methyl benzoate �4.69 2 Thr82,Thr82 �4.67 1 Tyr102Methylanisole �4.34 2 Lys89,Ile8 �4.97 0 �Methylisoeugenol �4.80 1 Ile8 �5.46 2 His201, His292Methylpyrrolidine �3.64 0 – �4.41 1 Leu200Myrcene �4.15 1 Ile8 �4.59 1 Tyr102Myrtenol �1.31 1 Lys79 �4.43 1 Asp255Neocnidilide �5.93 0 0 �0.76 1 His201Neryl Acetate �4.71 1 Lys89 �4.94 1 Tyr102Niacin �7.24 1 Asn17 �4.78 1 Val202Ocimene �4.1 1 Asn85 �1.17 1 His201Oleuropein �4.28 0 � �5.71 2 Tyr102, Leu200Osthol �3.84 0 � �5.36 1 His292,Tyr102Palmatine �3.56 1 Ser10 �4.03 1 Asp255Phellandrene �5.57 2 Trp13, Lys89 �4.65 1 HIS201Pinene �3.72 1 Lys89 �4.39 0 �Pinocarvone �4.33 2 Lys89,Lys89 �3.66 1 Gly94Pulegone �4.32 1 Ile8 �3.63 1 Ser92Pyrocide �2.18 1 � �1.97 0 �Resveratrol �1.64 0 � �4.42 1 Val69Sabenene �4.17 1 Ser10 �3.43 1 Tyr98Scopoletin �1.33 1 Asp216 �4.19 1 Ile 157Senkyunolide �4.90 0 � �4.73 2 Thr 229, Gly 230Sotolon �3.71 1 Asn88 �3.76 2 Trp13, Ile8Taraxacin �4.14 1 Asp67 �7.28 2 Trp 13, Lys 89Terpinolene �2.11 1 Arg148 �4.78 0 �Thujone �3.28 0 – �2.17 1 Thr 229Thymol �3.04 0 � �5.71 1 His292,Tyr102Trans-jasmone �3.76 1 Glu7 �5.06 1 Asp255,Tyr295Trigonelline �3.17 0 � �2.03 1 His201

(Continued)

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 11

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 13: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

present in the binding cavities are Val 2, Ala 3, Ile 10,Phe 27, Tyr 42, Met 46, Ser 92, Gly 93, Gly 94, andTyr 98 (Figure 6(b), Table 2). The antimicrobial effectsof Resveratrol and Wogonin against various bacterialpathogens and their toxins have been studied (Chan,2002; Docherty, Fu, & Tsai, 2001; Schrader, 2010),which revealed that these phytochemicals have signifi-cant inhibitory properties toward virulent factors of manyMDR pathogens.

Herniarin (7-O-methylumbelliferone), a coumarinfound in Herniaria glabra (smooth rupturewort) andPyrocide [(3-Allyl-2-methyl-4-oxo-2-cyclopenten-1-yl 2,2-dimethyl-3-(2-methyl-1-propen-1-yl) cyclopropanecarb-oxylate)], a common flavone present in Daucus carota(carrot) showed the best binding activity towardsdihydrofolate reductase (dfrA1, Trimethoprim-resistantprotein) of V. cholerae. From our preliminary screening,it is evident that Herniarin and Pyrocide showed betterdrug-like features and pharmacokinetic properties(Supplementary materials, Tables 3 and 4). The dockedcomplex of dfrA1-Herniarin was stabilized by twohydrogen bonds (Ser 97, Gly 98) with binding energy of�8.06 kcal/mol. The main residues present in the bindingcavity of dfrA1 were Met 6, Val 7, Glu 23, Phe 32, Thr47, Met 52, Gly 95, Gly 96, Ser 97, Tyr 103, and Glu128 (Figure 7(a)). Studies on the antifungal and antibac-terial activities of various Herniarin derivatives(Céspedes et al., 2006) revealed that these phytoligandsshowed good inhibitory activities against various entericbacterial pathogens. Similarly, the interaction betweenPyrocide and dfrA1 was stabilized by a hydrogen bond(Tyr 103) with binding energy of -8.93 kcal/mol. Val 7,Ile 15, Pro 19, Asp 20, Glu 28, Phe 32, Ile 51, Met 52,Thr 47, Gly 99, and Tyr 103 are the main residues pres-ent in the binding cavity (Figure 7(b)) (Table 4). ThoughPyrocide exhibits better binding energy (8.93 kcal/40mol), only Tyr 103 is interacting with the receptor.Hence, better simulation studies are necessary to screenthis ligand and the present data pave significant insightfor such studies.

Luteolin and Taraxacin (3,5,8-Trimethyl-9,9a-dihydroazuleno[6,5-b]furan-2,7-dione), a sesquiterpene

guaianolide present in Taraxacum officinale (weber),showed better binding properties toward mecI protein(gene code for Methicillin resistance) of S. aureus. Ourin silico studies revealed that Luteolin and Taraxacinshowed good drug likeliness and pharmacokineticsfeatures (Supplementary materials, Tables 3 and 4).Computer-aided simulation studies revealed that thedocked complex of mecI and Luteolin were stabilized bytwo hydrogen bonds (Ala 101, Tyr 102) with bindingenergy of �7.58 kcal/mol. The main residues present inthe active sites were found to be Ile 80, Ala 101, Tyr102, Ile 157, His 201, Thr 229, and Gly 230 (Figure 8(a)). Similarly, Taraxacin binds with mecI by the forma-tion of two hydrogen bonds (Trp 13, Lys 89) with thebinding energy of -7.28 kcal/mol. The main residuespresent in the binding cavity were identified as Ile 8, Lys89, Trp13, Thr 82, and Asn 85 (Figure 8(b), Table 3).We have noticed that Gly, Lys, His, and Thr are the con-served residues that play a major role in receptor–ligandinteraction as reported recently (Kahlon, Darokar, &Sharma, 2012). However, Taraxacin was not qualified forCMC-like rule and Lead-like rule. CMC-like rule defineddrug-like character for the CMC database, whichremoved several classes of compounds such as diagnos-tic imaging agents, solvents, and pharmaceutical aids(Oprea, Davis, Teague, & Leeson, 2001). Similarly, lead-like rule defined to consider designing libraries withdrug-like physicochemical properties such as high-affinity leads, lead-like leads, and drug-like leads (Ghose,Viswanadhan, & Wendoloski, 1999). From our studies, itis evident that these phytochemicals have significantinhibitory properties toward probable drug targets ofMDR pathogens. Many studies revealed the inhibitoryproperties of Taraxacin and Luteolin (Ahmad et al.,2000) against various pathogenic microorganisms bydifferent mechanisms.

Apigenin (4′, 5, 7-trihydroxyflavone), a flavonefound in Coffee arabica (coffee), and Luteolin werefound to be interacted against vanH (gene for Vancomy-cin resistance) protein. In silico prediction of druglikeliness and ADMET features were found to besuitable for Apigenin and Luteolin (Supplementary

Table 3. (Continued)

Ligand

Vancomycin-resistant protein (vanH) Methicillin-resistant protein (mecI)

BindingEnergy (Kcal/

mol)

Number ofhydrogenbonds

Amino acidinteracting withligand

Bindingenergy (Kcal/

mol)

Number ofHydrogenbonds

Amino acidinteracting withligand

Umbelliferone �1.72 1 Gln35 �3.65 �Valeric acid �1.35 0 � �4.68 2 Leu200, Arg231Verbenene �4.33 1 Glu23 �4.45 2 Val202, Leu200Violaxanthin �3.82 0 � �4.15 1 Tyr102Wogonin �3.33 0 � �3.62 0 �Zingerone �1.12 0 � �2.03 1 Tyr102

12 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 14: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Table 4. Docking interaction between dihydrofolate reductase (dfrA1), virulent factor for Trimethoprim resistance of V. cholerae andselected inhibitors. Herniarin and Pyrocide were identified as the best inhibitors based on the binding energy. The best ligands arehighlighted.

Ligand

Interaction with dihydrofolate reductase (dfrA1)

Binding energy (Kcal/mol) Number of hydrogen bonds Amino acid interacting with ligand

Allicin �5.97 2 Ala8, Asp20Apigenin �4.08 2 Glu28, Glu28Baicalein �5.71 1 Glu28Borneol �4.19 1 Ser97Deoxyhexose �5.28 1 Tyr103Diosphenol �5.67 0 0Esculetin �3.12 2 Tyr103,Tyr103Eugenol �3.57 1 Glu28Fraxetin �4.78 0 0Gallic Acid �4.61 0 0Geraniol �5.22 0 0Geranyl Acetate �3.51 2 Ala8, Gly99Herniarin �8.06 2 Ser 97, Gly 98Isohamnetin �5.2 1 Ser97Kaempferol �5.58 0 0Ledol �5.14 1 Glu28Limonene �5.48 1 Tyr103Linalool �5.09 1 Ser97Linalyl Acetate �6.06 2 Gly99, Ser97Lutein �5.61 2 Ser97, Gln29Luteolin �4.54 1 Ser97Melatonin �5.17 0 0Menthol �4.53 2 Glu28, Ile15Methyl Benzoate �5.09 1 Tyr103Methylanisole �5.41 1 Ser97Methylisoeugenol �4.06 0 0Methylpyrrolidine �5.22 1 Ala8Myrcene �5.83 1 Gln29Myrtenol �4.64 0 –Neocnidilide �5.13 2 Tyr103, Tyr103Neryl Acetate �6.13 1 Glu28Niacin �4.93 0 0Ocimene �5.81 1 Ala8Oleuropein �3.92 1 Ala8Osthol �5.73 0 0Palmatine �3.53 1 Gln29Phellandrene �5.59 1 Ser116Pinene �4.27 0 �Pinocarvone �5.16 1 Ser 97Pulegone �5.68 1 Ile15Pyrocide �8.93 1 Tyr 103Resveratrol �4.63 1 Tyr 6Sabenene �5.06 0 �Scopoletin �4.32 1 Asn 88Senkyunolide �4.19 1 Lys 89Sotolon �4.89 1 Asn 85Taraxacin �3.87 1 Lys 89Terpinolene �4.43 1 Asn 85Thujone �3.85 1 Lys 89Thymol �4.13 0 �Trans�Jasmone �4.98 1 Thr 82Trigonelline �4.83 1 Thr 47Umbelliferone �2.93 0 �Valeric Acid �1.81 1 Ala 101Verbenene �3.62 0 �Violaxanthin �4.75 1 Ile 157Wogonin �4.03 0 �Zingerone �5.52 0 �

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 13

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 15: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

materials, Tables 3 and 4). The docked complex ofvanH–Apigenin was stabilized by two hydrogen bonds(Tyr 102, Leu 200; binding energy �6.07 kcal/mol). Thebinding site contains Ala 101, Thr 102, Val 107, Ile 157,Leu 200, His 201, Val 202, Thr 229, and Gly 230(Figure 9(a)). Similarly, Luteolin interacted with vanHby three hydrogen bonds (Gln 35, Asp 198, and Asp 64;binding energy �6.32 kcal/mol) and the main residuespresent in the active sites are Gln 35, Ser 36, Asp 64,Asp 67, Asp 198, Asp 216, and Arg 219 (Figure 9(b),Table 3). As mentioned in the present study, recentreports revealed that Gly, His, and Asp are the main con-served residues present in the active site of Vancomycin-resistant protein, they have a vital role in receptor–ligandinteraction (Kahlon et al., 2012). The antimicrobialactivities of all these lead molecules are well studied. Asignificant inhibitory property of Apigenin toward drug-resistant Enterobacter cloacae was recently reported incomparison with the known chemotherapeutic agent,Ceftazidime (Eumkeb & Chukrathok, 2013). Fromour simulation studies, we have identified various phytol-igands which showed effective binding and conforma-tional changes in drug targets. The structural featuresand pharmacophoric properties of all these ligands areshown in supplementary materials, Table 6. The bindingefficiency of these phytoligands toward various drug-resistant proteins paves better understanding of theinhibitory mechanism of herbal leads and such studieshave high relevance in clinical and preclinical trials.

Since many bacteria showed high drug resistanceagainst currently prescribed chemotherapeutics, designingof alternative remedies against the drug targets are essen-tial to block the virulent functions. Thus, there is a needto discover new leads and deeper understanding of themechanisms by which bacteria gain resistance to antibi-otics which will aid in identifying novel targets fordrugs. Computer-aided studies serve as ideal platforms toscreen novel inhibitors against these drug targets and thepresent study provides remarkable insights for furtherin vitro and in vivo studies.

4. Conclusion

Most of the notorious pathogens developed resistance toconventional chemotherapeutic agents and emerged asmultidrug-resistant (MDR) and extensive drug-resistant(XDR) strains, “the superbugs”. The emergence of suchpathogenic bacteria has become serious healthcareissue worldwide. Our previous studies on multidrug-resistant pathogens from water samples collected fromByramangala reservoir indicated that S. typhi, Staphyloc-cus aureus, and V. cholerae showed resistance to manyconventionally used drugs such as Trimethoprim,Kanamycin, Vancomycin, and Methicillin. Hence, there isa necessity to address this issue and screen alternative

therapeutic substances. Bacterial drug resistance seems tobe mainly due to virulent genes; inhibition of such geneproducts by appropriate ligands may prevent the virulentfunctions. The three-dimensional structures of most of thevirulent proteins are not available in their native form.Hence, we employed homology modeling to predict thethree-dimensional folding of virulent proteins toward theseantibiotics. We have identified many herbal inhibitorswhich are beneficial in providing a remedy to block theactivity of virulent proteins by structure-based virtualscreening. The present study suggests that active com-pounds from herbal sources exhibit promising inhibitoryroles against these MDR and XDR pathogens. However,further experimental analysis is essential for the conforma-tion of the selected lead molecules.

Acknowledgments

The authors thankfully acknowledge Dr. G S Jagannatha Rao,senior professor and head, Department of Biotechnology,Dayananda Sagar College of Engineering, Bangalore, and Dr.G A Ravishankar, vice president, (R & D) in Life Sciences,Dayananda Sagar Institutions, Bangalore for their constantsupport and encouragement throughout the study.

Supplementary material

The supplementary material for this paper is availableonline at http://dx.doi.10.1080/07391102.2013.819787.

ReferencesAdaramoye, O. A., Anjos, R. M., Almeida, M. M., Veras, R.

C., Silvia, D. F., Oliveira, F. A., … Medeiros, I. A. (2009).Hypotensive and endothelium-independent vasorelaxanteffects of methanolic extract from Curcuma longa L. inrats. Journal of Ethnopharmacology, 124, 457–462.

Ahmad, V. U., Yasmeen, S., Ali, Z., Khan, M. A., Choudhary,M. I., Akhtar, F., Miana, G. A., & Zahid, M. (2000). Tarax-acin, a new guaianolide from Taraxacum wallichii. Journalof Natural Products, 63, 1010–1011.

Ajay, A., Walters, W. P., & Murcko, M. A. (1998). Can we learnto distinguish between “drug-like” and “nondrug-like” mol-ecules? Journal of Medicinal Chemistry, 41, 3314–3324.

Altschul, S. F., Madden, T. L., Schäffer, A. A., Zhang, J., Zhang,Z., Miller, W., & Lipman, D. J. (1997). Gapped BLAST andPSI-BLAST: A new generation of protein database searchprograms. Nucleic Acids Research, 25, 3389–3402.

Apweiler, R., Bairoch, A., Wu, C. H., Barker, W. C., Boeck-mann, B., Ferro, S., & … (2004). UniProt: The UniversalProtein knowledgebase. Nucleic Acids Research, 32, D115–D119.

Avallone, R., Zanoli, P., Puia, G., Kleinschnitz, M., Schreier,P., & Baraldi, M. (2000). Pharmacological profile of apige-nin, a flavonoid isolated from Matricaria chamomilla. Bio-chemical Pharmacology, 59, 1387–1394.

Baananou, S., Bouftira, I., Mahmoud, A., Boukef, K.,Marongiu, B., & Boughattas, N. A. (2012). Antiulcerogenicand antibacterial activities of Apium graveolens essential oiland extract. Natural Product Research. doi:10.1080/14786419.2012.717284.

14 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 16: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Bao, L. J., Maruya, K. A., Snyder, S. A., & Zeng, E. Y.(2012). China’s water pollution by persistent organic pollu-tants. Environmental Pollution, 163, 100–108.

Bennett, P. M. (2008). Plasmid encoded antibiotic resistance:acquisition and transfer of antibiotic resistance genes in bac-teria. British Journal of Pharmacology, 153, S347–S357.

Berman, H., Henrick, K., Nakamura, H., & Markley, J.L.(2007). The worldwide Protein Data Bank (wwPDB):Ensuring a single, uniform archive of PDB data. NucleicAcids Research, 35, D301–D303.

Bhadra, K., & Kumar, G. S. (2011). Interaction of berberine,palmatine, coralyne, and sanguinarine to quadruplex DNA:A comparative spectroscopic and calorimetric study. Biochi-mica et Biophysica Acta, 1810, 485–496.

Buscemi, N., Vandermeer, B., Hooton, N., Pandya, R.,Tjosvold, L., Hartling, L., … Baker, G. (2006). Efficacyand safety of exogenous melatonin for secondary sleep dis-orders and sleep disorders accompanying sleep restriction:meta-analysis. British Medical Journal, 332, 385–393.

Carugo, O. (2007). Statistical validation of the root-mean-square-distance, a measure of protein structural prox-imity. Protein Engineering, Design & Selection, 20, 33–37.

Castrignanò, T., De Meo, P. D., Cozzetto, D., Talamo, I. G., &Tramontano, A. (2006). The PMDB protein modeldatabase. Nucleic Acids Research, 34, 306–309.

Céspedes, C. L., Avila, J. G., Martínez, A., Serrato, B., Calde-rón-Mugica, J. C., & Salgado-Garciglia, R. (2006). Anti-fungal and antibacterial activities of Mexican tarragon(Tagetes lucida). Journal of Agriculture and Food Chemis-try, 54, 3521–3527.

Chan, M. M. (2002). Antimicrobial effect of resveratrol ondermatophytes and bacterial pathogens of the skin. Bio-chemical Pharmacology, 63, 99–104.

Chaudhari, L. K., Jawale, B. A., Sharma, S., Sharma, H.,Kumar, C. D., & Kulkarni, P. A. (2012). Antimicrobialactivity of commercially available essential oils againstStreptococcus mutans. Journal of Contemporary DentalPractice, 13, 71–74.

Chen, L. J., Lebetkin, E. H., & Burka, L. T. (2001). Metabo-lism of R pulegone in F344 rats. Drug Metabolism andDisposition, 29, 1567–1577.

Chen, C. Y., Lindsey, R. L., Strobaugh, T. P.Jr., Frye, J. G.,Meinersmann, R. J., Chen, C. Y., … Meinersmann, R.J.(2010). Prevalence of ColE1-like plasmids and kanamycinresistance genes in Salmonella enterica serovars. Appliedand Environment Microbiology, 76, 6707–6714.

Clive, D. (1985). Mutagenicity in drug development: Interpreta-tion and significance of test results. Regulatory Toxicologyand Pharmacology, 5, 79–100.

Colovos, C., & Yeates, T. O. (1993). Verification of proteinstructures: patterns of nonbonded atomic interactions.Protein Science, 2, 1511–1519.

Cushnie, T. P., & Lamb, A. J. (2005). Antimicrobial activity offlavonoids. International Journal of Antimicrobial Agents,26, 343–356.

Daniels, R. (2011). Surviving the first hours in sepsis: Gettingthe basics right (an intensivist’s perspective). Journal ofAntimicrobial Chemotherapy, 66, 11–23.

Davies, J., & Davies, D. (2010). Origins and evolution ofantibiotic resistance. Microbiology and Molecular BiologyReviews, 74, 417–433.

de Almeida, R. N., Araújo, D. A., Gonçalves, J. C., Montenegro,F. C., de Sousa, D. P., Leite, J. R., & … (2009). Rosewoodoil induces sedation and inhibits compound action potentialin rodents. Journal of Ethnopharmacology, 124, 440–443.

de Sousa, E., Zanatta, L., Seifriz, I., Creczynski-Pasa, T. B.,Pizzolatti, M. G., Szpoganicz, B., & … (2004). Hypoglyce-mic effect and antioxidant potential of kaempferol-3,7-O-(alpha)-dirhamnoside from Bauhinia forficata leaves.Journal of Natural Products, 67, 829–832.

Dey, Y. N., Ota, S., Srikanth, N., Jamal, M., & Wanjari, M. (2012).A phytopharmacological review on an important medicinalplant – Amorphophallus paeoniifolius Ayu., 33, 27–32.

Docherty, J. J., Fu, M. M., & Tsai, M. (2001). Resveratrol selec-tively inhibits Neisseria gonorrhoeae and Neisseria meningiti-dis. Journal of Antimicrobial Chemotherapy, 47, 214–243.

Dong, P., Zhang, Y., Gu, J., Wu, W., Li, M., Yang, J., … Liu,Y. (2011). Wogonin, an active ingredient of Chinese herbmedicine Scutellaria baicalensis, inhibits the mobility andinvasion of human gallbladder carcinoma GBC-SD cells byinducing the expression of maspin. Journal of Ethnophar-macology, 137, 1373–1380.

Esmaeili, A., & Hashemi, E. (2011). Biotransformation of myr-cene by Pseudomonas aeruginosa. Chemistry Central Jour-nal 5, 26. doi: 10.1186/1752-153X-5-26

Eswar, N., Eramian, D., Webb, B., Shen, M. Y., & Sali, A.(2008). Protein structure modeling with modeller. Methodsin Molecular Biology, 426, 145–159.

Eumkeb, G., & Chukrathok, S. (2013). Synergistic activity andmechanism of action of ceftazidime and apigenin combina-tion against ceftazidime-resistant Enterobacter cloacae.Phytomedicine, 20, 262–269.

Eumkeb, G., Siriwong, S., & Thumanu, K. (2012). Synergisticactivity of luteolin and amoxicillin combination againstamoxicillin-resistant Escherichia coli and mode of action.Journal of Photochemistry and Photobiology, 117, 247–253.

Frimurer, T. M., Bywater, R., Naerum, L., Lauritsen, L. N., &Brunak, S. (2000). Improving the odds in discriminating“drug-like” from “non drug-like” compounds. Journal ofChemical Information and Computer Sciences, 40,1315–1324.

García-Castellanos, R., Marrero, A., Mallorquí-Fernández, G.,Potempa, J., Coll, M., & Gomis-Ruth, F. X. (2003). Three-dimensional structure of MecI. Molecular basis for transcrip-tional regulation of staphylococcal methicillin resistance.Journal of Biological Chemistry, 278, 39897–39905.

Gerson, T., Hawke, J. C., & Shorland, F. B. (1960). The roleof n-valeric acid in the synthesis of the higher saturatedstraight-chain acids containing an odd number of carbonatoms in bovine milk fat. Biochemical Journal, 74,366–368.

Ghose, A. K., Viswanadhan, V. N., & Wendoloski, J. J. (1999).A knowledge-based approach in designing combinatorial ormedicinal chemistry libraries for drug discovery. 1. A quali-tative and quantitative characterization of known drugdatabases. Journal of Combinatorial Chemistry, 1, 55–68.

Giedraitienė, A., Vitkauskienė, A., Naginienė, R., & Pavilonis,A. (2011). Antibiotic resistance mechanisms of clinicallyimportant bacteria. Medicina (Kaunas), 47, 137–146.

Gilani, A. H., Janbaz, K. H., & Shah, B. H. (1998). Esculetinprevents liver damage induced by paracetamol and CCL4.Pharmacological Research, 37, 31–35.

Giménez, B. G., Santos, M. S., Ferrarini, M., & Fernandes, J.P. (2010). Evaluation of blockbuster drugs under the rule-of-five. Pharmazie, 65, 148–152.

Hagedorn, C., Robinson, S. L., Filtz, J. R., Grubbs, S. M.,Angier, T. A., & Reneau, R. B. (1999). Determiningsources of fecal pollution in a rural Virginia watershed withantibiotic resistance patterns in fecal streptococci. Appliedand Environment Microbiology, 65, 5522–5531.

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 15

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 17: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

He, X., Liu, & , H. W. (2002). Mechanisms of enzymatic Cbond O bond cleavages in deoxyhexose biosynthesis.Current Opinion in Chemical Biology, 6, 590–597.

Hernández-Ortega, M., Ortiz-Moreno, A., Hernández-Navarro,M.D., Chamorro-Cevallos, G., Dorantes-Alvarez, L., &Necoechea-Mondragón, H. (2012). Antioxidant, antinoci-ceptive, and anti-inflammatory effects of carotenoidsextracted from dried pepper (Capsicum annuum L.). Jour-nal of Biomedical and Biotechnology, 2012, 524019. doi:10.1155/2012/524019

Hettne, K. M., Williams, A. J., van Mulligen, E. M., Kleinjans,J., Tkachenko, V., & Kors, J. A. (2010). Automatic vs.manual curation of a multi-source chemical dictionary: theimpact on text mining. Journal of Cheminformatics, 2, 3–7.

Hiramatsu, K., Asada, K., Suzuki, E., Okonogi, K., & Yokota,T. (1992). Molecular cloning and nucleotide sequencedetermination of the regulator region of mecA gene inmethicillin-resistant Staphylococcus aureus (MRSA). FEBSLetters, 298, 133–136.

Hollingsworth, S. A., & Karplus, P. A. (2010). A fresh look atthe Ramachandran plot and the occurrence of standardstructures in proteins. Biomolecular Concepts, 1, 271–283.

Holm, L., Kääriäinen, S., Rosenström, P., & Schenkel, A.(2008). Searching protein structure databases with DaliLitev. 3. Bioinformatics, 24, 2780–2781.

Hooft, R. W., Sander, C., & Vriend, G. (1997). Objectivelyjudging the quality of a protein structure from a Ramachan-dran plot. Computer Applications in the Biosciences, 13,425–430.

Jonkers, D., Sluimer, J., & Stobberingh, E. (1999). Effect ofgarlic on vancomycin-resistant enterococci. AntimicrobialAgents and Chemotherapy, 43, 3045.

Kabuto, H., & Yamanushi, T. T. (2011). Effects of zingerone[4-(4-hydroxy-3-methoxyphenyl)-2-butanone] and eugenol[2-methoxy-4-(2-propenyl) phenol] on the pathologicalprogress in the 6-hydroxydopamine-induced Parkinson'sdisease mouse model. Neurochemical Research, 36,2244–2249.

Kahlon, A. K., Darokar, M. P., & Sharma, A. (2012). Probingthe evolutionary conserved regions within functional site ofdrug-resistant target proteins of Staphylococcus aureus: Insilico phylogenetic motif profiling approach. Indian Journalof Biochemistry & Biophysics, 49, 442–450.

Kamkumo, R. G., Ngoutane, A. M., Tchokouaha, L. R., Fokou,P. V., Madiesse, E. A., Legac, J., … Rosenthal, P.J. (2012).Compounds from Sorindeia juglandifolia (Anacardiaceae)exhibit potent anti-plasmodial activities in vitro and in vivo.Malaria Journal, 11, 382. doi: 10.1186/1475-2875-11-382

Katsiki, M., Chondrogianni, N., Chinou, I., Rivett, A. J., &Gonos, E. S. (2007). The olive constituent oleuropeinexhibits proteasome stimulatory properties in vitro and con-fers life span extension of human embryonic fibroblasts.Rejuvenation Research, 10, 157–172.

King, D. J., Gleadow, R. M., & Woodrow, I. E. (2006). Regu-lation of oil accumulation in single glands of Eucalyptuspolybractea. New Phytologist, 172, 440–451.

Kolaskar, A. S., & Sawant, S. (1996). Prediction of conforma-tional states of amino acids using a Ramachandran plot.International Journal of Peptide and Protein Research, 47,110–116.

Konig, T., Gutsche, B., Hartl, M., Hubscher, R., Schreier, P., &Schwab, W. (1999). 3-Hydroxy-4, 5-dimethyl-2(5H)-furanone (Sotolon) causes an off-flavor: elucidation of

its formation pathways during storage of citrus soft drinks.Journal of Agriculture and Food Chemistry, 47,3288–3291.

Laskowski, R. A., Rullmann, J. A., MacArthur, M. W.,Kaptein, R., & Thornton, J. M. (1996). AQUA andPROCHECK-NMR: programs for checking the quality ofprotein structures solved by NMR. Journal of BiomolecularNMR, 8, 477–486.

Leite, J. R., Seabra Mde, L., Maluf, E., Assolant, K.,Suchecki, D., Tufik, S., … Carlini, E. A. (1986). Pharma-cology of lemongrass (Cymbopogon citratus Stapf). III.Assessment of eventual toxic, hypnotic and anxiolyticeffects on humans. Journal of Ethnopharmacology, 17,75–83.

Leung, A. Y. (1980). Encyclopedia of common natural ingredi-ent used in food, drugs and cosmetics. New York, NY:Wiley.

Lothrop, H. D., Lothrop, B., & Reisen, W. K. (2002). Noctur-nal microhabitat distribution of adult Culex tarsalis(Diptera: Culicidae) impacts control effectiveness. Journalof Medical Entomology, 39, 574–582.

Lupo, A., Coyne, S., & Berendonk, T. U. (2012). Originand evolution of antibiotic resistance: the common mech-anisms of emergence and spread in water bodies. Fron-tiers in Microbiology, 3, 18. doi: 10.3389/fmicb.2012.00018

Maclean, R. C., Hall, A. R., Perron, G. G., & Buckling, A.(2010). The evolution of antibiotic resistance: insight intothe roles of molecular mechanisms of resistance and treat-ment context. Discoveery Medicine, 10, 112–118.

Maglott, D., Ostell, J., Pruitt, K. D., & Tatusova, T. (2011).Entrez Gene: Gene-centered information at NCBI. NucleicAcids Research, 39, D52–D57.

Marcuzzi, A., Crovella, S., & Pontillo, A. (2011). Geraniolrescues inflammation in cellular and animal models ofmevalonate kinase deficiency. In Vivo, 25, 87–92.

Martínez, J. L. (2012). Natural antibiotic resistance andcontamination by antibiotic resistance determinants:The two ages in the evolution of resistance to antimicrobi-als. Frontiers in Microbiology, 3, 1–3.

Molina-Jimenez, M. F., Sanchez-Reus, M. I., & Benedi, J.(2003). Effect of fraxetin and myricetin on rotenone-induced cytotoxicity in SH-SY5Y cells: Comparison withN-acetylcysteine. European Journal of Pharmacology, 472,81–87.

Moraes, M. C., Birkett, M. A., Gordon-Weeks, R., Smart, L.E., Martin, J. L., Pye, B. J., … Pickett, J. A. (2008). Cis-Jasmone induces accumulation of defence compounds inwheat, Triticum aestivum. Phytochemistry, 69, 9–17.

Morsy, M. A., & Fouad, A. A. (2008). Mechanisms of gastro-protective effect of eugenol in indomethacin-induced ulcerin rats. Phytotheraphy Research, 22, 1361–1366.

Nazir, T., Abraham, S., & Islam, A. (2012). Emergence ofpotential superbug Mycobacterium tuberculosis, lessonsfrom New Delhi mutant-1 bacterial strains. InternationalJournal of Health Science (Qassim), 26, 87–94.

Nishioka, T., Kawabata, J., & Aoyama, Y. (1998). Baicalein, analpha-glucosidase inhibitor from Scutellaria baicalensis.Journal of Natural Products, 61, 1413–1415.

Notredame, C., Higgins, D. G., & Heringa, J. (2000).T-Coffee: A novel method for fast and accurate multiplesequence alignment. Journal of Molecular Biology, 302,205–217.

16 S. Skariyachan et al.

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3

Page 18: Exploring insights for virulent gene inhibition of multidrug resistant Salmonella typhi , Vibrio cholerae , and Staphylococcus areus by potential phytoligands via in silico screening

Oliveira, D. A, Salvador, A. A., Smânia, A. Jr., Smânia, E. F.,Maraschin, M., & Ferreira, S. R. (2012). Antimicrobialactivity and composition profile of grape (Vitis vinifera)pomace extracts obtained by supercritical fluids. Journal ofBiotechnology. doi:10.1016/j.jbiotec.2012.09.014.

Oprea, T. I., Allu, T. K., Fara, D. C., Rad, R. F., Ostopovici,L., & Bologa, C. G. (2007). Lead-like, drug-like or “Pub-like”: how different are they? Journal of Computer-AidedMolecular Design, 21, 113–119.

Oprea, T. I., Davis, A. M., Teague, S. J., & Leeson, P. D.(2001). Is there a difference between leads and drugs? Ahistorical perspective. Journal of Chemical Information andComputer Sciences, 41, 1308–1315.

Ow, Y. Y., & Stupans, I. (2003). Gallic acid and gallic acidderivatives: Effects on drug metabolizing enzymes. CurrentDrug Metabolism, 4, 241–248.

Paulsen, E., Otkjaer, A., & Andersen, K. E. (2010). Thecoumarin herniarin as a sensitizer in German chamomile[Chamomilla recutita (L.) Rauschert, Compositae]. ContactDermatitis, 62, 338–342.

Peana, A. T., D'Aquila, P. S., Panin, F., Serra, G., Pippia, P., &Moretti, M. D. (2002). Anti-inflammatory activity oflinalool and linalyl acetate constituents of essential oils.Phytomedicine, 9, 721–726.

Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S.,Greenblatt, D. M., Meng, E. C., & … (2004). UCSF chimera-a visualization system for exploratory research and analysis.Journal of Computational Chemistry, 25, 1605–1612.

Qiu, J., Luo, M., Dong, J., Wang, J., Li, H., Wang, X. …Deng, X. (2011). Menthol diminishes Staphylococcus aur-eus virulence-associated extracellular proteins expression.Applied Microbiology Biotechnology, l90, 705–712.

Rahman, S., Parvez, A. K., Islam, R., & Khan, M. H. (2011).Antibacterial activity of natural spices on multiple drugresistant Escherichia coli isolated from drinking water,Bangladesh. Annals of Clinical Microbiology and Antimi-crobiology, 10, 10. doi: 10.1186/1476-0711-10-10

Reimer, A. R., Van Domselaar, G., Stroika, S., Walker, M.,Kent, H., Tarr, C., … V. cholerae Outbreak Genomics TaskForce (2011). Comparative genomics of Vibrio choleraefrom Haiti, Asia, and Africa. Emerging Infectious Diseases,17, 2113–2121.

Rossi, P. G., Bao, L., Luciani, A., Panighi, J., Desjobert, J. M.,Costa, J., & … (2007). (E)-Methylisoeugenol and elemicin:antibacterial components of Daucus carota L. essential oilagainst Campylobacter jejuni. Journal of Agriculture andFood Chemistry, 55, 7332–7336.

Rúa, J., Fernández-Álvarez, L., de Castro, C., Del Valle, P., deArriaga, D., & García-Armesto, M. R. (2012). Antibacterialactivity against foodborne Staphylococcus aureus and anti-oxidant capacity of various pure phenolic compounds.Foodborne Pathogens and Disease, 8, 149–157.

Salari, M. H., Amine, G., Shirazi, M. H., Hafezi, R., &Mohammadypour, M. (2006). Antibacterial effects of Euca-lyptus globulus leaf extract on pathogenic bacteria isolatedfrom specimens of patients with respiratory tract disorders.Clinical Microbiology & Infection, 12, 194–196.

Schrader, K. K. (2010). Plant natural compounds with antibacterialactivity towards common pathogens of pond-cultured channelcatfish (Ictalurus punctatus). Toxins (Basel), 2, 1676–1689.

Semenya, S., Potgieter, M., Tshisikhawe, M., Shava, S., &Maroyi, A. (2012). Medicinal utilization of exotic plants byBapedi traditional healers to treat human ailments in Limp-opo province, South Africa. Journal of Ethnopharmacology,44, 646–655.

Shapiro, S., & Guggenheim, B. (1995). The action of thymolon oral bacteria. Oral Microbiology and Immunology, 10,241–246.

Singh, S., & Singh, J. (1993). Transdermal drug delivery bypassive diffusion and iontophoresis: a review. MedicinalResearch Reviews, 13, 569–621.

Sirois, J. C., & Miller, R. W. (1972). The mechanism of thescopoletin-induced inhibition of the peroxidase catalyzeddegradation of indole-3-acetate. Plant Physiology, 49,1012–1018.

Skariyachan, S., Lokesh, P., Rao, R., Kumar, A. U., Vasist, K.S., & Narayanappa, R. (2013). A pilot study on water pol-lution and characterization of multidrug-resistant superbugsfrom Byramangala tank, Ramanagara district, Karnataka,India. Environmental Monitoring Assessment, 185, 5483–5495

Skariyachan, S., Prakash, N., & Bharadwaj, N. (2012). In silicoexploration of novel phytoligands against probable drugtarget of Clostridium tetani. Interdisciplinary Science, 4,273–281.

Sun, J. (2007). D-Limonene: Safety and clinical applications.Alternative Medicine Review, 12, 259–264.

Taguchi, H., Sakaguchi, M., & Shimabayashi, Y. (1985). Trigo-nelline content in coffee beans and the thermal conversionof trigonelline into nicotinic acid during the roasting ofcoffee beans. Agricultural and Biological Chemistry, 49,3467–3472.

Toyama, D. O., Filho, D. E. B., Cavada, B. S., da Rocha,B. A., de Oliveira, S. C., Cotrim, C. A., … Toyama, M. H.(2011). Umbelliferone induces changes in the structure andpharmacological activities of Bn IV, a phospholipase A (2)isoform isolated from Bothrops neuwiedi. Toxicon, 57,851–860.

Trott, O., & Olson, A. J. (2010). AutoDock Vina: improvingthe speed and accuracy of docking with a new scoringfunction, efficient optimization, and multithreading. Journalof Computational Chemistry, 31, 455–461.

Varlet, V., & Augsburger, M. (2012). Monitoring of aglyconsof yew glycosides (3, 5-dimethoxyphenol, myrtenol and1-octen-3-ol) as first indicator of yew presence. Drug Testand Analysis, 5, 474–479.

Veber, D. F., Johnson, S. R., Cheng, H. Y., Smith, B. R.,Ward, K. W., & Kopple, K. D. (2002). Molecularproperties that influence the oral bioavailability of drugcandidates. Journal of Medicinal Chemistry, 45, 2615–2623.

Walsh, T. R., Weeks, J., Livermore, D. M., & Toleman, M. A.(2011). Dissemination of NDM-1 positive bacteria in theNew Delhi environment and its implications for humanhealth: An environmental point prevalence study. TheLancet Infectious Disease, 11, 355–362.

Wang, Q., & Xie, M. (2010). Antibacterial activity and mecha-nism of luteolin on Staphylococcus aureus. Sheng Wu GongCheng Xue Bao (Chinese Journal of Biotechnology), 50,1180–1184.

Weigel, L. M., Clewell, D. B., Gill, S. R., Clark, N. C., McDo-ugal, L. K., Flannagan, S. E., & … (2003). Genetic analy-sis of a high-level vancomycin-resistant isolate ofStaphylococcus aureus. Science, 302, 1569–1571.

Zhang, C., & Lai, L. (2011). Towards structure-based proteindrug design. Biochemical Society Transactions, 39, 1382–1386.

Zhang, L., & Skolnick, J. (1998). What should the Z-scoreof native protein structures be? Protein Science, 7,1201–1207.

Exploring insights for virulent gene inhibition of multidrug resistant pathogens 17

Dow

nloa

ded

by [

122.

172.

193.

255]

at 0

7:33

26

July

201

3