Top Banner
Intestinal cholecystokinin and leptin signaling and the regulation of glucose production By Brittany Anne Rasmussen A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Physiology University of Toronto © Copyright by Brittany Anne Rasmussen (2015)
214

Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

Apr 22, 2020

Download

Documents

dariahiddleston
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: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

Intestinal cholecystokinin and leptin

signaling and the regulation of glucose production

             

By

Brittany Anne Rasmussen

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Graduate Department of Physiology University of Toronto

© Copyright by Brittany Anne Rasmussen (2015)

Page 2: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

ii

Title: Intestinal cholecystokinin and leptin signaling and the regulation of glucose

production

Degree: Doctor of Philosophy

Year of Convocation: 2015

Full Name: Brittany Anne Rasmussen

Department: Physiology, University of Toronto

General Abstract

The number of individuals affected by diabetes is on the rise due in part to lifestyle

and/or genetic factors. Diabetes and obesity are characterized by a disruption in glucose

homeostasis due in part to an elevation in glucose production (GP). It is of utmost importance to

understand the regulation of GP in normal, obese and diabetic settings in hopes to unveil

therapeutic targets that lower blood glucose concentrations in diabetes and obesity.

The small intestine has also been documented to regulate glucose homeostasis

independent of changes in food intake although the intestinal signaling mechanism(s) remain

largely unknown. Specifically, the duodenum senses an increase in lipids and triggers release of

CCK and activates the CCK1 receptor in the duodenum to lower GP via a gut-brain-liver axis.

However, the downstream intestinal CCK1 receptor signaling effectors remain unknown. In

study 1 of this thesis, the signaling molecule PKA was shown, for the first time to our

knowledge, to lie downstream of the duodenal CCK1 receptor to trigger vagal afferent firing

and a gut-brain-liver axis to lower GP. Importantly, direct activation of duodenal PKA lowered

GP and bypassed duodenal CCK resistance in high fat fed rats.

Like the duodenum, the distal part of the small intestine, the jejunum, also senses lipids

to lower GP via a gut-brain-liver axis, but whether hormonal action mediates this GP-lowering

Page 3: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

iii

effect of the jejunum remains unknown. In study 2 of this thesis, a novel GP-lowering effect of

leptin in the jejunum was described. Specifically, jejunal leptin activated the long form leptin

receptor and PI3K to lower GP in normal, high fat fed or uncontrolled diabetic rodents via a

neuronal network, and contributed to the early anti-diabetic effect of bariatric surgery.

In conclusion, this doctoral thesis demonstrates that independent activation of duodenal

CCK-PKA and jejunal leptin-PI3K signaling potently lowers GP in normal, high-fat fed and

diabetic rodents via a gut-brain-liver neuronal axis. Thus, targeting hormonal (i.e., CCK and

leptin) signaling in the small intestine represents a potential therapeutic strategy to lower GP and

restore glucose homeostasis in diabetes and obesity, and may mimic the anti-diabetic effect of

bariatric surgery.

Page 4: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

iv

Acknowledgements

I would first like to thank my supervisor Dr. Tony Lam who has constantly challenged

me throughout my studies to help me become an independent and critical thinker. I appreciate

all the effort you have made in helping me become the best I can be academically and for your

support in my career decisions. I am also thankful for my committee members, Dr. Adria Giacca

and Dr. Khosrow Adeli who have taught me to think on the spot, think critically, and search for

the best explanation. Their knowledge and wisdom continues to inspire me throughout every

committee meeting and presentation I give on my research. I would not be where I am today

without the support of the Lam Lab. I have met some incredible people along this journey who

have acted not only as mentors but also as great friends. I would like to especially thank Dr.

Danna Breen, for all of the laughs we shared as well as challenging me to become a better

student. I would also like to thank the rest of the “gut” team, Clémence Côté, Melika Zadeh

Tahmesabi, Dr. Frank Duca, Sophie Hamr and Paige Bauer. You have been an amazing team to

work with and have also provided moral support throughout my studies. I seriously could not

have done it without you guys! I would also like to thank the rest of the lab members, Penny

Wang, Elena Burdett, Claire Yang, Patricia Mighiu, Dr. Jessica Yue, Dr. Beatrice Filippi, Mona

Abraham, Mary LaPierre, and Beini Wang. It has been wonderful getting to know each and

everyone of you and your help and support was always appreciated. My family is of utmost

importance to me and I would like to extend a big thank you to my parents, Rodney and Patricia

Rasmussen, who have never questioned my career choices and have supported me with in any

decisions I have made. Also to by brothers Ryan and Jordi (and Loy and John) and sister Lauren

for their continued support throughout my academic career. Lastly, to my husband Oliver. All I

can say is that I would have never made it to this point without you and I love you very much.

You have always provided comfort and support and I am forever grateful to have you in my life.

Page 5: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

v

Table  of  Contents    Introduction  ......................................................................................................................  1  Chapter  1

1.1 Obesity and Diabetes  ................................................................................................................................  1  1.2 The small intestine and the regulation of metabolic homeostasis  ...................................................  6  

1.2.1 Local gut-brain paracrine effect versus the endocrine effect of gut-derived hormones  ...............  6  1.3 Gastrointestinal Peptides  .........................................................................................................................  9  

1.3.1 Gastric Peptides  ....................................................................................................................................................  9  1.3.2 Proximal Intestinal Peptides  ...........................................................................................................................  15  1.3.3 Distal Intestinal Peptides  .................................................................................................................................  20  

1.4 Small intestine control of glucose production through a gut-brain-liver neuronal axis  .........  30  1.4.1 Duodenal lipid sensing and CCK secretion triggers a gut-brain-liver axis to lower glucose production  ........................................................................................................................................................................  30  1.4.2 Jejunal nutrient sensing triggers a gut-brain-liver axis to lower glucose production  ..................  37  

1.5 Bariatric surgery, gut hormones and intestinal nutrient sensing  ................................................  39  1.5.1 Bariatric surgical procedures and changes in gut hormones  ................................................................  39  1.5.2 Duodenal jejunal bypass surgery, nutrient sensing and beyond  .........................................................  44  

1.6 Summary of Introduction  .....................................................................................................................  49  1.6 Rationale and Significance of the Studies  .........................................................................................  49  1.7 General Hypothesis  ................................................................................................................................  51  1.8 Specific Aims  ...........................................................................................................................................  51  

 General  Methods  ..........................................................................................................  52  Chapter  22.1 Animals  .....................................................................................................................................................  52  

2.1.1 High Fat Feeding Animal Model  ..................................................................................................................  52  2.2 Surgical Procedures  ...............................................................................................................................  52  

2.2.1 Vessel Cannulation  ............................................................................................................................................  53  2.2.2 Intestinal Cannulation  .......................................................................................................................................  53  

2.3 Pancreatic Euglycemic (Basal Insulin) Clamp Technique  ............................................................  54  2.4 Protein Assay  ...........................................................................................................................................  55  2.5 Biochemical Analyses  .............................................................................................................................  56  

2.5.1 Plasma Glucose  ...................................................................................................................................................  56  2.5.2 Plasma Glucose Tracer Specific Activity  ..................................................................................................  56  2.5.3 Plasma Insulin  .....................................................................................................................................................  57  

2.6 Calculations  .............................................................................................................................................  58  2.7 Statistical Analysis  ..................................................................................................................................  58  

 Study  1  .............................................................................................................................  60  Chapter  33.1 Abstract  ....................................................................................................................................................  61  3.2 Introduction  .............................................................................................................................................  62  3.3 Materials and Methods  ..........................................................................................................................  64  

3.3.1 Animal Preparation  ............................................................................................................................................  64  3.3.2 Animal Surgeries  ................................................................................................................................................  64  3.3.3 Intraduodenal Infusions and Treatments  ....................................................................................................  66  3.3.4 Pancreatic Euglycemic (Basal Insulin) Clamp Technique in Rats  ....................................................  66  3.3.6 PKA Activity Assay  ..........................................................................................................................................  68  3.3.7 PCR methods  .......................................................................................................................................................  69  3.3.9 Biochemical Analysis  .......................................................................................................................................  71  3.3.10 Calculations and Statistical Analysis  ........................................................................................................  71  

3.4 Results  .......................................................................................................................................................  72  3.4.1 Direct activation of PKA lowers glucose production  ............................................................................  72  3.4.2 Activation of PKA lowers glucose production via a vagal afferent firing  ......................................  73  

Page 6: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

vi

3.4.3 Activation of NR1-containing NMDA receptors is required for duodenal PKA to lower glucose production  ........................................................................................................................................................  74  3.4.4 Duodenal PKA activation requires brain to liver communication to lower glucose production  .............................................................................................................................................................................................  76  3.4.5 CCK lowers glucose production via PKA activation  .............................................................................  76  3.4.6 The CCK1 receptor fails to activate PKA after short term high fat feeding  ..................................  77  

3.5 Discussion  .................................................................................................................................................  78    Study  2  .............................................................................................................................  97  Chapter  4

4.1 Abstract  ....................................................................................................................................................  98  4.2 Introduction  .............................................................................................................................................  99  4.3 Materials and Methods  ........................................................................................................................  100  

4.3.1 Animal Preparation  .........................................................................................................................................  100  4.3.2 Animal Surgeries  .............................................................................................................................................  103  4.3.3 Intraintestinal infusions and treatments  ...................................................................................................  105  4.3.4 Pancreatic (Basal Insulin) Euglycemic Clamp Technique  ................................................................  106  4.3.5 Rat [3–3H] glucose infusion protocol (non-clamped conditions)  ....................................................  107  4.3.6 Fasting and refeeding protocol  ...................................................................................................................  108  4.3.7 Gut tissue collection and preparation for western blotting and enzymatic activity assay  ......  108  4.3.8 Western blotting  ..............................................................................................................................................  109  4.3.9 RNA extraction, reverse transcription and PCR methods  .................................................................  110  4.3.10   PI3K Activity Assay  ...............................................................................................................................  112  4.3.11 Biochemical Analysis  ..................................................................................................................................  113  4.3.12 Calculations and Statistical Analysis  .....................................................................................................  114  

4.4 Results  .....................................................................................................................................................  115  4.4.1 Jejunal leptin requires jejunal leptin receptor activation to lower glucose production  ............  115  4.4.2 A STAT3-independent and PI3K-dependent signaling pathway is required for jejunal leptin to lower glucose production via a neuronal network  ..........................................................................................  117  4.4.3 Jejunal leptin’s action remain intact in high fat fed or diabetic rats  ..............................................  119  4.4.4 The antidiabetic effect of DJB surgery is mediated by jejunal leptin action  ...............................  121  

4.5 Discussion  ...............................................................................................................................................  122    Summary  and  Conclusions  ......................................................................................  144  Chapter  5

5.1 Summary of Studies in this Thesis  ....................................................................................................  144  5.2 General Summary  ................................................................................................................................  145  5.3 General Conclusion  ..............................................................................................................................  145  

 General  Discussion  ....................................................................................................  147  Chapter  66.1 Do nutrient sensing mechanisms interact with both CCK and leptin?  ....................................  147  6.2 What other intestinal hormones share similar signaling mechanisms as CCK and leptin?  149  

6.2.1 PKA  .....................................................................................................................................................................  149  6.2.2 PI3K  .....................................................................................................................................................................  150  

6.3 What is the cellular location of CCK-PKA and leptin-PI3K signaling in the intestine?  ......  151  6.4 What is the relevance of CCK and leptin signaling in disease models?  ...................................  153  

 Limitations  of  the  Studies  ........................................................................................  156  Chapter  7

 Future  Directions  .......................................................................................................  159  Chapter  8  References  ....................................................................................................................  164  Chapter  9

   

Page 7: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

vii

List of Abbreviations

AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin AMPK Adenosine monophosphate activate protein ANOVA Analysis of variances ARC Arcuate nucleus ATP Adenosine trisphosphate B Bound fraction B0 Total binding BB-dp Diabetes-prone BioBreeding rat BBB Blood brain barrier BMI Body mass index BPD/DS Bilio-pancreatic diversion/duodenal switch BSA Bovin serum albumin cAMP Cyclic adenosine monophosphate cAMP-GEFII; Epac2

cAMP guanine nucleotide exchange factor II Exchange protein directly activated by cAMP 2

CaSR CCK1 receptor

Calcium sensing receptor Cholecystokinin 1 receptor

CCK2 receptor Cholecystokinin 2 receptor CD36 Cluster determinant 36 CCK Cholecystokinin CNS Central nervous system CPT-1 Carnitine almitoyltrasnferase-1 DAG diacylglyercol db/db mouse Long form leptin receptor knock out mouse DJB Duodenal jejunal bypass DPP-IV Dipeptidyl peptidase IV DVC Dorsal vagal complex Ex-4 Exendin-4 Ex-9 Exendin-9 Fa/fa rat Lean koletsky rat Fak/fak rat Obese koletsky rat FFA FTO

Free fatty acids Fat mass and obesity associated gene

GCGR GWAS

Glucagon receptor Genome wide association studies

GHSR Growth hormone secretagogue receptor 1a GIP Glucose-dependent insulinotropic peptide GIPR Glucose-dependent insulinotropic peptide receptor GLP-1 GLP-1R

Glucagon like peptide-1 Glucagon like peptide-1 receptor

GLP-2 Glucagon-like peptide-2 GLP-2R Glucagon-like peptide-2 receptor GLUT2 Glucose transporter 2

Page 8: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

viii

GOAT Ghrelin O-acyltransferase gp130 Class I cytokine receptor family GPCR G-protein coupled receptor GPR120 G-protein coupled receptor 120 GPR40 G-protein coupled receptor 40 GSIS Glucose stimulated insulin secretion HFD High fat diet HRP Horseradish peroxidase i.c.v Intracerebroventricular i.p. Intraperitoneal IP3 Inositol triphosphate IRS1 Insulin receptor substrate 1 KATP channels ATP-sensitive potassium channels LAGB Laparoscopic adjustable gastric band LCFA Long chain fatty acids MAPK Mitogen activated protein kinase MUNC18-1 mammalian uncoordinated-18 1 NMDA receptor N-methyl-D-aspartate receptor NPY Neuropeptide y NTS Nucleus of the solitary tract OAG 1-oleoyl-2-acetyl-sn-glycerol Ob-Ra Leptin receptor isoform A Ob-Rb; Leprb Long form leptin receptor Ob-Re Leptin receptor isoform E OXN oxyntomodulin PC Prohormone convertase PI3K Phosphatidylinositol-3-OH kinase PKA Protein kinase A PKC Protein kinase C PLC Phospholipase C POMC Pro-opimelanocortin PYY Peptide YY Ra Rate of appearance Rd RC RIA

Rate of disappearance Regular chow Radioimmunoassay

RYGB Roux-en-Y gastric bypass SD rat Sprague dawley rat SG Sleeve gastrectomy SGLT-1 Sodium glucose transporter-1 SLR Soluble leptin receptor SNARE Soluble NSF Attachment Protein REceptor STAT3 Signal transduction and activator of transcription -3 STAT3 PI Signal transduction and activator of transcription -3 peptide

inhibitor STC-1 cell line Secretin tumor cell-1 cell line STZ Streptozotocin TBST Tris buffered saline-tween VAMP2 Vesicle-associated membrane protein-2

Page 9: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

ix

List of Tables

Table 2.1 Diet content of the regular chow and lard-oil enriched high fat diet. .......................... 59  

Table 3.1 Plasma insulin and glucose concentrations of the groups receiving an intraduodenal infusion during basal and clamp conditions. ....................................................................... 94  

Table 3.2. Plasma insulin and glucose concentrations of the groups receiving both an intraduodenal infusion and DVC infusion during basal and clamp conditions ................... 95  

Table 3.3 Plasma insulin and glucose concentrations of the groups receiving an intraduodenal infusion during basal and clamp conditions. ....................................................................... 96  

Table 4.1 Plasma insulin and glucose concentrations of groups receiving intrajejunal infusions during the basal and clamp conditions .............................................................................. 143  

Page 10: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

x

List of Figures

Figure 1.1 Site of synthesis and secretion of gastrointestinal peptide hormones. …………….....8

Figure 1.2 Duodenal and jejunal nutrient sensing mechanisms trigger a gut-brain-liver neuronal axis to lower glucose production. ………………………………………………………………47

Figure 3.1 Schematic representation of working hypothesis – duodenal Sp-CAMPS activates PKA to lower glucose production, which is abolished upon co-infusion of Sp-CAMPS and H-89 or Rp-CAMPS, and experimental design. .................................................................. 81  

Figure 3.2 Duodenal PKA activation lowers glucose production. .............................................. 82  

Figure 3.3 Schematic representation of working hypothesis – duodenal PKA activation increases vagal afferent firing ............................................................................................................. 83  

Figure 3.4 Direct activation of duodenal PKA increases the spontaneous discharge rate of the mesenteric nerve and inhibits spinal afferent firing of the duodenum. ............................... 84  

Figure 3.5 Schematic representation of working hypothesis – duodenal PKA activation triggers a neuronal network to lower glucose production and experimental design. ........................ 85  

Figure 3.6 Duodenal PKA activation lowers glucose production through a neuronal network. . 86  

Figure 3.7 Schematic representation of working hypothesis – duodenal PKA activation lowers glucose production through a gut-brain-liver neuronal axis and experimental design. ....... 87  

Figure 3.8 Duodenal PKA activation lowers glucose production through activation of the DVC NR1- containing NMDA receptor and hepatic innervation. ................................................ 88  

Figure 3.9 Schematic representation of working hypothesis – Duodenal CCK requires PKA activation to lower glucose production and experimental design. ....................................... 89  

Figure 3.10 Duodenal CCK requires PKA activation to lower glucose production. ................... 90  

Figure 3.11 Schematic representation of working hypothesis – Duodenal CCK fails to suppress glucose production upon high fat feeding, which is rescued upon PKA activation and experimental design ............................................................................................................. 91  

Figure 3.12 Duodenal CCK fails to activate duodenal PKA and lower glucose production after three days of high fat feeding. ............................................................................................. 92  

Figure 3.13 Duodenal Sp-CAMPS activates duodenal PKA activity and lowers glucose production in high fat diet fed rats. ...................................................................................... 93  

Figure 4.1 Schematic representation of the working hypothesis – Gastric leptin activates the intestinal long form leptin receptor to activate a PI3K-dependent and STAT-3 independent signaling axis to lower glucose production through a neuronal network. ......................... 126  

Figure 4.2 Leptin receptor expression in intestinal tissue. ........................................................ 127  

Page 11: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

xi

Figure 4.3 Jejunal leptin administration lowers glucose production. ........................................ 128  

Figure 4.4 Jejunal leptin lowers glucose production independent of changes in portal and circulating leptin levels. ..................................................................................................... 129  

Figure 4.5 Leptin activates leptin receptors to lower glucose production in rats (chemical approach). .......................................................................................................................... 130  

Figure 4.6 Leptin activates leptin receptors to lower glucose production in lean fa/fa rats but not in fak/fak (Kolestky) long form leptin receptor deficient rats (molecular approach). ........ 131  

Figure 4.7 Jejunal leptin activates leptin receptors to lower glucose production in C57BL/6 but not db/db mice (molecular approach). ............................................................................... 132  

Figure 4.8 Jejunal leptin lowers glucose production in C57BL/6 independent of changes in circulating leptin levels. ..................................................................................................... 133  

Figure 4.9 Jejunal leptin lowers glucose production through a STAT3-independent and PI3K dependent pathway. ........................................................................................................... 134  

Figure 4.10 Jejunal and duodenal leptin activate intestinal STAT3, and only jejunal leptin activates jejunal PI3K. ....................................................................................................... 135  

Figure 4.11 Jejunal leptin lowers glucose production through a neuronal network. ................. 136  

Figure 4.12 Jejunal leptin lowers glucose production in high fat diet fed rats. ......................... 137  

Figure 4.13 Jejunal leptin lowers glucose production in high fat diet fed rodents independent of a rise in plasma leptin levels. ............................................................................................. 138  

Figure 4.14 Jejnual leptin lowers plasma glucose levels and glucose production in uncontrolled diabetic rodents independent of changes in plasma insulin and glucagon levels. ............. 139  

Figure 4.15 Jejunal leptin lowers plasma glucose levels and glucose production in uncontrolled diabetic rodents independent of a rise in plasma leptin levels. .......................................... 140  

Figure 4.16. Schematic of duodenal-jejunal bypass (DJB) surgery and jejunal catheter placement. .......................................................................................................................... 141  

Figure 4.17 Jejunal leptin action mediates the rapid anti-diabetic effect of DJB surgery. ........ 142  

Figure 5.1 Summary of duodenal and jejunal hormonal signaling that triggers a neuronal network to lower glucose production ................................................................................ 146  

Page 12: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

xii

Published Manuscripts that Contributed to this Thesis

Review papers: Rasmussen, BA*, Breen, DM* and Lam, TK. Lipid sensing in the gut, brain and liver. Trends Endocrinol Metab 23, 49-55, 2011 *Equal contribution Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: Elsevier Limited Rasmussen, BA*, Breen, DM*, Côté, CD, Jackson, M, and Lam, TK. Nutrient sensing mechanisms in the gut as therapeutic targets for diabetes. Diabetes 62, 3005-3013, 2013 *Equal contribution Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: American Diabetes Association Côté, CD*, Zadeh-Tahmasebi, M*, Rasmussen, BA, Duca, FA, and Lam, TK. Hormonal Signaling in the gut. J Biol Chem, 289, 11642, 2014 *Equal contribution Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: American Society for Biochemistry and Molecular Biology Study 1 (Chapter 3): Rasmussen, BA, Breen, DM, Luo, P, Cheung, GW, Yang, CS, Sun, B, Kokorovic, A, Rong, W, and Lam, TK. Duodenal activation of cAMP-dependent protein kinase induces vagal afferent firing and lowers glucose production in rats. Gastroenterology 142, 834-843, 2012 Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: Elsevier Limited Study 2 (Chapter 4): Rasmussen, BA*, Breen, DM*, Duca, FA, Côté, CD, Zadeh Tahmasebi, M, Filippi, BM, and Lam, TK. Jejunal leptin-PI3K signaling lowers glucose production. Cell Metabolism 19, 1-7, 2014 *Equal contribution Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: Elsevier Limited Other studies contributing to the completion of this thesis: Breen, DM, Yue, JT, Rasmussen, BA, Kokorovic, A, Cheung, GWC, and Lam, TK. Duodenal PKC-δ and cholecystokinin signaling axis regulates glucose production. Diabetes 60, 3148-3153, 2011

Page 13: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

xiii

Breen, DM, Rasmussen BA, Kokorovic, A, Rennian, W, Cheung, GW, and Lam, TK. Jejunal nutrient sensing is required for duodenal-jejunal bypass surgery to rapidly lower glucose concentrations in uncontrolled diabetes. Nature Medicine 18, 950-955, 2012 Duca, FA, Côté, CD, Rasmussen, BA, Zadeh-Tahmasebi, M, Rutter, GA, Filippi, BM, and Lam, TK. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production. Nature Medicine In Press Côté, CD*, Rasmussen, BA*, Duca, FA*, Zadeh-Tahmasebi, M, Baur, JA, Daljeet, M, Breen, DM, Filippi, BM, and Lam TK. Duodenal Sirt1 activation reverses insulin resistance through a neuronal network in rats. Nature Medicine In Press *Equal contribution

Page 14: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

1

Chapter 1Introduction

1.1 Obesity and Diabetes

The incidence of individuals who are obese or overweight has more than doubled since

1980 with 2.1 billion people worldwide being either overweight (BMI ≥ 25) or obese (BMI ≥

30) as of 20131. More alarmingly, the number of children and adolescents (ages 2-19) that are

obese or overweight has risen since 1980 in both boys and girls in developing and developed

countries1, which will likely persist into adulthood2. Obesity is a serious risk factor for life

threatening co-morbidities such as cardiovascular disease, hypertension, type 2 diabetes, cancer

and premature mortality3 and costs the Canadian Healthcare System an average of $5.5 billion

annually4. Given that the number of obese and overweight individuals is predicted to increase1,

it is of utmost importance to understand the pathogenesis of this disease in hopes to reduce its

associated health and economic burdens.

Under normal physiological conditions mammals achieve a remarkably stable body

weight by maintaining energy homeostasis by matching overall energy intake and expenditure

over long periods of time. This tight homeostatic regulation is traditionally believed to involve a

complex integration of acute and chronic metabolic, neural and hormonal factors. For example,

postprandial release of gut hormones activate a local paracrine gut-brain axis5 and gut-brain-

brown fat neuronal axis6 to acutely regulate food intake and energy expenditure, respectively,

where circulating endocrine chronic signals such as insulin and leptin control the overall

metabolic state of adipose stores7,8 as well as feeding9 and energy expenditure10 via the central

nervous system (CNS). Obesity is caused by a shift in energy balance, favoring increased energy

Page 15: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

2

intake and decreased expenditure due to disruptions in the aforementioned gut-brain food intake

axis11–16, as well as central insulin17–21 and leptin resistance22–28. Contributors to these defects

include genetic, environmental and/or social factors. For example, studies have demonstrated

that humans with mutations in genes such as leptin29 (long term energy regulator) or the

melanocortin 4 receptor gene30 (regulates gut peptides release31 thus may acutely regulate

energy homeostasis, as well as chronically regulate adipose stores32) are obese, demonstrating a

monogenic effect on the development of obesity. However, genetic predisposition to obesity in

most individuals is polygenic whereby the presence of genetic variations in multiple genes

contributes to its development. For example, common gene variations associated with increased

BMI are beginning to be uncovered by the genome-wide association studies (GWAS)33 such as

in the MC4R gene and the fat mass and obesity associated gene (FTO)34 (although the FTO link

to obesity is recently debated35). However, given the rapid rise in the incidence of obesity over

the last 50 years, it is unlikely that genetic changes are the main culprit for the obesity pandemic,

and indeed, identical twins are discordant for obesity36. As such, it is more likely that changing

environmental and lifestyle factors are the primary engines of the current pandemic. For

example, daily stress as well as social habits and cues all play a significant role in both daily

consumption (or overconsumption in the case of obesity including increased intake of energy

dense foods) as well as decreased energy expenditure (or reduced physical activity in the case

for obesity, although still debated37,38). Taken together, obesity is likely resultant of a complex

interplay of environmental factors and each individual’s genetic susceptibility to these factors,

which leads to a disruption in energy homeostasis.

Despite the complex and multifactorial etiology of this disease, recent advancements in

the understanding of the pathogenesis of obesity has led to the development of drug therapies

aimed at reducing energy intake and/or increasing energy expenditure. Many have demonstrated

moderate to substantial weight loss such as Orlistat (which inhibits pancreatic lipase and thus

Page 16: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

3

lipid absorption from the gut thereby reducing caloric intake) and sibutramine (a serotonin

reuptake inhibitor which reduces energy intake and increases energy expenditure39) among

others. However, these therapeutic options come with mild side effects such as uncontrolled and

oily bowel movements (seen with orlistat treatment) as well as more serious side effects such as

cardiovascular problems (i.e., serotonin releasing agents fenfluramine, dexfenfluramine and

sibutramine, which have now been removed from the market) or depression (such as rimonabant

which activates cannabinoid CB1 receptors in the brain)40,41. Given the limited success and

potential risks of obesity drug treatments, currently the most successful weight loss intervention

is gastric bypass surgery, which has shown significant and sustained clinical improvements such

as decreased body weight and food intake (while the effects on energy expenditure are currently

debated42). However these surgeries are extremely invasive and have various surgical

complications and at times a need for reoperation43. Thus, it remains of utmost importance to

continue to dissect the mechanism(s) underlying the regulation of body weight to unveil

potential targets to develop successful therapies without associated risks and side effects.

Interestingly, surgical intervention techniques44 aimed at weight loss improve

hyperglycemia, the hallmark of diabetes, highlighting the pathological interconnectivity of the

two diseases. Indeed, as mentioned previously, obesity is a primary cause of type 2 diabetes, as

80-90% of type 2 diabetic individuals are obese/overweight in Canada alone45. Diabetes affects

an alarming amount of people, estimated at 382 million worldwide by the International Diabetes

Federation46. More worryingly, similar to obesity, the number of children and adolescents

affected by the disease has also risen47,48. Within Canada, more than 9 million individuals live

with diabetes or prediabetes and it is estimated that diabetes will cost the Canadian healthcare

system $16.9 billion a year by 202049. Understanding the regulation of glucose homeostasis in

both a normal and diabetic setting will begin to uncover targets to restore the regulation of

glycemia.

Page 17: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

4

Similar to energy homeostasis, under normal conditions, glucose homeostasis is tightly

regulated by controlling the rate of glucose production and uptake in both the fasted and fed

state. In the fasting state, glucose levels are maintained by the hormone glucagon, which after its

secretion from pancreatic α cells binds to its receptor on the liver to increase glucose

production50. This works in an opposite fashion to the hormone insulin, produced from

pancreatic β-cells, which prevents hyperglycemia through a suppression of glucose production

and the stimulation of glucose uptake51. In the fasting state, insulin levels remain low as to not

counteract the effect of glucagon and to prevent peripheral glucose uptake. Thus, through a

counterbalance of these two hormones, circulating glucose levels will rise under fasting

conditions to ensure sufficient energy for distribution to various organs.

In direct contrast, intake of a meal leads to exogenous sources of glucose entering the

system from absorption of glucose from the intestinal lumen, changing the state from fasting to

fed conditions. In this fed condition, there now exists both endogenous and exogenous sources

of glucose and the regulation of glucose homeostasis shifts to ensure circulating glucose levels

are not too high. The control of nutrient delivery into the small intestine is based on the gastric

emptying rate which is a major physiological determinant of postprandial glycemia after a meal,

accounting for ~35% of peak glucose concentrations after ingestion of oral glucose in healthy

volunteers52,53. In addition to promoting secretion of incretin hormones, glucose then enters the

circulation to trigger the first phase insulin response whereby insulin is rapidly secreted to reach

an initial short lived peak within 5 to 7 minutes, lasting around 10-15 minutes54. Following this

initial first phase of insulin secretion, the second phase is characterized by a steady and long-

lasting increase in plasma insulin concentrations54 where glucagon secretion is suppressed as to

not counteract the effects of insulin. In addition, nutrient induced gut peptide release locally

activates a gut-brain-liver axis to regulate glucose production, which will be described in detail

later in this introduction. Together, these mechanisms ensure that endogenous glucose

Page 18: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

5

production is suppressed and peripheral glucose uptake is stimulated to account for the

exogenous glucose entering the system and ensure glucose concentrations are maintained in

their normal homeostatic range.

Defects in the aforementioned homeostatic regulation of glucose levels result in fasting

hyperglycemia in type 1 and 2 diabetes. As type 1 diabetes is characterized by an autoimmune

response that destructs insulin producing pancreatic β-cells55, medications aim to increase

insulin levels to lower plasma glucose levels. Type 2 diabetes, the current focus of this thesis, is

characterized by increased glucose production, peripheral insulin resistance (possibly caused

through an increase in circulating fatty acids seen with obesity56), reduced/altered insulin

secretion, and elevated glucagon levels57,58. Given that fasting hyperglycemia in type 2 diabetes

is largely due to an increase in the rate of glucose production59, development of drug therapies

aimed at reducing glucose production may prove efficacious. Indeed, metformin, the most

widely prescribed type 2 diabetic drug, reduces hyperglycemia via a reduction in glucose

production60, however treatment has been associated with gastrointestinal discomfort and lactic

acidosis60. More recently, incretin based therapies, aimed at increasing the levels and action of

gut-derived incretin hormones have proven successful to lower glucose levels61 and body

weight41. Even more effective than pharmacological interventions for glucose control is bariatric

surgery62,63, which lowers glucose production64 and glycemia in association with changes in gut

peptides65. Taken together, the early anti-diabetic effect of these drugs and bariatric surgery

highlights the role of the intestine in the development of obesity and diabetes and its therapeutic

potential as a target site to lower glucose levels to reduce the risk of diabetic complications. The

focus of this current thesis is to characterize the gluco-regulatory role of gut-derived peptides

and how they contribute to the pathogenesis of obesity and diabetes.

Page 19: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

6

In summary, the small intestine has been viewed as an organ that acutely regulates

feeding, nutrient digestion and metabolism via nutrient induced gut peptide release and

subsequent activation of a local paracrine gut-brain axis as well as through an endocrine fashion

activating peripheral and/or CNS targets. While the ability of gut derived hormones to regulate

glycemia via direct tissue action has been largely studied, only more recently has it been

demonstrated that gut-derived peptides locally trigger a gut-brain axis to acutely regulate

glycemia, similar to feeding regulation. In the following section, the contribution of the local

gut-brain axis versus direct tissue endocrine action of gut derived hormones on feeding and

glucose regulation will be discussed.

1.2 The small intestine and the regulation of metabolic homeostasis  

1.2.1 Local gut-brain paracrine effect versus the endocrine effect of gut-derived hormones

The gastrointestinal tract is the first point of contact between nutrients and the host

whereby initiation of negative feedback mechanisms to maintain metabolic homeostasis first

takes place. The gastrointestinal tract relays information of an incoming meal, such as the size

and composition via both gastric and intestinal signals, which are integrated within the CNS,

more specifically the hindbrain, to ultimately reduce food intake. More specifically, the

hindbrain integrates signals of both chemical (endocrine) and neural origin (via

mechanoreceptors or local activation of neurons innervating the intestine). For example, nutrient

delivery into the stomach can be sensed by mechanoreceptors that detect tension66, stretch67 and

volume68, and these mechanical signals are relayed to the brain via spinal and spinal nerves. In

addition to gastric mechanical signals, both the stomach and small intestine can send chemical

signals to relay nutritional status via secretion of gut-derived hormones. More specifically, the

inner lining of the GI tract houses a single layer of epithelial cells containing specialized

enteroendocrine (EEC) cells, which express nutrient sensing elements on the apical side.

Page 20: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

7

Following a meal, activation of the nutrient sensing elements leads to the triggering of

intracellular signaling pathways, which results in the depolarization of the cell membrane

causing the release of gut hormones expressed within the EEC. These hormones can directly

enter the bloodstream to act within the periphery to control food intake and pass through the

leaky blood brain barrier to target the central nervous system directly. In contrast, these gut

peptides can act in a paracrine fashion to activate their corresponding receptors on vagal

afferent terminals innervating the small intestine to trigger a gut-brain neuronal axis to acutely

control feeding and metabolism. Indeed, intestinal nutrient infusions reduce food intake within

minutes suggesting activation of local intestinal signals is required for the acute effects on

feeding rather than postabsorptive affects69.

Similar to appetite regulation, nutrient induced secretion of gut-derived hormones has

been demonstrated to regulate glycemia via endocrine actions via either inhibition or stimulation

of the release of insulin or glucagon directly or via their action within the CNS to control

glucose production or pancreatic hormone release70. In addition, nutrients in the preabsorptive

state can activate sensing mechanisms in the small intestine via local gut peptide release and

subsequent activation of a gut-brain negative feedback system to regulate gastric emptying71 and

thus control the rate of glucose entry into the blood, as well as inhibit glucose production by the

liver69 to acutely regulate glucose levels. The purpose of the current thesis is to dissect the local

paracrine effect of gut-derived hormones on the regulation of glucose homeostasis.

The next focus will review the paracrine versus endocrine metabolic regulatory

mechanisms of specific hormones such as from the stomach: leptin and ghrelin, from the

duodenum/jejunum: CCK and glucose-dependent insulinotropic peptide (GIP) (and possibly L

cell derived hormones), and from the ileum: glucagon-like peptide-1/2 (GLP-1/2),

oxyntomodulin (OXN), and peptide YY (PYY) (Figure 1.1).

Page 21: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

8

Figure 1.1 Site of synthesis and secretion of gastrointestinal peptide hormones

Shown is the site of synthesis as well as secretion of both stomach and small intestinal derived hormones. The stomach produces and secretes ghrelin and leptin. The duodenum and jejunum synthesize and secrete CCK and GIP. It is currently debated whether the duodenum contains L cells and thus synthesizes and secretes GLP-1/2, OXN and PYY, which are more commonly thought to arise from the ileum. Adapted from Côté, CD*& Zadeh-Tahmasebi, M* et al. Hormonal Signaling in the gut. J Biol Chem, 289, 11642, 2014 *Equal contribution. Permission to reproduce this figure has been obtained from the copyright owner: American Society for Biochemistry and Molecular Biology

Page 22: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

9

1.3 Gastrointestinal Peptides

1.3.1 Gastric Peptides

1.3.1.1 Ghrelin

Ghrelin is a 28 amino acid peptide expressed in various tissues such as the stomach,

intestine, pituitary, pancreas, kidney, lung, ovaries and brain72. The stomach is the major source

of ghrelin, which is synthesized in endocrine X/A cells of the gastric mucosa73 and is highly

concentrated in the fundic region74,75. In order to become active, ghrelin must undergo multiple

cleavage steps starting as a 117 amino acid pre-prohormone. First, the removal of a secretory

signal peptide at its N-terminus and cleavage at its C-terminus by prohormone convertase

(PC)1/3 is needed to result in a prohormone76,77. Second, ghrelin undergoes esterification by an

acyl-transferase, ghrelin O-acyltransferase (GOAT)78 becoming acylated-ghrelin (AG), which

accounts for approximately 10-20% of circulating ghrelin79. It is traditionally believed that only

after acylation by GOAT that ghrelin binds to its widely expressed receptor, the growth

hormone secretagogue receptor 1a (GHSR)80, however recent data suggests that des-acyl ghrelin

(originally believed to be a non-active form) may also bind to the GHSR to exert biological

effects81. With respect to nutrient induced regulation of ghrelin secretion, glucose, amino acids,

and lipids can all suppress ghrelin secretion. However, carbohydrates are its most potent

suppressor, followed by proteins and lipids82,83.

After its secretion, ghrelin acts as a “hunger hormone”. This is due to the fact that an

increase in ghrelin levels has been associated with timing of a meal in both rodents and

humans84,85, peaking at meal initiation followed by a postprandial decrease back to baseline72.

Indeed, peripheral and central administration of ghrelin increases food intake in both rodents

and humans86–88 which is abolished upon intraperitoneal (i.p.) co-administration of a GHSR

antagonist or anti-ghrelin immunoglobulin, instead resulting in a decrease in food intake89,90.

Given these findings, it is evident that ghrelin may act in an endocrine and/or paracrine fashion.

Page 23: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

10

Indeed, ghrelin can cross the blood brain barrier (BBB) where it is thought to activate different

regions of the brain to control food intake, including the hypothalamus91,92 and brain stem93,94

which has been widely studied (recently reviewed in95). Alternatively, vagal afferents that

innervate the stomach express the ghrelin receptor and in both rodents and humans, and

vagotomy (which eliminates communication between the stomach and brain) abolishes the

ability of ghrelin to increase food intake96,97, demonstrating a local paracrine effect of ghrelin on

feeding regulation. Thus, it is evident that ghrelin induces feeding, whether through its intestinal

and/or brain action. Likewise, ghrelin also down regulates receptor expression for anorexigenic

peptides such as PYY, GLP-1, and CCK98,99, further emphasizing its orexigenic effects.

Not only does ghrelin regulate feeding, but it also plays a role in the maintenance of

blood glucose levels in the fasting condition. In rodents, administration of AG can cause a rapid

inhibition of glucose stimulated insulin secretion (GSIS)100, through its direct action on the

pancreas101. This is strengthened by the findings that blockade of AG102 and the GHSR101

improves the insulin response to a glucose challenge, and that the GHSR is located in pancreatic

islets103–107. These findings have been translated to humans, where administration of higher

doses of AG suppressed GSIS108. In contrast to its peripheral actions, central ghrelin works in an

opposite fashion by acting as a positive regulator of insulin secretion81,109. This implies that

peripheral AG action may counteract the hyperinsulinemic action of central AG. However,

future studies are needed to better understand the direct versus indirect effects of ghrelin on

GSIS. In addition to glucose regulation via pancreatic β cell insulin release, α cell secretion of

glucagon can also regulate glycemia. Traditionally ghrelin is thought to have no affect on

glucagon secretion110, however recent literature suggests the presence of the GHSR in α cells in

mice and demonstrates secretion of glucagon following AG administration to mouse islets111. In

contrast to the regulation of insulin secretion, central ghrelin does not appear to affect glucagon

levels109.

Page 24: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

11

In addition to the regulation of insulin secretion, studies suggest that ghrelin may also

affect insulin sensitivity. Indeed, ghrelin and ghrelin receptor knock out mice exhibit decreased

body weight and insulin levels, while being more insulin sensitive112. In line with these findings,

administration of AG in humans caused insulin resistance in association with a rise in

circulating FFA113,114. In contrast to these results, in mice undergoing the hyperinsulinemic

euglycemic clamp studies, AG administration improved peripheral, but not hepatic insulin

sensitivity115. Additionally, both a positive and negative correlation between ghrelin levels and

the incidence of type 2 diabetes and insulin resistance has been reported in humans116–119. Thus,

these studies collectively suggest that the role of ghrelin in the regulation of insulin sensitivity

remains controversial, with recent studies still claiming opposing results120,121.

In contrast to the controversial findings on the regulation of insulin sensitivity by

ghrelin, it is commonly accepted that ghrelin can increase gastric emptying thus increasing the

amount of glucose that can be absorbed by the duodenum and subsequently enter into the

circulation122,123. This effect of ghrelin may be mediated by neuronal mechanisms, as the ability

of ghrelin to increase gastric emptying was abolished when neural communications were

negated by surgical or chemical techniques124 demonstrating that ghrelin can locally regulate

glycemia in the presence of luminal glucose. Thus, in addition to alleviating the inhibition of

ghrelin on GSIS, an increase in the gastric emptying rate stimulated by ghrelin will increase

nutrient release into the duodenum to cause secretion of gut peptides to inhibit further ghrelin

release to prevent an increase in glucose absorption and resultant hyperglycemia.

Taken together, these studies suggest that while ghrelin may act within the periphery to

regulate food intake and glucose homeostasis, its local regulatory actions may be of equal

importance. This suggests that local gut-derived hormonal signals may indeed play an integral

role in mediating metabolic homeostasis, which is a common theme amongst the different

peptide hormones secreted within the gastrointestinal tract.

Page 25: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

12

1.3.1.2 Leptin

In contrast to the well-known stomach derived hormone ghrelin, leptin is less studied in

regards to its synthesis and secretion from the stomach. Although it is conventionally believed

that the hormone leptin is mainly produced by adipose tissue125 and circulates in proportion to

fat mass126, leptin is also produced in the stomach127 as well as the placenta128, skeletal

muscle129, and mammary epithelium130. No matter where its site of secretion, leptin’s effects are

mediated through its long form leptin receptor (Ob-Rb; Leprb) belonging to the class I cytokine

receptor family (also known as the gp130 receptor family)131,132. There are six isoforms of the

receptor that have been identified and termed A-F, including Leprb. All six isomers of the

receptor share the same 805 amino acids at the N-terminus and are products of the db gene133.

The smallest of the receptors is the isoform E (Ob-Re) which does not contain a transmembrane

or cytoplasmic domain but rather is a soluble binding protein, also known as the soluble leptin

receptor (SLR)132. Interestingly, the transcript for this isoform has not been detected in humans,

likely due to differences in post-translational processing134,135. The remaining 5 isoforms contain

a transmembrane domain and a short intracellular portion. It is generally believed that Leprb is

the only isoform capable of signaling by activating molecules such as signal transduction and

activator of transcription-3 (STAT3) and insulin receptor substrate 1/phosphatidylinositol-3-OH

kinase (PI3K) signaling through phosphorylation of tyrosine residues on the receptor136,137.

However, this has recently been challenged by the findings that Ob-Ra may have signaling

capacity138.

Within the stomach, leptin has been localized to pepsinogen secreting gastric chief cells

mainly in the fundic region127,139, as well as in a small number of epithelial cells of the

stomach140. The concentration of leptin in the stomach has been estimated to be around half that

found in adipose tissues in rats of the same age141. In contrast, in humans, the amount has been

found to be double142, however a direct comparison is difficult to make with differences in

Page 26: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

13

sampling between humans and rats. Interestingly, due to the fact that leptin is localized within

different cells of the stomach, it can undergo both endocrine127,139 and exocrine143 secretion.

Indeed, it has been demonstrated that refeeding with a diet containing carbohydrates, protein

and lipids stimulates leptin secretion into the gastric juices within the stomach in both rats127 and

humans139 suggesting that all class of nutrients may cause release of gastric leptin. This

secretion is rapid, where leptin content in the stomach has been shown to rapidly decline within

20 minutes of refeeding127. The exocrine secretion of leptin has been demonstrated by the

finding that leptin from the stomach lumen survives the acidic gastric environment as it has been

measured and detected in the duodenal juice after refeeding143. It is hypothesized that leptin is

synthesized within gastric chief cells at the level of the rough endoplasmic reticulum separately

from the Leprb, where the receptor first undergoes maturation to the soluble leptin receptor

isoform (SLR; Ob-Re) and is then bound to leptin at the level of the secretory granule144. Thus,

upon nutrient stimulation of leptin release, leptin is complexed to its SLR to increase its survival

in the gastric juices. In contrast to these findings, human studies suggest that leptin secreted into

the intestine was not found to be associated with macromolecules142 (such as the SLR) and the

SLR has been suggested to bind to leptin and antagonize its effects145,146. Given that the SLR-

leptin complex does not affect unbound leptin induced activation of the Leprb146, it remains to be

assessed whether the amount of free versus bound leptin is regulated by nutritional status. Such

findings will clarify how leptin reaches the intestinal lumen intact.

In regards to food intake regulation, studies mostly focus on adipocyte derived leptin and

demonstrate that its central actions can control feeding9. Given that leptin must cross the BBB147

and modulate feeding through release of brain neuropeptides and subsequent changes in gene

expression, this satiety effect is considered to be effective over a long-term period. In contrast,

gastric leptin is thought to modulate short-term satiation through activation of gastric and/or

intestine nerve endings. This is based on the findings that Leprb is found to be expressed on cells

Page 27: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

14

of, or vagal afferents innervating the stomach and small intestine144,148–151. It may be that vagal

afferent Leprb play a more important role in food intake regulation as selective deletion of these

receptors in vagal afferents resulted in increased food intake and weight gain152, which is not

seen with their deletion in epithelial cells153,154. Nonetheless, given that leptin makes its way to

the duodenal lumen, it may work in concert with other gut peptides to regulate feeding. In fact,

leptin has been demonstrated to cause secretion of CCK, which results in a positive feedback

loop to increase the amount of leptin released143. In addition, both Leprb149 and CCK1

receptors155 are expressed on vagal afferent neurons, and activate common targets to trigger

vagal firing, whereby leptin enhances CCK induced satiety156,157. Taken together, these findings

suggest a cooperative and synergistic mechanism for CCK and gastric derived leptin to regulate

short-term satiation.

Similar to feeding regulation, the glucoregulatory role of leptin has been mostly studied

in the hypothalamus158. However, given its role in CCK secretion, it also plays a role in

regulating gastric emptying. Thus, leptin indirectly (through CCK action) regulates glucose

homeostasis by inhibiting the amount of nutrients entering into the intestinal lumen to be

absorbed into the circulation159. Given that CCK has been shown to acutely regulate glucose

homeostasis through a gut-brain-liver axis (described in detail below), it remains to be

elucidated whether intestinal leptin action has a similar glucoregulatory role. Leptin also

regulates GLP-1 secretion160, which affects glucose homeostasis through a variety of

mechanisms and will be described later. However, it should be noted that these studies do not

directly implicate gastric derived leptin in these effects, as leptin was administered i.p..

Conversely, a direct role for gastric derived leptin on intestinal regulation of glucose

homeostasis comes from the finding that leptin regulates glucose absorption as luminal leptin

administration reduced the recruitment of the sodium glucose transporter-1 (SGLT-1) to the

apical membrane161, thus impeding glucose absorption within the intestine.

Page 28: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

15

In addition to the stomach derived peptides ghrelin and leptin, the small intestine

secretes a variety of hormones to trigger both local and peripheral signaling to regulate both

feeding and glucose homeostasis, which will be described in detail below.

1.3.2 Proximal Intestinal Peptides

1.3.2.1 Cholecystokinin

In 1928, Ivey and Oldberg discovered the gut peptide CCK and implicated its role in

stimulating gall bladder contractions162. CCK is predominantly found within intestinal I cells in

the proximal intestine (duodenum and jejunum), but also within the enteric and central nervous

system and pancreas163. The 115 amino acid prepro-CCK polypeptide must undergo multiple

posttranslational modifications including sulfation of its C terminus via protein tyrosine

sulfotransferase, multiple cleavage steps via endoprotease and carboxypeptidase E and

amidation via amidating enzyme to generate CCK-8, the shortest and biologically active form of

CCK164. All forms of nutrients (glucose, lipids, and proteins) have all been shown to stimulate

CCK secretion165–168. More specifically, the breakdown of triglycerides into long chain fatty

acids (LCFA) is required for lipids to stimulate CCK secretion169–171, and individual amino acids

such as phenylalanine172 and tryptophan173 can stimulate CCK release. The mechanisms

initiating CCK release remain largely unknown, but have begun to be uncovered. For instance,

the involvement of the lipid transporter, cluster determinant 36 (CD36) and G-protein coupled

receptors (GPCRs), such as GPR40 are required174,175 and protein kinase C (PKC), a

serine/threonine kinase may be involved for lipid induced secretion of CCK as the LCFA oleic

acid has been shown to release CCK in vitro through activation of PKC176. PKC may then

activate Soluble NSF Attachment Protein REceptor (SNARE) proteins or accessory proteins, as

PKC induces insulin secretion in pancreatic β cells through mammalian uncoordinated-18-1

(MUNC18-1) and vesicle-associated membrane protein-2 (VAMP2)177, and stimulates CCK

Page 29: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

16

secretion in vitro through VAMP2178. More recently, the involvement of immunoglobulin-like

domain containing receptor-1 on the basolateral surface of I cells has been shown to mediate fat

stimulated CCK secretion179, which may elevate calcium levels and cause docking of SNARE

proteins for CCK release. After secretion from intestinal I cells, CCK plays an important role

pertaining to the digestion and absorption of nutrients through stimulation of pancreatic

secretion, excretion of bile from the gall bladder, and delaying gastric emptying, which enables

the intestine to effectively digest nutrients180–184.

Studies have demonstrated that CCK also plays an important role in mediating hunger

suppression in rodents, primates, and humans185–193 through a neuronal network194–196. This

appetite suppressive effect of CCK is mediated through the CCK receptor, of which there are

two known isoforms, namely the CCK-1 receptor (predominantly expressed in the

gastrointestinal tract) and the CCK-2 receptor (predominantly expressed in the brain).

Activation of the CCK-1 receptor is necessary for lipids to lower food intake167,197–199 through

vagal afferent firing200,201 and subsequent hindbrain N-methyl-D-aspartate (NMDA) receptor

activation202 involving mitogen activated protein kinase (MAPK) signaling203. Thus, like the

previously described peptides, CCK activates a local paracrine gut-brain axis to control feeding.

In fact, as mentioned, CCK may not work alone but through its interaction with leptin by

affecting the capacity of vagal afferent neurons to regulate expression of transcription factors156.

As to which isoform mediates these effects remains debated as while CCK-8 is the biologically

active form, it has also been suggested that CCK-33 and CCK-58 may play a role in food intake

through reducing meal size and prolonging the intermeal interval, possibly due to their long half

life204,205.

Although these findings suggest a local paracrine vagal mediated pathway for CCK-

induced satiation, there are studies that suggest that CCK may induce satiety through an

endocrine action at the level of the brain. This is based on the following findings: 1) the CCK-2

Page 30: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

17

receptor is also expressed within the hindbrain and hypothalamus, 2) microinjections of CCK

into various hypothalamic nuclei decreases food intake206,207, and 3) lesions of the hindbrain

attenuate CCK-induced satiation208. However, the findings that infusion of CCK into the celiac

artery (which directly supplies the GI tract) significantly reduced food intake in comparison to

infusion the jugular vein209 suggests that the energy intake suppressive effect of CCK is likely

via activation of a local paracrine gut-brain axis, rather than an endocrine effect in the brain.

In addition to its food intake suppressive effects, it is generally believed that CCK

regulates GSIS to regulate glucose homeostasis. However reports on CCK-induced GSIS have

mixed findings, some demonstrating an effect210–219, where in others, this effect is absent220–224.

This may be due to the differences in the dose of CCK administered in these studies suggesting

that CCK’s effects may be pharmacological rather than physiological. Nonetheless, an

intravenous CCK infusion results in a drop in plasma glucose levels in conjunction with

biphasic insulin secretion225 and CCK deficient mice have impaired insulin secretion226. This

effect is likely due to CCK interaction with its receptor, as an i.p. injection of a CCK1 receptor

antagonist negated CCK-induced insulin release227 and may be mediated via its endocrine action

on pancreatic β cells, where its receptor is expressed 228. It is suggested that CCK may potentiate

GSIS through activation of G proteins and the classical phospholipase C (PLC) system229. This

involves an increase in the production of inositol triphosphate (IP3) and diacylglycerol (DAG)

which activates PKC230 and increases intracellular calcium levels227 to induce insulin secretion.

Alternatively, after PKC activation, CCK may activate phospholipase A2 that forms arachidonic

acid (AA)231 to cause insulin release, which has been demonstrated to be independent of

changes in calcium levels232. A recent study also suggests that CCK may regulate insulin

sensitivity226 although the mechanism(s) involved are unknown and require further

investigation.

Page 31: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

18

CCK also regulates glucose homeostasis through modulating gastric emptying. This has

been demonstrated in humans, where CCK modulates postprandial glycemia by delaying gastric

emptying of nutrients from the stomach into the proximal gut180. As with many other functions

of CCK, its effect on gastric emptying is mediated through the CCK1 receptor233–237. Further

evidence suggests that CCK may not need to bind to its CCK1 receptor located on the pyloric

sphincter, but rather may act in a paracrine fashion and directly activate CCK1 receptors present

on gastric vagal afferents to delay gastric emptying238. Intestinal motility and pyloric pressure239

are also affected by CCK, further demonstrating the ability of CCK to regulate nutrient transit in

the intestine and subsequent absorption into the circulation. Thus, through regulating GSIS and

gastric emptying, CCK can regulate postprandial glucose homeostasis.

More recently the ability of CCK to activate a gut-brain-liver axis to acutely regulate

glucose production has been demonstrated (described in detail below). However, the signaling

cascade required for such regulation remains to be determined. Given that pancreatic CCK1

receptor signaling is well known, perhaps a similar signaling cascade exists at the level of the

gut to regulate glucose homeostasis, which is a focus of the current thesis. Another duodenal

hormone, GIP, also shares similar pancreatic effects as CCK and will be described in detail

below.

1.3.2.2 Glucose-dependent Insulinotropic Polypeptide

GIP is a single 42-amino acid peptide that is derived from a 153-amino acid precursor

through post-translational processing of proGIP240. This peptide was originally observed to

inhibit gastric acid secretion and motility in dogs241 and is highly expressed in K cells of the

duodenum and jejunum242. In the fasting condition, GIP circulates at low levels but becomes

elevated upon feeding by glucose243–247, proteins248 and fats, where in regards to fats, long chain

triglycerides are the most potent stimulator of GIP release249. The ability of various nutrients to

Page 32: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

19

stimulate GIP release appears to be species dependent, where fat is a more potent stimulator

than carbohydrates in humans, and in contrast, carbohydrates are more potent than fat in rodents

and pigs250. The release of GIP by nutrients involves nutrient associated receptors or transporters

such as GPR40, GPR120, SGLT-1251,252.

Interestingly, GIP does not regulate food intake, which has been confirmed in

humans253,254. GIP is most commonly known and demonstrated as an incretin hormone. An

incretin hormone (INtestine seCRETion INsulin) is defined as a gut-derived hormone that

induces a greater insulin secretory response upon an oral glucose load in comparison to an

intravenous glucose infusion of the same amount. A direct infusion of GIP enhances GSIS in

healthy humans and rats by affecting the early-phase of insulin release255–259. The insulinotropic

affect of GIP is mediated via its endocrine action on pancreatic islets as GIP receptors (GIPR)

are expressed on pancreatic β cells260. Upon activation of the GIPR, there is an increase in cyclic

adenosine monophosphate (cAMP) levels261 which activate downstream mediators, protein

kinase A (PKA) and cAMP guanine nucleotide exchange factor II (cAMP-GEFII; Epac2)262.

Both of these downstream molecules are involved in a variety of intracellular functions such as

altered ion channel activity that leads to an increase in calcium levels and enhanced insulin

exocytosis. More specifically, activation of PKA leads to adenosine triphosphate (ATP)-

sensitive potassium (KATP) channels closure and subsequent depolarization of the plasma

membrane263. This depolarization event leads to the opening of voltage gated calcium channels,

which allows the entry of calcium to further increase intracellular calcium levels through

mobilization from intracellular stores264 via PKA and Epac2. This increase in calcium results in

insulin secretion through calcium dependent exocytosis. Given that CCK and GIP both regulate

GSIS through activation of GPCR pathway(s), and that duodenal CCK regulates glucose

production through its receptor (discussed below), it remains to be investigated whether GIP

Page 33: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

20

may also regulate glucose homeostasis via local paracrine activation of a gut-brain axis

through activation of common downstream signaling.

In addition to insulin secretion, GIP also stimulates the secretion of glucagon255,265 likely

through a direct action on pancreatic islets as the GIPR is found to be expressed on pancreatic α

cells260. In humans, it has been demonstrated that GIP up regulates glucagon levels during

fasting and hypoglycemic conditions266. This suggests that this hormone has diverging glucose

dependent effects with opposite actions on the two main pancreatic hormones, therefore acting

as a bi-functional blood glucose level stabilizer.

In contrast to the other gastrointestinal peptides discussed, GIP does not regulate the rate

of gastric emptying from the stomach254,267. However, a recent study suggest that GIP inhibits

intestinal glucose absorption and intestinal motility through a somatostatin mediated pathway268.

This is contrast to previous findings that GIP increases SGLT-1 expression and thus increases

glucose absorption from the small intestine269. In addition, another previous study also reports

that similarly to GLP-1, GIP also regulates intestinal motility263 which may affect glucose

absorption. Thus, while GIP may regulate glycemia via its effect on intestinal glucose

absorption, it is likely that GIP does not activate a gut-brain axis directly to regulate glycemia

given that its receptor is not expressed in vagal afferents270.

More commonly known for its incretin action is the hormone GLP-1 that is found in the

more distal intestine, and will be described in detail below.

1.3.3 Distal Intestinal Peptides

1.3.3.1 Glucagon-like peptide-1

GLP-1 is a posttranslational product of proglucagon which is mainly found to be

expressed in L cells271 in the distal intestine (the ileum and colon) but is also found in the

CNS272 and pancreas273. There are two forms of GLP-1, GLP-1(7-36)NH2 and GLP-1(7-

Page 34: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

21

37)274,275 that arise from enzymatic cleavage of the proglucagon by prohormone convertase 1/3

to become active and then enter the lymphatic system or bloodstream to exert their actions276.

GLP-1 has a very short half-life (1-2 minutes)277 due to its rapid degradation by the enzyme

dipeptidyl peptidase IV (DPP-IV)278, and as such it is debated whether a rapid neuronal

mechanism may be needed for GLP-1 action. Moreover, the secretion of GLP-1 is biphasic,

with an early phase of secretion, followed by prolonged second phase279. GLP-1 binds to its

receptor (GLP-1R), a GPCR280 that is found to be expressed in many tissues281, to exert its wide

variety of physiological effects.

Given that nutrients do not likely reach the distal intestine within the time frame of GLP-

1 secretion, it is suggested that intestinal hormones may signal via vagal nerves to cause its

secretion282. However, L cells are also found in the proximal small intestine, and may contribute

to the secretion of GLP-1 from the intestine into the circulation271. Given that CCK activates a

neuronal network to regulate glucose homeostasis at the level of the duodenum (described in

detail below), and that GLP-1 may be released in the duodenum and activate GLP-1Rs on vagal

afferents283 to increase vagal firing284,285, it remains to be investigated whether GLP-1 and CCK

work together to regulate this axis. Nonetheless, all forms of nutrients cause GLP-1 secretion

and their mechanism of secretion have been studied. Glucose-mediated secretion may be

mediated by sweet taste receptors or SGLT-1286,287, although glucose sensing and signaling

mechanisms in the intestine are still under debate288. For lipids, similar to what is seen for the

release of other peptide hormones, the hydrolysis of triglycerides to release LCFAs is required

for GLP-1 release289, and may be mediated by fatty acid transport protein 4290, GPR40291, and

GPR120292. As mentioned earlier, LCFA entry into the duodenum also stimulates CCK

secretion, which has also been implicated in GLP-1 release as blockade of CCK1 receptor

signaling during fat intake abolished the rise of GLP-1 in humans289. The mechanism of amino

acid induced secretion of GLP-1 remains largely unknown, but a recent studies suggests the

Page 35: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

22

involvement of G-protein coupled receptor (GPCR) 6A293. Beyond nutrient stimulation, GLP-1

may mediate its own secretion through an autoregulatory loop294 or be stimulated by bile acids

via a TGR5-dependent pathway295,296.

After entering the circulation, GLP-1 is widely known for its food intake suppressive

effects in both rodents and humans, which are mediated both through peripheral and central

mechanisms. Its peripheral effects are likely mediated via activation of vagal afferents297 where

its receptor is expressed283. This is demonstrated by the fact that peripheral exendin-9 (Ex-9)

administration abolishes the food intake suppressive effects of peripheral GLP-1298, and surgical

or chemical inactivation of the vagus attenuates GLP-1R activation and satiety299. However, the

involvement of the vagus nerve in mediating GLP-1 anorectic effects has been recently

challenged and may be due to CNS GLP-1R signaling300. Indeed, GLP-1 may act in various

regions of the brain including the hindbrain and hypothalamus and studies have demonstrated

that an injection of Ex-9 into the third ventricle abolishes the ability of GLP-1 to decrease food

intake301–303. Further, direct hindbrain administration of Exendin-4 (Ex-4; GLP-1 analog)

reduces food intake304 which is mediated by PKA/MAPK signaling305. However, a connection

between the suppressive effects on food intake of both the peripheral and central GLP-1R may

exist as peripheral GLP-1 administration fails to affect food intake after abolishment of a vagal-

brainstem-hypothalamic pathway299.

In addition to the food intake suppressive effect of GLP-1, GLP-1 also belongs to the

family of incretin hormones and potentiates GSIS through either a paracrine or endocrine

fashion, similar to GIP. Indeed, intestinal GLP-1 reaches the pancreas via the portal vein, but

blockade of vagal activation attenuates GSIS, indicating that GLP-1 may act within a paracrine

fashion to stimulate insulin release306. Moreover, GLP-1 also stimulates GSIS through binding

to its GLP-1R expressed on the β-cell307. Similar to its effects in the brain, increased AC activity

occurs after binding of GLP-1 to its receptor, resulting in formation of cAMP which

Page 36: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

23

subsequently increases PKA activity308. Additionally, increased cAMP concentrations results in

the activation of cAMP-GEFII or Epac2262. Both proteins alter ion channel activity, which leads

to the closure of KATP-channels through phosphorylation of the SUR1 subunit309. Interestingly,

GLP-1 has been shown to sensitize the KATP channels to ATP, as less ATP is required for

closure of the channels310. The closure of these channels shifts the membrane potential and

opens internal voltage-gated calcium channels, increasing intracellular calcium levels through

release from intracellular calcium stores311 and the number of readily releasable insulin

secretory vesicles. In addition to PKA activation, other pathways have been implicated such as a

calmodulin mediated pathway. Briefly, a calmodulin inhibitor reversed the actions of GLP-1 on

KATP channels and subsequent depolarization of the membrane312.

In contrast to the similarity of GLP-1 and GIP to increase insulin secretion, GLP-1

inhibits glucagon secretion313 to regulate circulating glucose levels, which is glucose

dependent314–316. This has been demonstrated in isolated rat islets and is proposed to be

mediated by somatostatin secretion from pancreatic δ cells by GLP-1 and subsequent binding to

the somatostatin receptor-2317. This effect is likely paracrine in nature as treatment with

somatostatin antibodies abolished the inhibitory effect of GLP-1 on glucagon secretion317. In

addition, GLP-1Rs are not found to be expressed on α cells307 which strengthens the hypothesis

that this inhibition is likely through an indirect mechanism. This may be through its action on δ

cells, although the findings of GLP-1R expression on δ cells is inconsistent318. Moreover, this

effect of GLP-1 is likely in hypoglycemic conditions, when glucagon levels are high in order to

increase circulating glucose levels. This is based on findings that during a hypoglycemic clamp

in humans, the inhibitory effect of GLP-1 on glucagon secretion was lost when circulating

glucose levels were near or just below normal fasting levels319.

In addition to its direct effect on the pancreas, GLP-1 also regulates glucose

homeostasis via extrapancreatic mechanisms. Given that the portal vein is exposed to higher

Page 37: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

24

levels of active GLP-1, it is not surprising that GLP-1 acts within this region to regulate

glucose disposal, given the existence of a hepatoportal glucose sensor, and the finding that

portal infusion of glucose with exendin-9 attenuated the ability of portal glucose to increase

glucose clearance320. In line with these findings, an increase in intestinal incretin action via

selective inhibition of intestinal and not systemic DPP-IV is also sufficient to enhance glucose

tolerance in association with increased vagal firing285. Thus, GLP-1R activation within the

portal vein triggers a portal-brain-muscle neuronal axis to control glucose disposal. Given these

findings, it remains to be addressed whether intestinal GLP-1 locally activates its receptors on

vagal afferents innervating the small intestine to trigger a gut-brain-liver axis, like CCK, to

regulate glucose production.

Although unknown for intestinal GLP-1 action, within the arcuate nucleus of the

hypothalamus, GLP-1 has been demonstrated to regulate glucose production321. In addition,

central GLP-1 signaling has also been shown to reduce insulin stimulated muscle glucose

utilization under hyperglycemic conditions to favor hepatic glycogen storage322. However, the

role of CNS GLP-1 signaling (as well as vagal signaling) in the regulation of glucose

homeostasis has recently been challenged, similar to feeding regulation. This is due to the

recent finding that liraglutide, a long acting GLP-1 agonist, still has glucose lowering effects in

mice lacking the GLP-1R in either the vagus nerve or brain300. Given that this study utilized

mice lacking the GLP-1R from birth, additional studies are warranted to dissect the role of both

vagal and brain GLP-1R signaling in the regulation of glycemia.

GLP-1 also regulates glucose homeostasis through its ability to control GI motor

functions through the “ileal brake” where the rate of specific unabsorbed nutrients reach the

distal intestine is controlled. In humans, intravenous GLP-1 administration slows gastric

emptying in a dose-dependent manner which is likely mediated via vagal afferents323. In

addition, GLP-1 also controls pressure waves in the duodenum and can alter pyloric pressure,

Page 38: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

25

effects that are abolished upon Ex-9 administration324–326. Thus, GLP-1 alters the amount of

nutrients entering the small intestine, another way in which it regulates glucose homeostasis.

A close relative to GLP-1, GLP-2, is also secreted from L cells, and its involvement in

glucose and food intake regulation is described below.

1.3.3.2 Glucagon-like peptide-2

GLP-2 is a 33 amino acid peptide that is created by posttranslational cleavage of

proglucagon at the same time as GLP-1, and is also found to be located in intestinal L cells. It is

believed that GLP-2 is secreted upon nutrient ingestion327, along with GLP-1. Therefore, GLP-

2’s secretion is likely predominantly mediated by long chain fatty acids282,290,291,328. However, as

discussed for GLP-1, other L cell secretagogues include glucose and bile acids329,330 as well as

peptide hormones such as GIP and leptin may also cause GLP-2 secretion. In regards to its

secretion profile, GLP-2 also exhibits a biphasic pattern of secretion with an acute increase that

occurs rapidly followed by a more delayed and prolonged response327,328,331. After secretion,

GLP-2 mediates its effects through binding to its receptor (GLP-2R), a GPCR, and activates a

diverse set of downstream signaling molecules such as PKA332.

GLP-2 is widely known for its intestinal growth actions such as affecting barrier

function and intestinal protection333. However, its effect on suppressing food intake have been

debated. For instance, its peripheral effects have been demonstrated in mice where an i.p

injection of GLP-2 reduced short term feeding, which was abolished when a GLP-2R antagonist

was co-administered334. However, this same effect was not demonstrated in humans335,336. Thus

the peripheral effects of GLP-2 on food intake remain to be resolved. Rather, it is more widely

accepted that the food intake suppressive effects of GLP-2 are mediated centrally. Indeed,

multiple regions of the brain express the GLP-2R including hypothalamic and

extrahypothalamic regions337 and it has been demonstrated that an intracerebroventircular (i.c.v)

Page 39: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

26

GLP-2 infusion inhibits food intake337,338 through hypothalamic neuron signaling339. This food

intake suppression is likely through a relay between the hypothalamus and hindbrain338.

As mentioned previously, the main site of action for GLP-2 is within the GI tract where

GLP-2 affects intestinal nutrient transit. It has been demonstrated to inhibit gastric

emptying,340,341 enhance gastric capacity by decreasing gastric fundic tone342 and reduce

intestinal transit in vivo343. This inhibitory effect may be mediated by central signaling by GLP-

2, as neuronal specific deletion of the GLP-2R in hypothalamic nuclei resulted in accelerated

gastric emptying339. However, GLP-2’s effects on gastric emptying are not as potent as seen for

GLP-1341, but nonetheless, its affect on intestinal transit of nutrients ultimately affects glucose

homeostasis.

In contrast to GLP-1, GLP-2 is not an incretin, and as such it does not affect GSIS by

pancreatic β cells. However, it has been suggested that GLP-2 does affect glucagon secretion

from α cells through activation of its receptor in rats344 suggesting that it may play a role in the

regulation of glucose homeostasis. In line with these findings in rats, the GLP-2R has been

detected in α cells in humans344 and an exogenous GLP-2 administration rapidly increased

plasma glucagon levels345. In contrast, other studies have not been able to detect the GLP-2R in

murine islets and did not see an increase in glucagon levels after GLP-2 administration346. Thus,

the importance of GLP-2R signaling in the control of glucagon secretion requires further

clarification as differences are detected among different species.

In addition to its effects described above, it is recently suggested that GLP-2 may play a

role in regulating insulin sensitivity through central GLP-2R. This was demonstrated by the

findings that GLP-2R deletion in the hypothalamic pro-opimelanocortin (POMC) expressing

neurons impaired glucose tolerance and hepatic insulin sensitivity and GLP-2R activation in

these neurons activated PI3K/Akt signaling which was required for GLP-2 to regulate hepatic

glucose production347. These findings uncover a new role for GLP-2 in the regulation of glucose

Page 40: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

27

homeostasis in addition to its known affects on intestinal growth. Given these findings, future

investigation into whether GLP-2 regulates glucose production at the level of the small intestine

is warranted, which has been demonstrated for the gut-derived hormone CCK (please see

below).

Another L-cell derived hormone is OXN, which has been suggested to regulate

metabolic homeostasis as described below.

1.3.3.3 Oxyntomodulin

OXN is a 37 amino acid peptide derived from glucagon whose structure was elucidated

in 1981348. Similar to GLP-1 and GLP-2, OXN is also found in L-cells of the small intestine349

and its secretion is stimulated upon nutrient ingestion, with GLP-1350. There is no clear

demonstration of the existence of an OXN receptor, and it is more widely accepted that OXN

binds to the GLP-1R or glucagon receptor (GCGR) to exert its effects351. Thus, similar to that

postulated for GLP-1, local OXN activation of the GLP-1R may regulate glucose homeostasis

similar to that seen for CCK within the intestine. Interestingly, in regards to the GLP-1R, OXN

activates downstream molecules β-arrestin 2 and results in an increase in cAMP levels but has

less preference to MAPK signaling352. This suggests that OXN and GLP-1 may differ in their

downstream signaling and in vivo effects.

Similar to many of the peptide hormones already discussed, OXN has food intake

suppressive effects both peripherally and centrally depending on the species studied. Indeed,

OXN administration dose-dependently inhibited food intake in rats353 and humans354. In

contrast, peripheral OXN administration in mice did not affect feeding. However, i.c.v

administration of OXN transiently inhibited food intake likely through its activation of the GLP-

1R as the food intake suppressive effects of OXN are abolished in GLP-1R knock out mice355.

Thus, the food intake suppressive effects of OXN remain to be clarified.

Page 41: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

28

Moreover, the ability of OXN to regulate glucose homeostasis remains largely unknown,

but some studies do suggest such a role. For instance, OXN has been demonstrated to regulate

gastric emptying in humans356, but these findings were not seen in mice357. In regards to GSIS,

OXN has been shown to increase cAMP levels in association with insulin secretion in mice357,

and similar results have been demonstrated in rats358. Additional studies are warranted to

address the glucoregulatory role of OXN.

The last L-cell derived metabolic regulatory hormone is PYY, which is described in

detail below.

1.3.3.4 Peptide YY

PYY is a 36 amino acid peptide that is secreted from L cells together with the previously

described peptides GLP-1/2 and OXM359. In the circulation, there are two forms of PYY arising

from its cleavage leading to PYY(1-36)NH2 and PYY(3-36)NH2360,361 by DPP-IV278, with

PYY(3-36)NH2 being the major circulating from of PYY. Both carbohydrates and lipids

stimulate its release. In regards to lipids, the conversion to LCFA170,362 is required to stimulate

PYY release and may involve GPCRs as demonstrated for GLP-1 release. In regards to

carbohydrates, the mechanisms described for GLP-1 may also be involved as described

previously. CCK can also stimulate PYY release in humans suggesting that a neuronal axis

exists between the duodenum and ileum363. PYY is also similar to GLP-1 in terms of its

secretion profile which is biphasic, resulting in an increase in PYY levels in as short as 15

minutes which peak after 1-2 hours after a meal364. After secretion, PYY(1-36)NH2 binds to its

Y receptor subtypes Y1, Y2, Y4 and Y5 receptors, where PYY(3-36)NH2 binds only to Y2 and

Y5359,365.

Similar to other peptide hormones discussed, peripheral and central PYY(3-36)NH2

administration has been demonstrated to regulate feeding. PYY(3-36)NH2’s peripheral affects

Page 42: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

29

are likely mediated via vagal afferents where the Y2 receptor is expressed366, as vagotomy

abolished exogenous PYY(3-36)NH2 induced c-fos activation in the ARC and did not affect

feeding299. Furthermore, direct PYY(3-36)NH2 administration is hypothesized to inhibit ARC

neuropeptide Y (NPY) neurons to suppress feeding, as PYY(3-36)NH2 binding to Y2 receptors

resulted in reduced NPY release through a reduction in cAMP production and neurotransmitter

exocytosis367. Thus, PYY(3-36)NH2 may inhibit food intake through a neuronal gut-brain axis.

Given that CCK regulates feeding and glucose homeostasis through a gut-brain and gut-brain-

liver axis, respectively, it remains to be addressed whether PYY may also regulate glucose

homeostasis through activation of its receptor expressed on vagal afferents, in addition to

feeding.

In contrast to many of the other peptides discussed, PYY(1-36)NH2 inhibits GSIS. This

is likely by direct action on the pancreas as suggested by several studies: 1) Pyy knockout mice

exhibit hyperinsulinemia in both fasted and fed states368, 2) studies have demonstrated in

isolated islets that direct PYY(1-36)NH2 administration reduced GSIS in a dose dependent

manner369,370 and 3) mice lacking the Y1 receptor hypersecrete insulin371. Moreover, it is

suggested that PYY(3-36)NH2 does not directly affect GSIS372 as neither Y2R and Y5R are

detected in murine islets371,372. Interestingly, PYY(3-36)NH2 instead may stimulate insulin

secretion through increasing GLP-1 levels372. Thus, the ability of PYY to regulate insulin

secretion remains controversial but if in fact PYY inhibits GSIS, it may be that it works in an

opposite fashion to other gut peptides to prevent hypoglycemia.

In contrast to the findings that PYY may inhibit GSIS and thus elevate glucose levels, it

may inhibit gastric emptying, suggesting that it could play a role in ensuring glucose levels do

not get too high after a meal. This has been demonstrated in various species, including

monkeys373 and humans, where PYY(3-36)NH2 is more effective in comparison to PYY(1-36)

NH2374,375. In addition to gastric emptying inhibition, PYY also regulate intestinal motility,

Page 43: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

30

suggesting that PYY regulates entry and subsequent movement of nutrients in the intestinal

tract.

Taken together, the introduction to the above gastrointestinal peptide hormones

demonstrates that local hormonal signals induce neuronal activation to regulate metabolic

homeostasis. The mechanism of local hormonal signaling to trigger a gut-brain-liver axis,

specifically, to regulate glucose production will be described below.

1.4 Small intestine control of glucose production through a gut-brain-liver neuronal

axis

Modified from: Rasmussen, BA*, Breen, DM* and Lam, TK. Lipid sensing in the gut, brain and liver. Trends Endocrinol Metab 23, 49-55, 2011 *Equal contribution (Review)

As mentioned above, upon nutrient entry, the small intestine secretes a variety of

peptides that regulate glucose homeostasis and food intake through a variety of mechanisms.

Throughout this introduction, it has become apparent that, in addition to their indirect effects,

many studies already point to direct hormonal signaling in the gut to regulate feeding and

glucose regulation. In line with this hypothesis, the gut-derived hormone CCK can locally

activate a gut-brain-liver neuronal network to lower glucose production. In the following

section, the neuronal axis triggered by nutrient sensing and subsequent peptide hormone

secretion will be discussed in detail as applicable.

1.4.1 Duodenal lipid sensing and CCK secretion triggers a gut-brain-liver axis to lower

glucose production

1.4.1.1 LCFA!LCFA-CoA

After ingestion of a meal, dietary lipids accumulate in the small intestine. The most

prominent forms of lipid in a typical western diet include triglycerides, cholesterol,

phospholipids and LCFA, where triglycerides are the primary form of lipid. Triglycerides are

Page 44: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

31

emulsified through secretion of bile salts from the gallbladder. Pancreatic lipase secretion from

the exocrine pancreas breaks down triglycerides to fatty acids and monoglycerides to be

absorbed by enterocytes. Lipid sensing is then said to begin with the absorption of triglycerides

in the intestinal lumen. Fatty acids are transported into enterocytes via CD36, which functions as

a fatty acid transporter in the proximal sections (duodenum and jejunum) of the small

intestine174. Sensors of FFA in the small intestine include GPCRs such as GPR40 and GPR120,

whose ligands both include medium- and long-chain saturated and unsaturated FFA. Both

GPR40 and GPR120 have been shown to play a role in FFA-stimulated secretion of gut derived

hormones291,292, as discussed previously.

Once inside the cell, LCFAs are metabolized by acyl-CoA synthetase (ACS) to form

LCFA-CoA, which is mediated via the specific ACS isoform, ACS3376. After conversion,

LCFA-CoA have two fates: 1) LCFA-CoA and monoglycerides are recombined into

triglycerides and packaged into chylomicrons for exocytosis, or alternatively, 2) LCFA-CoA are

then transported into the mitochondria via carnitine almitoyltransferase-1 (CPT-1) to undergo β-

oxidation. The process through which β-oxidation occurs at the level of the intestine is similar to

what occurs in the brain377 and liver378.

Regardless of the fate of LCFA-CoA, its formation is required for intestinal lipids to

lower glucose production379. This has been demonstrated by an intraduodenal infusion of

Intralipid (a soybean emulsion of mono- and polyunsaturated fatty acids), which was able to

lower hepatic glucose production during the pancreatic basal insulin clamp technique. Of note,

Intralipid was infused at a rate that ensured the effects observed were in the pre-absorptive

state380. Co-infusion of Intralipid with triacsin C (an inhibitor of ACS3381) prevented the

suppression on glucose production induced by upper intestinal lipids, suggesting that conversion

of LCFA to LCFA-CoA is essential for upper intestinal lipids to lower hepatic glucose

production. The suppression of glucose production was mediated by a gut-brain-liver axis as 1)

Page 45: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

32

interruption of the neuronal connection between the gut and brain by co-infusion of tetracaine,

or performing subdiaphragmatic vagotomy or gut vagal deafferentation surgical procedures, also

blocked the ability of LCFA to suppress glucose production 2) administration of MK-801

(NMDA ion channel blocker) into the nucleus of the solitary tract (NTS) abolished the LCFA

induced inhibition of glucose production and 3) infusion of Intralipid into rats that had received

hepatic vagotomy (a surgical technique that interrupts the connection between the brain and

liver) also abolished the ability of fats to lower glucose production379. All of the above effects

were independent of weight loss, consistent with all the studies discussed below. In line with

these findings, a recent study showed that activation of NMDA receptors within the dorsal vagal

complex (DVC) by the agonist glycine decreases glucose production382 strengthening the

existence of a gut-brain (at the level of the DVC and activation of NMDA receptors)-liver axis

to regulate glucose production (Figure 1.2).

The ability of upper intestinal lipids to regulate glucose homeostasis has recently been

shown to be mediated by PKC-δ activation383 and CCK-1 receptor activation384. The importance

of both of these molecules in mediating upper intestinal lipid regulation of glucose production is

shown by the inability to regulate plasma glucose levels in fasting/refeeding experiments upon

inhibition of either PKC-δ383 or CCK-1 receptor activation384. These experiments highlight the

possible downstream mediators of intestinal lipid sensing and will be discussed in the following

sections.

1.4.1.2 PKC-δ! CCK

It is evident from the previous section that changes in the availability of LCFA-CoA

regulate glucose homeostasis. Although this is well established, the associated signaling

mechanisms downstream need to be elucidated and studies suggest that PKC, a serine/threonine

kinase, is a potential molecule to mediate the effect of lipid. The PKC family consists of at least

Page 46: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

33

10 isoforms, which are divided into three subfamilies based upon their second messenger

requirements385. Conventional PKCs (α, βI, βII, and γ) require both calcium and the lipid DAG

for activation whereas novel PKCs (δ, ε, η, and θ) require DAG but not calcium. Unlike both

conventional and novel PKCs, the atypical PKCs (ζ, ι, and λ) require only phospholipids/lipids

and not calcium or DAG for activation. As there are several different PKC isoforms and PKC

protein expression varies between different tissues, it is not surprising that numerous biological

functions have been ascribed to PKC activity.

As mentioned previously, short-term accumulation of LCFA-CoA in the duodenum

lowers glucose production through a gut-brain-liver neuronal axis379. However the necessary

step(s) that mediate this effect on glucose production were not elucidated in that study. Given

that most PKC isoforms are present in the small intestine and the pattern of expression between

rodents and humans is similar, with a few exceptions386,387, a potential role for PKC in

regulating glucose production is warranted. In fact, activation of duodenal mucosal PKC-δ was

found to be sufficient and necessary for lipid sensing to regulate glucose production383. In brief,

an intraduodenal infusion of 1-oleoyl-2-acetyl-sn-glycerol (OAG, PKC activator), during the

pancreatic basal insulin clamp, lowered glucose production, which was blocked by co-

administration of a PKC-δ inhibitor or an adenovirus expressing the dominant negative form of

PKC-δ. Furthermore, this effect was shown to be mediated by a gut-brain-liver neuronal axis as

administration of tetracaine or NTS MK-801, and hepatic vagotomy all prevented PKC-δ

activation from decreasing glucose production (Figure 1.2).

The above results discussed suggest that duodenal lipid sensing and subsequent

activation of PKC-δ occur in the fasting state as the pancreatic clamp technique is conducted in

animals that have undergone 5 hours of fasting. What remains in question is whether these

mechanisms are activated during refeeding. Thus, a more physiological assessment of the role of

Page 47: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

34

intestinal lipid sensing mechanisms in the regulation of glucose homeostasis involves the use of

fasting/refeeding experiments. During fasting/refeeding, circulating glucoregulatory hormones

are changed at will while plasma glucose levels rise, and this elevation of plasma glucose is

counteracted by an inhibition of hepatic gluconeogenesis388. Direct inhibition of PKC-δ382 in the

duodenum during a fasting/refeeding experiment disrupts glucose homeostasis causing plasma

glucose levels to rise. This observation strengthens the role of PKC-δ in the regulation of

glucose homeostasis. This is also true for CCK-1 receptor inhibition384 which will be discussed

in more detail below.

What is the signaling pathway(s) downstream of PKC-δ that is required for lipids to

lower glucose production? As discussed briefly in section 1.3.2.1, in vitro studies report a

mechanistic link between PKC and CCK as the LCFA oleic acid activates PKC to stimulate the

release of CCK in the secretin tumor cell (STC)-1 cell line176,390. Furthermore, in addition to

PKC-δ being the downstream effector of lipid sensing, activation of duodenal CCK was also

found to be sufficient and necessary for lipid sensing to regulate glucose production384, which

will be discussed in detail in the following section. Based on all of these findings, this

relationship was addressed using both pharmacological and molecular approaches to inhibit

PKC-δ and illustrated that duodenal PKC-δ stimulation requires CCK-1 receptor activation to

lower hepatic glucose production during a pancreatic basal insulin clamp. However, duodenal

PKC-δ is not required for CCK to decrease glucose production391. Whether PKC isoforms other

than PKC-δ play a role either upstream or downstream of CCK in the regulation of glucose

production remains possible and warrants further investigation. Thus, it is proposed that PKC-δ

is upstream of CCK and stimulates CCK release, subsequently leading to the activation of the

CCK-1 receptor to regulate glucose homeostasis. The mechanistic link between PKC-δ

activation and the stimulation of CCK release remains unknown. However, also discussed in

Page 48: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

35

section 1.3.2.1, the potential involvement of SNARE proteins (i.e. Munc18-1 and VAMP-2)

could be the subject of future studies. This is due to the fact that PKC-δ has been shown to

enhance insulin secretion coupled with increased phosphorylation of Munc18-1 in pancreatic β-

cells177 and VAMP-2 mediates CCK secretion in STC-1 cell lines178.

1.4.1.3 CCK!CCK-1 receptor

As discussed in the previous section, CCK lies downstream of PKC-δ and is an

important mediator of duodenal lipid sensing391. A recent study reported that CCK in the

duodenum lowers glucose production through a neuronal network and is downstream of

lipids384. Briefly, a CCK-8 infusion into the duodenum during the pancreatic basal insulin clamp

lowered glucose production. This glucose production suppression effect was abolished upon co-

administration of CCK-8 with the CCK-1 receptor blocker MK-329 as well as infusion of CCK-

8 in CCK-1 receptor knockout rats. In addition, co-infusion of lipids with MK-329 abolished the

ability of the lipid administration within the duodenum to lower glucose production. The gut-

brain axis was also defined, as co-administration of CCK-8 with the anesthetic tetracaine

abolished the glucose production suppression effect of CCK-8. These data suggest that duodenal

CCK-8 stimulates the vagal afferent to lower glucose production and is the downstream

mechanism of lipid sensing (Figure 1.2). Furthermore, inhibition of the NMDA receptor

through administration of MK-801, or hepatic vagotomy surgery blocked the ability of duodenal

CCK-8 administration to lower glucose production384. This finding suggests that activation of

NMDA receptors and subsequent neuronal relay to the liver is required for intestinal lipid

sensing/CCK-1 receptor activation to lower glucose production.

Taken together, the data mentioned provides evidence for the existence of a duodenal

lipid ! LCFA ! LCFA-CoA ! PKC-δ ! CCK ! CCK-1 receptor pathway that triggers a

gut-brain-liver axis to lower hepatic glucose production (Figure 1.2).

Page 49: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

36

1.4.1.4 Effects of High Fat Feeding on the Gut-Brain-Liver Axis

As discussed, several advances have recently been made that have uncovered part of the

downstream signaling mechanisms of intestinal lipids in normal rodents. Interestingly, the

signaling pathway mentioned above fails to lower glucose production in rodents fed a high fat

diet for 3 days, a model of diet-induced hepatic392,393 and hypothalamic394 insulin resistance,

discussed in more detail below. Thus, these studies have allowed us to get closer to identifying

the location of the defect induced by high-fat feeding.

First, as mentioned previously, PKC-δ activation was found to lie downstream of

duodenal lipids to activate the gut-brain-liver neuronal axis to lower glucose production in

normal rodents383. As in the case of duodenal lipid infusion, high-fat feeding also prevented

direct stimulation of PKC-δ, through duodenal OAG infusion, from lowering glucose

production391. These findings suggest that the signaling defect does not lie within the inability of

lipid to trigger signaling events like PKC-δ activation. Similar to PKC-δ, CCK activation was

also demonstrated to be required for duodenal lipids to lower glucose production384. Again, rats

overfed with a HFD completely failed to respond to duodenal CCK-8 to lower glucose

production384. These findings are consistent with the fact that duodenal PKC-δ activation is

upstream of CCK signaling, and that diet-induced CCK resistance is postulated to lie within the

downstream signaling cascade of CCK-1 receptors384, as direct activation of duodenal mucosal

PKC-δ still fails to overcome intestinal CCK resistance to lower glucose production391. Taken

together, these findings strengthen the argument that duodenal lipid resistance lies downstream

of the CCK/CCK-1 receptor signaling cascade (Figure 1.2). However, the exact location of

resistance at the level of the duodenal CCK-1 receptor still remains to be explored. Thus, it is

essential to investigate the downstream signaling mechanisms of the CCK1 receptor to begin to

Page 50: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

37

uncover where the resistance lies to uncover possible ways to restore the functionality of this

axis, a focus of the current thesis.

In addition to the duodenum, gut peptides are also found more distally in the jejunum, as

previously described (i.e. CCK and GIP). This suggests that hormonal signaling in the jejunum

could play a role in regulating glucose homeostasis. First, what is known in regards to the role

of nutrient sensing in the jejunum and the regulation of glucose homeostasis will be discussed in

detail below.

1.4.2 Jejunal nutrient sensing triggers a gut-brain-liver axis to lower glucose production

Modified from: Rasmussen, BA*, Breen, DM*, Côté, CD, Jackson, M, and Lam, TK. Nutrient sensing mechanisms in the gut as therapeutic targets for diabetes. Diabetes 62, 3005-3013, 2013 *Equal contribution (Review)

It is traditionally believed that nutrients reach the distal gut only in malabsorptive

conditions395. However, during the early phases of food ingestion, nutrients have been shown to

reach the distal intestine in both animals396–399 and humans400,401 suggesting that the more distal

intestine may also regulate glucose production through a gut-brain-liver neuronal axis. In fact, a

recent study demonstrates such an axis exists in the jejunum402, and that the jejunum shares

similar nutrient sensing mechanisms as the duodenum, which will be discussed in detail below.

First, the ability of the jejunum to sense lipids was tested. Similar to the findings in the

duodenum, a jejunal Intralipid infusion lowered glucose production during the pancreatic basal

insulin euglycemic clamp technique. This was abolished upon co-infusion of an ACS inhibitor

suggesting that the conversion of LCFA to LCFA-CoA is also required for the jejunum to lower

glucose production402 (Figure 1.2). These effects were independent of weight loss, which is

consistent for all of the findings in the study. What still remains unknown is the downstream

signaling pathway of jejunal lipid sensing. This may require CCK or other gut derived

hormones, which is a focus of the current thesis.

Page 51: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

38

Next the ability of the jejunum to sense glucose to lower glucose production was tested.

Indeed, a direct glucose infusion into the jejunum lowered glucose production during the

pancreatic clamp. This required glucose uptake into intestinal cells as co-infusion of glucose

with phlorizin, a SGLT inhibitor, abolished the ability of glucose to lower glucose production

(Figure 1.2)402. Importantly, a direct glucose infusion into the portal vein at the same

concentration did not affect the glucose kinetics, suggesting that infusion of glucose into the

jejunum activates local signaling mechanisms to lower glucose production402. Similar to lipids,

the downstream hormonal signaling involved in jejunal glucose-induced suppression of glucose

homeostasis remains to be assessed. However, this may also involve CCK or other gut derived

hormones, which is a focus of this current thesis.

The involvement of a gut-brain-liver axis was then tested for jejunal nutrient sensing.

Similar to the duodenum, blockade of gut to brain signaling via infusion of the anesthetic

tetracaine abolished both lipid and glucose-induced suppression of glucose production402.

Further, NTS administration of MK-801 or hepatic vagotomy negated the glucose production

suppression effects of lipids and glucose402. Thus, a gut-brain-liver neuronal axis also exists for

jejunal nutrient sensing as seen in the duodenum.

Given that the direct disruption of duodenal nutrient sensing mechanisms results in a

dysregulation of glucose homeostasis during a fasting and refeeding protocol, the same

experimental procedure was performed with or without blockade of jejunal nutrient sensing

mechanisms to address the relevance of jejunal nutrient sensing. Interestingly, blockade of either

nutrient (lipid and glucose) sensing in the jejunum did not disrupt glucose homeostasis during

the refeeding study402. This suggests that nutrient sensing mechanisms in the jejunum may

become apparent under conditions of disrupted nutrient flow such as when sections of intestine

are surgically removed for either cancer or bariatric surgical procedures. Thus, the ability of

jejunal nutrient sensing mechanisms to regulate glucose homeostasis was tested after duodenal-

Page 52: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

39

jejunal bypass surgery. This surgical technique, as well as the findings of the involvement of

jejunal nutrient sensing to mediate the beneficial effects of this surgery, will be discussed in

greater detail below. Before such discussion, the different types of bariatric surgical procedures

as well as changes in gut peptide hormone secretion associated with bariatric surgery will be

reviewed first.

1.5 Bariatric surgery, gut hormones and intestinal nutrient sensing

1.5.1 Bariatric surgical procedures and changes in gut hormones

Bariatric surgery encompasses many surgical procedures that are either restrictive in

nature (i.e. gastric banding or vertical sleeve gastrectomy) by altering the stomach size or

nutrient flux into the stomach, or in addition to changing the stomach size, bypass sections of

the small intestine thus altering the amount of nutrients entering the stomach and the intestinal

tract (i.e. Roux-en-Y gastric bypass). Bariatric surgery was primarily used as a weight loss

procedure for obese subjects (BMI > 35). Indeed, these surgical procedures have profound

weight loss effects, where gastric banding results in ~20% weight loss, and Roux-en-Y gastric

bypass (RYGB) results in ~25% weight loss403–405. In addition to the dramatic weight loss

effects of these surgeries, surgeons noticed that many patients with type 2 diabetes who had

undergone the surgery for morbid obesity experienced complete diabetes remission. Indeed,

bariatric surgery normalizes glucose levels in type 2 diabetes, and these effects have been shown

to be independent of weight loss406. Excitingly, one study indicates that bariatric surgery caused

diabetes remission, and this effect is still present even after 6 years407. In addition, bariatric

surgery has been demonstrated to reduce the risk of developing diabetes by 80% over 7 years408.

Given such success, there has been a world-wide effort to i) better understand which surgical

procedures have the best results and ii) begin to uncover the mechanism(s) of the surgery in

hopes to discover molecular candidates that can be targeted to mimic the beneficial effects.

Many scientists have focused on changes in gut hormone secretion after surgery as potential

Page 53: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

40

candidates for mediators in both the weight loss and glucose lowering effects. Indeed, changes

in gut hormone profiles have been demonstrated which, in addition to the four main surgical

procedures currently used in patients, are described below.

1.5.1.1 Common types of bariatric surgical procedures and beneficial outcomes

1.5.1.1.1 Laparoscopic adjustable gastric band surgery

The laparoscopic adjustable gastric band (LAGB) surgery is characterized by the

insertion of a synthetic band just below the gastro-esophageal junction that creates a gastric

pouch. In order to control the amount of food entering the stomach, the band size can be

changed through inflation or deflation409. Thus patients can limit their caloric intake and delay

gastric emptying into the small intestine. Typically used for its weight loss effects, LAGB is

shown to cause substantial body weight loss, however this depends on the starting BMI410. This

surgical procedure has very little complication associated with it compared to the other surgical

interventions described below and is the safest of all of procedures. In addition to its weight loss

effects, patients with mild obesity and type 2 diabetes underwent remission following LAGB410.

However, not all patients experience weight loss with this surgery, a common complaint for

LAGB.

1.5.1.1.2 Sleeve Gastrectomy

The sleeve gastrectomy (SG) procedure involves removal of 80% of the stomach

creating a small stomach pouch. This procedure originated from another form of bariatric

surgery, the bilio-pancreatic diversion/duodenal switch, as surgeons noticed that substantial

weight loss occurred before the second part of the procedure was performed409. This procedure

has substantial weight loss effects for both morbidly obese and extremely obese patients411. The

potential risk associated with this particular procedure is B12 deficiency409 but initial data

suggests that diabetes remission has occurred for some patients412.

Page 54: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

41

1.5.1.1.3 Roux-en-Y Gastric Bypass Surgery

Roux-en-Y gastric bypass (RYGB) surgery is the most commonly used bariatric surgical

procedure and accounts for ~60% of bariatric surgical procedures conducted in the United

States411. This procedure was developed in the 1970s and was modified to its current form.

There are two components to this surgical procedure: a restrictive and malabsorptive

component. The restrictive element of the procedure involves reducing the size of the stomach

by creating a gastric pouch out of the upper portion of the stomach. The malabsorptive

component involves changing the intestinal tract as follows: the jejunum is divided into two

limbs, the upper bilio-pancreatic limb and a lower limb (also called the Roux limb). The Roux

limb is brought up and connected to the restricted stomach, which results in bypassing nutrient

entry into the duodenum and proximal jejunum. The bilio-pancreatic limb is then connected to

the Roux limb through a distal jejunostomy, and delays the interaction of food coming into

contact with pancreatic enzymes and bile409. This is one of the most complicated procedures

surgically, but has substantial effects on diabetes remission410. However, one of the negative

consequences of this surgery is “dumping syndrome” (as the pyloric sphincter is removed)

which encompasses a group of symptoms including weakness and abdominal discomfort and

sometimes increased bowel evacuation after ingestion of a meal. Interestingly, although more

invasive surgically, there are less complications associated with RYGB in comparison to LAGB.

1.5.1.1.4 Bilio-pancreatic diversion/duodenal switch

The bilio-pancreatic diversion/duodenal switch (BPD/DS) was developed by the

combination of two different surgical procedures413–415. The restrictive component of this

surgery involves partial removal of the stomach as well as a change in stomach curvature. In

contrast to RYGB, this surgical procedure keeps the pyloric sphincter of the stomach intact

which eliminates dumping syndrome as a complication. Similar to RYGB, the malabsorptive

Page 55: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

42

component of this surgery involves the separation of food to digestive enzymes and bile. Further

down the intestinal tract, these separated intestinal components are rejoined into a common tract

where food, bile and enzymes join allowing for limited fat absorption. This is the least common

form of bariatric surgical procedures conducted even though there are substantial weight loss

effects, even greater than RYGB416. In addition to weight loss, there is excellent diabetes

resolution410. However, this procedure has the largest mortality rates and poses the greatest risk

for nutritional deficiencies409.

1.5.1.2 Changes in gut hormones

The resolution of diabetes following gastric bypass surgeries is thought to be explained

by the “foregut/hindgut” hypothesis. The foregut hypothesis states that by excluding the

proximal small intestine, there is a reduction in some negative/anti-incretin hormone, which

consequently improves glucose control. The hindgut hypothesis states that by excluding the

proximal portion of the small intestine, there is an increase in the secretion of distal

hormones417. Indeed, many studies focus on GLP-1 levels after bypass surgery. After RYGB

there is an increase in circulating GLP-1 levels418–420 which is thought to be a potential mediator

of the weight loss and glucose lowering effect of this surgery, and this increase is higher than in

patients who received gastric banding421. However, not all studies are consistent in their findings

in regards to GLP-1 levels, which may be due to the fact that the precision of GLP-1 assays can

vary, or that GLP-1 measurements were taken during fasting conditions422–424. Moreover,

changes in PYY levels have been seen after RYGB surgery in comparison to other surgeries,

and have been shown to increase within 2 days and remain elevated up to 24 months after

surgery425. Similar to GLP-1, the increase in PYY levels is greater in individuals who have

received RYGB in comparison to other forms of bypass surgery421. Another hormonal change

seen after both RYGB and SG is a reduction in circulating ghrelin levels426,427 which is not

Page 56: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

43

surprising given that both surgical techniques have a stomach-reducing component to the

surgery. However, other studies saw an increase in ghrelin levels following RYGB428 and thus

the involvement of ghrelin in gastric bypass surgery remains controversial. In addition to GLP-

1, PYY and ghrelin, other studies in humans suggest that GIP may be involved. However, the

results among different studies are not consistent, some demonstrating and increase429, decrease

or no changes of postprandial levels of GIP430. Circulating leptin levels have also been assessed

in patients are RYGB and have consistently been found to be lower after surgery, likely due to a

decrease in body fat431,432. Thus, it is evident that the exact gut hormone profile found after

gastric bypass surgery remains controversial and warrants further investigation.

It is clear that data collected in human studies is limited to correlative findings and

remains inconclusive. Thus, the use of animal models helps to dissect the potential mechanisms

responsible for the resolution of diabetes after bariatric surgery. However, similar to findings in

humans, studies in rodents are also controversial with different findings amongst different

groups. In regards to RYGB surgery, it is suggested that changes in GLP-1 may mediate the

beneficial effects of the surgery433. However, the finding that RYGB still has beneficial effects

in GLP-1R knock out rodents434 questions this hypothesis. However, another group suggests

that rodents may have different responses to gastric bypass surgery due to differences in GLP-1

responsiveness435. Thus GLP-1 may indeed play a role but is likely not the sole mediator of the

beneficial effects of the surgery. Moreover, an increase in PYY concentrations has also been

suggested to improve glucose homeostasis436, although studies in Y receptor knockout models

are lacking. Therefore, the relative contribution of GLP-1 and PYY signaling in mediating the

beneficial effects of RYGB remains unresolved. Moreover, similar conflicting results are seen

after SG surgery where changes in both GLP-1 and ghrelin are postulated to mediate the

beneficial effects of this surgery437. However, the use of receptor knock out models suggests

otherwise, as this surgical procedure still has its beneficial effects in both ghrelin438 and GLP-

Page 57: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

44

1R439 knock out rodents. Thus, the exact mechanisms through which these surgeries exert their

beneficial effects are still largely unknown. It is likely a combination of different hormonal

signaling tied in with the complexity of diabetes that creates difficulties in searching for

nonsurgical tools to recapitulate the effects of these surgeries.

Given the fact many forms of bariatric surgery change the anatomy of the intestinal tract,

and the findings that intestinal nutrient sensing mechanisms trigger a gut-brain-liver axis

independent of weight loss, intestinal nutrient sensing may be involved in mediating some of the

beneficial effects of bariatric surgery. In order to test this hypothesis, a surgical procedure that

only modifies the intestinal tract is needed, which will be described in more detail below.

1.5.2 Duodenal jejunal bypass surgery, nutrient sensing and beyond

Modified from: Rasmussen, BA*, Breen, DM*, Côté, CD, Jackson, M, and Lam, TK. Nutrient sensing mechanisms in the gut as therapeutic targets for diabetes. Diabetes 62, 3005-3013, 2013 *Equal contribution

Duodenal jejunal bypass (DJB) surgery involves repositioning the intestinal tract without

restriction or exclusion of the stomach. More specifically, this procedure first involves exclusion

of the duodenum and proximal jejunum, and connection of the distal jejunum to the stomach.

Thus, nutrients from the stomach bypass the duodenum and enter directly into the jejunum. This

surgical procedure has been shown to have glucose lowering effects in non-obese rodents440 and

in non-obese or mild-obese humans with type 2 diabetes441–444, independent of weight loss. This

experimental form of bypass surgery is conducted in order to tease out the stomach restricting

effects from the intestinal specific effects. Thus, what remains in question is whether intestinal

nutrient sensing mechanisms are mediating the glucose lowering effects of this surgery.

In this regard, DJB surgery was conducted in two different models of non-obese

uncontrolled insulin deficient diabetes402. The first model involved injection of streptozotocin

(STZ), a cytotoxic agent that selectively kills pancreatic β cells and reduces insulin

Page 58: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

45

concentrations by ~80%. Due to low insulin levels, these rodents display fasting and fed

hyperglycemia. DJB surgery in this model rapidly lowered plasma glucose levels which was

independent of changes in circulating insulin and glucagon levels402. Similar results were seen in

the insulin deficient, nonobese diabetes-prone BioBreeding (BB-dp) rats that spontaneously

develop type 1 diabetes. Importantly, the glucose lowering effect induced by this surgery is

mediated by jejunal nutrient sensing mechanisms (Figure 1.2). This is demonstrated by the fact

that 2 days after DJB surgery, blocking jejunal nutrient sensing mechanisms during a fasting and

refeeding study resulted in a rise in glucose levels402. To further confirm these findings, jejunal

nutrient infusions were conducted in STZ rodents without DJB surgery, which lowered plasma

glucose levels as well as glucose production. Importantly, these findings are consistent with

those seen in mild-obese type 2 diabetic humans441. That is, the glucose response during a

refeeding study in these rodents while blocking nutrient-sensing mechanisms402 closely

resembled the glucose response during an oral glucose tolerance test in patients before

surgery441. This suggests that nutrient sensing mechanisms may a play a role in the glucose

lowering effect of bariatric surgery in humans. Whether gut-derived hormones play a role in

mediating the early anti-diabetic effects of DJB surgery, in addition to nutrient sensing

mechanisms, is currently unknown, which is a focus of this current thesis.

Given that jejunal nutrient sensing has been demonstrated to trigger a gut-brain-liver

neuronal axis to lower glucose production, it is likely that such an axis is activated after DJB

surgery to lower glucose production402. This is consistent with other groups’ findings that

demonstrate that DJB64 or a variant of DJB445 lower blood glucose concentrations and glucose

production through the CNS in type 2 diabetic rodents. As discussed above, DJB surgery

lowered glucose levels in insulin deficient rodents suggesting that an increase in circulating

insulin levels does not account for the early glucose lowering effect402. Furthermore, the glucose

production lowering effect seen in obese type 2 diabetic rodents was independent of an

Page 59: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

46

improvement in insulin action64. However, it is important to note that these findings do not

exclude the possibility that DJB surgery lowers blood glucose concentrations via increased

insulin-dependent or independent glucose uptake in a more long term setting, as it has been

suggested that an improvement in β cell function occurs after surgery in obese type 2 diabetic

subjects446.

As stated above for RYGB, it is thought that changes in insulin secretion are accounted

for by changes in circulating GLP-1, although this is controversial447. Complicating matters

more, in STZ induced uncontrolled diabetic rats, DJB surgery lowered glucose levels in

association with a rise in circulating GLP-1 levels. However, in BB-dp rats with DJB surgery,

the rapid glucose lowering effect was seen without changes in GLP-1 concentrations. Other

studies also reports no changes in GLP-1 after DJB in Zucker Diabetic Fatty rats437, and Goto-

Kakizaki rats448, similar to the findings in BB-dp rodents. Moreover, bariatric surgery still has

profound effects on glucose tolerance in high fat fed rodents deficient of GLP-1Rs, as discussed

previously. Thus, the relative contribution of GLP-1 in mediating the glucose lowering effect of

DJB surgery remains to be resolved. The focus of this current thesis is to address whether other

gut-derived hormones play a role in the improvement in glucose regulation following DJB

surgery.

In addition to increasing distal gut peptide secretion, altering the intestinal tract during

this surgical procedure also alters the mixing of bile with nutrients in the proximal small

intestine and thus may alter bile acid levels. Bile acids have recently been implicated in playing

a role in glucose homeostasis through their effects on glucose production and increased glucose-

induced insulin secretion330,449. Bile acids are postulated to play a role in mediating the

beneficial effects of RYGB in dogs450 and an increase in circulating bile acids has been detected

in humans450 after bypass surgery. Indeed, DJB and other bariatric surgical procedures

Page 60: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

47

performed in rodents have been associated with an increase in bile acids levels64,451, and the

glucoregulatory as well as body weight regulatory effects of SG were abolished in nuclear

receptor FXR knockout mice452. It still remains in question whether bile acids action is required

for the rapid glucoregulatory effects of DJB surgery.

Interestingly, less invasive procedures that mimic DJB surgery have been developed

such as the duodenal endoluminal sleeve, which involves inserting a flexible tube that inhibits

the interaction of nutrients with the duodenum and has been shown to have similar effects on

glucose regulation451. This is a step in the right direction in regards to finding less invasive ways

to lower glucose levels in diabetes. Nonetheless, a lot of work still remains in order to uncover

the mechanisms of this surgery. By doing so, we may be able to find noninvasive target

strategies to improve the lives and outcomes for patients who are diabetic and/or obese.

Page 61: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

48

Figure 1.2 Duodenal and jejunal nutrient sensing mechanisms trigger a gut-brain-liver neuronal axis to lower glucose production. Upon lipid entry into the duodenum a LCFA-CoA ! PKC-δ ! CCK ! CCK1 receptor signaling pathway activates vagal afferents to signal to the NTS to activate NMDA receptors to lower glucose production. This duodenal pathway is abolished upon high fat feeding for three days. Like the duodenum, the more distal intestine, the jejunum, is capable of sensing both glucose and lipids to trigger a neuronal network to lower glucose production, which is required for the early anti-diabetic effect of DJB surgery. Adapted from Breen, DM* and Rasmussen, BA* et al. (2013) Diabetes 62, 3005-3013 *Equal contribution. Permission to reproduce this figure has been obtained from the copyright owner: American Diabetes Association

Page 62: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

49

1.6 Summary of Introduction

Diabetes and obesity are characterized by a variety of factors including the dysregulation

in food intake and glucose homeostasis. The intestine is the first line of defense against nutrient

excess and activates local hormonal signaling to regulate glucose levels as well as satiety. More

recently, the duodenum has been demonstrated to sense lipids to trigger the release of CCK to

lower glucose production via a gut-brain-liver axis. However, this signaling pathway is impaired

upon short-term high fat feeding, suggesting duodenal CCK resistance. The downstream

signaling of the CCK-1 receptor to trigger this neuronal network remains unknown as well as

whether direct activation of these signaling molecules could bypass CCK resistance. Like the

duodenum, the jejunum is also capable of sensing nutrients and has been shown to activate a

gut-brain-liver neuronal axis and mediate the glucose lowering effect of DJB surgery. Whether

other gastrointestinal hormones trigger a similar axis and contribute to this glucoregulatory

effect also warrants future investigation.

1.6 Rationale and Significance of the Studies

Diabetes is a worldwide epidemic with the number of individuals affected by the disease

increasing at an alarming rate, in large part due to the combination of genetic and lifestyle

factors453. Diabetes and/or obesity are often characterized by hepatic insulin resistance,

reduced/altered insulin secretion, muscle insulin resistance and increased glucose production,

where fasting hyperglycemia in type 2 diabetes has been shown to be due largely to an increase

in glucose production59. Chronic hyperglycemia can lead to diabetic complications such as

neuropathy, nephropathy, and retinopathy454. Thus, uncovering novel mechanisms that lower

glucose production in diabetes or obesity will unveil therapeutic targets to lower glucose levels

and reduce the risk of diabetic complications.

Page 63: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

50

It has been demonstrated that a duodenal lipid ! LCFA ! LCFA-CoA ! PKC-δ !

CCK ! CCK-1 receptor pathway triggers a gut-brain-liver axis to lower hepatic glucose

production379,383,384,391. Furthermore, direct administration of CCK-8 into the duodenum fails to

lower glucose production in rats fed a high-fat diet384. Although the site(s) of this defect remains

unclear, evidence confirms that it is located downstream of CCK release, as both lipid

administration379 and PKC-δ activation391 (both of which stimulate CCK release) still fail to

lower glucose production. Whether this resistance lies at the level of the CCK-1 receptor and/or

within the signaling cascade of the receptor currently remains to be explored. The purpose of

Study 1 in this thesis was to address the downstream signaling of the CCK-1 receptor, namely

PKA, to regulate glucose production, and to determine whether direct activation of PKA can

bypass duodenal CCK resistance in rodents fed a high fat diet for 3 days.

Like the duodenum, the jejunum is capable of sensing nutrients to trigger a gut-brain-

liver neuronal axis to lower glucose production402. These nutrient sensing mechanisms become

apparent after DJB surgery, whereby the influx of nutrients into the jejunum lowers glucose

levels. Whether hormonal action mediates this glucose production-lowering effect remains

unknown. The purpose of Study 2 in this thesis was to address whether leptin (produced by the

stomach) action in the intestine triggers a neuronal network to lower glucose production and

whether intestinal leptin action mediates the glucose lowering effect of DJB surgery.

The pancreatic (basal insulin) euglycemic pancreatic clamp technique in combination with

intestinal infusion of various compounds was performed in both normal and diseased rats. These

studies provide evidence for PKA as a duodenal target to lower glucose production in high fat

diet fed rodents and for the possible role of jejunal leptin signaling in mediating the beneficial

effects of DJB surgery.

Page 64: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

51

1.7 General Hypothesis

Independent CCK and leptin signaling in the intestine triggers a neuronal network to lower

glucose production.

1.8 Specific Aims

This thesis consists of two studies that examined intestinal hormonal signaling involvement

in the regulation of glucose production and whether these signaling axes remain intact in

diseased settings.

Study 1. To determine whether duodenal PKA activation plays a role in the CCK1 receptor

mediated decrease in glucose production and whether direct activation of duodenal PKA

bypasses duodenal CCK-resistance acquired upon high fat feeding.

Study 2. To investigate whether leptin activates jejunal Leprb-mediated signaling

pathway(s) to regulate glucose production via the central nervous system and whether enhanced

gastric leptin action in the jejunum contributes to the glucose-lowering effect of DJB surgery in

non-obese uncontrolled diabetes.

Page 65: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

52

Chapter 2General Methods

2.1 Animals All animal study protocols were reviewed and approved by the Institutional Animal Care

and Use Community of the University Health Network. For all studies, adult male (8-week old)

male Sprague-Dawley (SD) rats (~300g) were obtained from Charles Rivers Laboratories

(Montreal, Quebec, Canada). Rats were individually housed and maintained on a standard 12-

12h light dark cycle, and had ad libitum access to water and rat chow (Harlan Teklad 6%

mouse/Rat diet; composition: 49% carbohydrate, 33% protein and 18% fat; total calories

provided by digestible nutrients: 3.1 kcal/g). Rats were given at least 5 days to acclimatize upon

arrival before surgeries were performed.

2.1.1 High Fat Feeding Animal Model

A subgroup of male SD rats were placed on a lard-oil enriched high fat diet ad libitum

for three days after intestinal and vascular catheter implantation (see Table 1 for high fat diet

and standard chow composition; Ren’s Pet Depot, ON, Canada). Rats that were hyperphagic and

consumed more calories as rats on regular chow were used for the clamp experiments. These

rats have previously been shown to develop hepatic392,393 and hypothalamic394 insulin resistance

and duodenal Intralipid379 and CCK384 resistance.

2.2 Surgical Procedures

Rats were first anesthetized with an i.p. cocktail of (60-90 mg/kg) ketamine (Ketalean;

Bimeda-MTC, Cambridge, Ontario) and (8-10mg/kg) Xylazine (Rompun; Bayer) before

performing surgical procedures described below. All surgical procedures were preceded through

Page 66: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

53

shaving both the abdominal and neck area and cleaning with 70% ethanol and 10% povidone-

iodine (Betadine solution, ON, Canada) before incisions were made. Recovery from surgical

procedures was ensured through monitoring body weight gain and food intake for 4-6 days after

the surgery.

2.2.1 Vessel Cannulation

Indwelling catheters were made with polyethylene tubing (PE 50, Clay Adams, Boston,

MA) with a cuff extension (15mm, internal diameter of 0.02 inches) of Silastic tubing (Dow

Corning, Midland, MI). After blunt dissecting through the muscle layers, the carotid artery was

isolated from connective tissue and the vagus nerve. Using a 4-0 silk thread, the exposed vessel

was ligated at the cranial end. At the caudal end, another thread was loosely tied and the two

ligatures were pulled taut. A small incision was made into the vessel wall. The indwelling

catheter was then inserted past the overlap and the catheter was secured through tightening the

loose ligature. Blood withdrawal and infusion were tested from the catheter. The same

procedure was conducted for the right internal jugular vein. After insertion, the catheters were

tunneled subcutaneously with a 16G needle and filled with a 10% heparin mixture (saline with

1000 U/ml of heparin) to maintain patency of the cannula and closed with a metal pin until the

day of the procedure.

2.2.2 Intestinal Cannulation

Duodenal and jejunal cannulation surgeries were performed as described379,384,402. Three

to four days before the clamp studies, exposure of the gastrointestinal tract within the

peritoneum was conducted through a laparotomy incision made on the ventral midline as well as

the abdominal muscle wall. After identifying the pyloric sphincter, the duodenum was identified

as 1.5 cm distal to the sphincter. In separate rats, the jejunum was identified as 8–10 cm from

the Ligament of Treitz. With a 21-gauge needle, a small hole was made on the ventral aspect of

Page 67: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

54

the duodenum or jejunum (in a region with the least vascularization to minimize bleeding) to

allow insertion of an intestinal catheter made of silicone tubing (0.04 in ID, 0.085 in. OD; Sil-

Tec, Technical Products, USA) with a 0.2 cm extension of smaller silicone tubing (0.025 in ID,

0.037 in. OD; Sil-Tec, Technical Products, USA). To ensure the cannula was placed in the

lumen of the duodenum, the cannula was flushed with saline. In order to ensure the catheter

remained in place after surgery, it was anchored to the outer serosal surface of the duodenum or

jejunum with 3M adhesives (Vetbond) and a 0.5 cm2 piece of Marlex mesh sewn to the surface

with a 6-0 silk suture. Through the laparotomic incision, the proximal portion of the catheter

exited the abdominal cavity and the abdominal wall was closed with a 4-0 silk suture. At the

back of the neck, a 2 cm midline incision was made in the skin, rostral to the interscapular area,

and the cannula was tunneled subcutaneously to exit the incision. This 2 cm incision was sewn

closed with 4-0 silk sutures and the proximal portion of the cannula was closed with a metal pin.

The cannula was flushed daily with 0.1 ml of saline to ensure patency on the day of the clamp

studies.

2.3 Pancreatic Euglycemic (Basal Insulin) Clamp Technique

The night before the in vivo clamp experiments, the rats were restricted to ~57 kcal to

ensure the same post-absorptive nutritional status. The total length of the experiment was 200

minutes. At t = 0, a primed-continuous infusion of [3–3H] glucose (Perkin Elmer, MA, USA; 40

µCi bolus; 0.4 µCi/ min) was initiated and maintained until t = 200 min to assess glucose

kinetics based on the tracer-dilution methodology. Blood samples were collected in heparinized

tubes at 10 minute intervals and subjected to centrifugation at 6000 rpm to separate the plasma

and plasma glucose was measured as described below (2.8.1 Plasma Glucose) to obtain basal

glucose readings (t = 60-90 min). At t = 90 min until the end of the experiment (t = 200) a

pancreatic (basal insulin) clamp was initiated by providing a continuous insulin (1.2

Page 68: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

55

mU/kg/min; porcine insulin; Sigma-Aldrich, St. Louis, MO, USA) and somatostatin (3

µg/kg/min; Bachem, CA, USA) infusion to inhibit endogenous insulin and glucagon secretion.

After initiation of the pancreatic clamp, a variable infusion of a 25% glucose solution (45%

glucose; Sigma-Aldrich, St. Louis, MO, USA) was provided to maintain basal plasma glucose

levels (t = 60-90 min) and adjusted every 10 minutes if needed. From t = 150 to t = 200,

intraduodenal or intrajejunal infusions (0.01 ml/min) were performed. Additional samples were

obtained at the 10-minute intervals for the determination of [3–3H] glucose specific activity,

insulin, and leptin levels (see 2.8.2-2.8.4 for details). Rats were anesthetized at the end of the

experiments through a direct infusion of ketamine into the jugular vein and portal plasma

samples were taken followed by tissue collection. Tissues were freeze-clamped in situ with steel

tongs pre-cooled in liquid nitrogen. All tissue samples were stored at –80 ºC and plasma

samples were stored –20 ºC until use. The Harvard Apparatus PHD 2000 infusion pumps (MA,

USA) were used for all infusions during the clamp.

2.4 Protein Assay

The Thermo Scientific Pierce 660nm Protein Assay (Thermo Scientific, IL, USA) was

used to measure the protein concentration of different tissue samples with BSA used as a

standard. This assay is a colorimetric assay based on the binding of a dye-metal complex to

protein under acidic conditions, which causes a shift in the dye’s maximum absorption,

measured at 660nm. The color produced by the assay is stable and the color increases in

intensity with increasing protein concentration. The tissue samples were aliquoted for the

protein assay, thawed, vortexed and kept on ice. The samples were diluted 1:20 with distilled

water in an eppendorf tube. Standards were prepared using stock BSA (2 mg/ml) diluted with

distilled water to prepare a curve ranging from 0 to 2 mg/ml. 10 µl of the BSA standards were

transferred to a 96 microwell plate in duplicate. Then, 10 µl of the diluted tissue samples were

Page 69: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

56

added to the plate in duplicate. 150 µl of the Thermo Scientific Pierce 660nm Protein Assay

Reagent was added to each well and allowed to change color. After 5 minutes, the plate was

transferred to a spectrophotometer and the absorbance was read at 660nm. Through

interpolation, the protein concentrations of the tissue samples were determined.

2.5 Biochemical Analyses

2.5.1 Plasma Glucose

The measurements of plasma glucose concentrations were conducted by the glucose

oxidase methods using a GM9 Analox Glucose Analyzer (Analox Instruments, Lunenburg,

MA). Blood samples were collected into heparinized tubes and centrifuged at 6000 rpm to

separate the plasma. Upon calibration of the analyzer with a provided standard, a 10 µl D-

glucose containing plasma sample was pipetted into the reaction well containing a solution with

glucose oxidase and oxygen. The following reaction occurs after injection of a sample:

β-D-Glucose + O2 Glucose oxidase

D-gluconic acid +H!O!

The rate of oxygen consumption is proportional to the amount of glucose in the plasma sample.

A polarographic sensor measures the rate of oxygen consumption to determine the plasma

glucose concentration. More specifically, the partial pressure of oxygen in the sample is

measured as Clark-type amperometric oxygen electrodes are immersed in the sample and a

potential is applied between them that reduces dissolved oxygen at the working electrode.

Results are obtained within 20 seconds of inserting the sample into the apparatus.

2.5.2 Plasma Glucose Tracer Specific Activity

50 µl of plasma was used to determine the specific activity of [3-3H] in the plasma. The

samples were first deproteinized by the addition of 100 µl of Ba(OH)2 and ZnSO4 followed by

vortexing and centrifugation at 6000 rpm for 5 minutes at 4°C. The supernatant of each sample

was transferred to scintillation vials and evaporated to dryness to remove tritiated water (since

Page 70: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

57

tritium on the C-3 position of glucose is lost to water during glycolysis). Thus, radioactivity

represents the [3-3H] glucose in the plasma only. Scintillation fluid (Bio-Safe Scintillation

Cocktail, Research Products International Corp., Mount Prospect, IL, USA) was added to the

dried sample to detect the radioactive signal and counted in a LS6500 Multipurpose Scintillation

Counter (Beckman, USA).

2.5.3 Plasma Insulin

A radioimmunoassay (RIA) was used to determine plasma insulin concentrations using a

rat insulin kit (100% specificity) from Linco research (St. Charles, MO). The antigen-antibody

binding principle is used in the RIA. Briefly, the amount of insulin present in the plasma sample

is in competition for binding to antibodies raised against insulin (guinea pig anti-rat insulin

antibody) with a labeled tracer antigen (125I labeled insulin). Thus the amount of radiolabeled

125I-labeled insulin that binds is in reverse proportion to the amount of known standards and the

amount of insulin in the plasma sample. Separation of the 125I-labeled insulin and unbound

fractions is conducted through the use of a double antibody solid phase.

Specifically, a 2-day protocol as per the supplier’s instructions was used. First, the

generation of standard curve is constructed with the use of 50 µl of standards with a range of

known concentrations (0.1, 0.2, 0.5, 1.0, 2.0, 5.0 and 10.0 ng/ml). Then 50 µl of the plasma

samples was pipetted into appropriate tubes and the addition of 50 µl of 125I-labeled insulin and

50 µl of the rat insulin antibody is added to both the standards and samples, and were vortexed.

1.0 ml of precipitating reagent is added after overnight incubation at 4°C followed by vortexing

and incubation at 4°C for 20 minutes. To pellet the bound insulin, the samples were then

centrifuged. A gamma counter (Perkin Elmer 1470) is used to count the radioactivity of the

pellet. The radioactivity counts (B) for the standards and samples are expressed as a percentage

of the mean counts of total binding reference tubes (B0):

Page 71: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

58

% total binding=%BB0

= Standard or sample

B0 x 100%

A standard curve is constructed by plotting the % !!!

for each standard against the known

concentration. Through interpolation, the concentration of the insulin samples was determined.

2.6 Calculations

During the pancreatic clamp experiments, a radioactive [3–3H] glucose tracer was

infused at a constant rate to allow for equilibration of the tracer glucose with the glucose in the

body. After equilibration, using the steady state formula, glucose production and uptake can be

determined. That is, in the steady state basal condition, the rate of glucose uptake (Rd) is equal

to the rate of glucose appearance (Ra) or rate of endogenous glucose appearance. Thus, using

the steady state formula, the Ra and Rd can be can be determined by the following equation:

Ra=Rd=Constant tracer infusion rate ( µCimin )

Specific activity ( µCimg )

During the pancreatic clamp where an exogenous glucose infusion is given to maintain

euglycemia, glucose production is calculated by subtracting the exogenous glucose infusion rate

from the Rd:

Ra=Rd-Glucose Infusion Rate

2.7 Statistical Analysis

Data are presented as means + SEM. When a comparison was made between two

groups, an unpaired Student’s t-test was performed. Where comparisons were made across

more than two groups, analysis of variances (ANOVA) was performed, and if significant, this

was followed by Tukey’s post-hoc test, which enabled comparisons of all treatment groups. A

probability of P < 0.05 was accepted as significant. The statistical software program Prism

(GraphPad Software Inc., CA, USA) was used for statistical calculations.

Page 72: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

59

Table 2.1 Diet content of the regular chow and lard-oil enriched high fat diet.

Page 73: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

60

Chapter 3Study 1

Duodenal Activation of cAMP-Dependent Protein Kinase Induces Vagal Afferent Firing and Lowers Glucose Production in Rats

Modified From: Rasmussen, BA, Breen, DM, Luo, P, Cheung, GW, Yang, CS, Sun, B, Kokorovic, A, Rong, W, and Lam, TK. (2012) Duodenal activation of cAMP-dependent protein kinase induces vagal afferent firing and lowers glucose production in rats. Gastroenterology 142, 834-843 Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: Elsevier Limited

Page 74: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

61

3.1 Abstract

Background and Aims: The duodenum detects a rise in nutrients to maintain energy and

glucose homeostasis. However, the signaling and neuronal mechanisms involved still remain

unknown. In the present study, we examined whether activation of adenosine 3’,5’-cyclic

monophosphate (cAMP)-dependent protein kinase A (PKA) in the duodenum lies downstream

of CCK to trigger vagal afferent firing and regulate glucose production. Methods: We

selectively activated duodenal PKA in rats through a duodenal infusion of a PKA activator (Sp-

CAMPs) and assessed changes in glucose kinetics during the pancreatic (basal insulin)

euglycemic clamps and vagal afferent firing. To assess whether duodenal PKA signaling is

required for glucose regulation, PKA activation induced through infusion of Sp-CAMPS or a

CCK1 receptor agonist (CCK-8) was blocked through co-infusion of two independent cell-

permeable PKA inhibitors H-89 and Rp-CAMPs. We also tested whether a neuronal network is

required and if the gluco-regulatory effects of duodenal PKA activation remain intact in rats fed

a high fat diet. Results: In normal rats, an intraduodenal infusion of Sp-CAMPs increased both

PKA activation and vagal afferent firing and lowered glucose production. Co-infusion of Sp-

CAMPs with H-89 or Rp-CAMPs (PKA inhibitors) negated the metabolic and neuronal effects

of duodenal PKA activation. The metabolic effects were also negated upon co-infusion with

tetracaine, inhibition (both molecular and pharmacologic) of NR1-containing NMDA receptors

within the DVC, or hepatic vagotomy. Duodenal CCK-8 infusion failed to lower glucose

production upon duodenal PKA inhibition, whereas duodenal CCK resistance in high fat diet fed

rats was bypassed upon duodenal Sp-CAMPs administration, which activated PKA and lowered

glucose production. Conclusions: A neural glucoregulatory function of duodenal PKA signaling

was identified.

Page 75: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

62

3.2 Introduction

It is approximated that a staggering 220 million people have type 2 diabetes with almost

half of this population living in China455,456. Diabetes and/or obesity are often characterized by

hepatic insulin resistance, reduced/altered insulin secretion, muscle insulin resistance and

increased glucose production, where fasting hyperglycemia in type 2 diabetes has been shown to

be due largely to an increase in glucose production. Chronic hyperglycemia can lead to diabetic

complications such as neuropathy, nephropathy, and retinopathy454. Thus, uncovering novel

mechanisms that lower glucose production in diabetes or obesity will unveil therapeutic targets

to lower glucose levels and reduce the risk of diabetic complications.

An acute rise in nutrients is detected by the duodenum to trigger negative feedback

systems to maintain peripheral homeostasis457. The absorption and metabolism of pre-absorptive

lipids, through activation of biochemical pathways within the duodenum concurrently inhibits

glucose production and food intake15. The underlying mechanisms of duodenal lipid metabolism

induced suppression of glucose production and food intake remain elusive. However the

secretion of CCK from the duodenal I-cells, and subsequent binding of CCK to its gut CCK1

receptors, are sufficient and necessary for lipids to trigger in parallel a gut-brain and a gut-brain-

liver axis to lower appetite163,167,198,458,459 and glucose production384, respectively. In addition to

glucose production regulation, CCK plays a role in digestion and improves nutrient absorption

by stimulating pancreatic amylase secretion, promotes bile release from the gall bladder, and

delays gastric emptying184. Importantly, the physiological relevance of duodenal CCK action in

glucose regulation is highlighted by the findings that either molecular or pharmacological

inhibition of duodenal CCK1 receptor signaling during refeeding disrupts glucose

homeostasis384.

Page 76: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

63

The CCK1 receptor is a G-protein coupled receptor460 which is mostly expressed in the

gut. Classical G-protein coupled receptor signaling involves both PKA and PLC signaling

activated by upstream G-proteins Gαs and Gαq, respectively. The CCK1 receptor signaling

pathways have been studied in the pancreatic acinar cell461 and both signaling pathways have

been described to mediate the direct CCK/CCK1 receptor signaling cascade in pancreatic

secretions461–464. To date, the signaling pathway mediating the duodenal CCK1 receptor induced

suppression of glucose production remains unknown. Whether PKA activation mediates CCK1

receptor signaling in the duodenum to regulate peripheral glucose homeostasis will be tested in

the current study.

Duodenal CCK-resistance is acquired in response to short term high fat

feeding5,384,458,459. Although the site(s) of this defect remains unclear, evidence confirms that for

glucose production regulation it is located downstream of CCK release, as both lipid

administration379 and PKC-δ activation382 (both of which stimulate CCK release) still fail to

lower glucose production. Thus, studies aimed at uncovering the duodenal CCK1 receptor

signaling cascade (i.e., PKA signaling) that regulates glucose production in normal and high-fat

fed rats will begin locating the molecular defects that occur in duodenal CCK-resistance. Such a

finding will potentially uncover novel signaling molecules within the duodenum that could be

targeted in diabetes and obesity to restore glucose homeostasis.

In the current study, we propose that direct activation of the duodenal PKA signaling

pathway is necessary for the CCK/CCK1 receptor and sufficient to trigger vagal afferent firing

to activate a neuronal network to lower glucose production in rats in vivo.

Page 77: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

64

3.3 Materials and Methods

3.3.1 Animal Preparation

Male SD rats weighing between 280-300g were obtained and maintained as described in

General Methods Section 2.1.

3.3.1.1 High Fat Diet Feeding

A subgroup of rats were fed a lard oil enriched high fat diet for three days. Rats that

were hyperphagic underwent the clamp studies. Please refer to General Methods 2.1.1 for details

on high fat feeding.

3.3.2 Animal Surgeries

3.3.2.1 Intestinal and vascular cannulation

Rats were anesthetized and a duodenal catheter was inserted 0.5 cm proximal to the

pyloric sphincter. A subgroup of rats underwent jejunal catheter placement (8-10 cm from the

Ligament of Treitz). After the intestinal cannulation, the jugular vein and carotid artery were

cannulated. Please refer to the General Methods Section 2.2.1 and 2.2.2 for details regarding

these surgical procedures.

3.3.2.2 Selective Hepatic Branch Vagotomy

A subgroup of rats underwent hepatic vagotomy surgery as previously

described379,384,402. On the ventral midline a laparotomy incision was made, followed by an

incision through the abdominal muscle wall to exposure the gastrointestinal tract within the

peritoneum. The stomach was gently retracted using sterile saline soaked cotton gauze to reveal

the descending esophagus and ventral subdiaphragmatic trunk. A 6-0 suture with a needle was

used to make a small puncture within the bottom portion of the stomach to allow easy

visualization throughout the surgery. The hepatic vagus of the ventral subdiaphragmatic vagal

trunk was transected by microcautery. This disrupts neural communication between the liver

Page 78: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

65

and brain. This also results in a slight decrease in the innervations to the intestine as the hepato-

duodenal sub-branch supplies a small portion of the intestine. Intestinal and vascular

cannulation surgeries were performed immediately after the vagotomy surgery.

3.3.2.3 Stereotaxic Surgery

For a subgroup of rats, implantation of a bilateral catheter targeting the nucleus of the

solitary tract within the dorsal vagal complex was performed. Specifically, after rats were

anesthetized, they were mounted onto a stereotaxic apparatus (David Kopf Instruments,

Tunjunga, CA) with ear bars and a nose piece set at +5.0mm. For implantation into the NTS, 26-

gauge stainless steel double guide cannulae were inserted using the following coordinates: 0.0

mm on the occipital crest, 0.4 mm lateral to the midline for both sides, and 7.9 mm below the

skull surface. The double guide cannula was secured with cyanoacrylate glue (HRS Scientific,

QC, Canada) and electric ortho-jet powder liquefied with Ortho-jet Acrylic liquid (Central

Dental, ON, Canada). After the glue and powder had hardened, the double guide cannula was

closed with a dummy cannula and dust cap (HRS Scientific, QC, Canada) until the day of the

experiment. Rats were given a week of recovery time after the stereotaxic surgery before

intestinal and vascular cannulation surgeries were performed.

3.3.2.3.1 Adenoviral Infection in the DVC after Stereotaxic Surgery

Immediately after the stereotaxic surgery, and before placement of the dummy cannula

and dust cap, a subgroup of rats received 3 µL of adenovirus (adenovirus containing short

hairpin RNA NR1: 4.0 x1011 plaque-forming units/mL; mismatch, 4.0 x 1011 plaque-forming

units/mL) per side of the cannulae over a 30-second injection using Hamilton syringes

(Hamilton Company, NV, USA). In order to prevent backflow, microsyringes were left in the

cannula for 20 minutes before removal. After removal, the dummy cannula and dust cap were

placed in and on the guide cannula, respectively, until the day of the clamp experiment. We

Page 79: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

66

have previously verified that direct injection of this adenovirus into the DVC knocked down the

NR1 subunit of the NMDA receptors382,465.

3.3.3 Intraduodenal Infusions and Treatments

The following substances were infused into the duodenum through a duodenal catheter during

the pancreatic clamp (t = 150-200) at a rate of 0.01 ml/min:

(1) saline

(2) Sp-cAMPS (PKA agonist; 30 µmol/L; Tocris Bioscience, Ellisville, MO, USA); a

subgroup of rats also received this treatment in the jejunum during the pancreatic clamp.

(3) H-89 (PKA antagonist; 12 µmol/L; Tocris Bioscience, Ellisville, MO, USA)

(4) Rp-CAMPS (PKA antagonist; 12 µmol/L; Tocris Bioscience, Ellisville, MO, USA)

(5) tetracaine (local anesthetic; 0.015 mg/min; Sigma-Aldrich, St. Louis, MO, USA)

(6) CCK-8 (35 pmol /kg/min; Sigma-Aldrich, St. Louis, MO, USA)

(7) MK-329 (CCK1 receptor antagonist; 1.6 µg/kg/min; Tocris Bioscience, Ellisville, MO,

USA)

Reagents #3, and 5-7 were dissolved in DMSO, #2 and #4 in distilled water. The rates for Sp-

CAMPs, H-89 and Rp-CAMPs were based on dose-response studies. The dose for the anesthetic

tetracaine and CCK-8 were based on previous studies379,384.

3.3.4 Pancreatic Euglycemic (Basal Insulin) Clamp Technique in Rats

Please refer to the General Methods section 2.3 for a detailed description of the clamp

procedure. Rats were restricted the night before the clamp experiment. A primed-continuous

constant infusion of [3–3H] glucose was given throughout the experiment (t = 200) to reach

steady state. The pancreatic clamp was then initiated at t = 90 where insulin (1.2 mU/kg/min)

and somatostatin (3 µg/kg/min) were infused at a constant rate. Blood samples were taken to

determine if a variable 25% glucose infusion was needed to maintain euglycemia. At t = 150, a

Page 80: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

67

duodenal infusion at 0.01 ml/min was conducted. A subgroup of rats received a jejunal infusion

from t = 150-200 minutes at 0.01 ml/min. Another group of rats received an MK-801 (NMDA

receptor antagonist; dissolved in 0.9% NaCl) infusion (0.03 ng/min; Sigma-Aldrich, St. Louis,

MO, USA) into the NTS at t = 90 until t = 200 at a rate of 0.006 µl/min using the CMA/400

syringe microdialysis infusion pump (Chromatographysciences, Montreal, QC, Canada), in

addition to the duodenal infusion.

3.3.5 Electrophysiological Ex Vivo Recordings of Duodenum Preparation

Just below the sphincter of Oddi (~5 cm long), the duodenum was removed from

anesthetized (80mg/kg pentobarbital i.p) male SD rats (200-250g). This duodenal tissue was

immediately placed in a recording chamber and subsequently perfused with oxygenated (95%

O2 + 5% CO2) Krebs’ solution (composition: NaCl 120 mM; KCl 5.9 mM, NaH2PO4 1.2 mM;

MgSO4 1.2 mM; NaHCO3 15.4 mM; CaCl2 2.5 mM; glucose 11.5 mM) at room temperature

with a Miniplus 3 perfusion pump (World Precision Instruments (WPI), USA) as previously

described466,467. Both ends of the duodenal segment were cannulated with two Genie syringe

pumps (WPI) connected in parallel to an intraluminal inflow cannula through a T-piece

connector to allow for perfusion of Krebs solution or test solutions through the lumen. In 15-

minute intervals, intraluminal infusions (9 mL/h) were conducted. Ramp distensions (up to 60

mm Hg) were performed using a three-way tap on the intraluminal outlet cannula, which was

closed while Krebs’ solution was perfused. Using a suction electrode, a branch of mesenteric

nerves, containing both spinal and vagal afferents, was dissected and pressure was recorded via

a pressure amplifier (NL 108, Digitimer, UK) and nerve activity was recorded with a Neurolog

headstage (NL100, Digitimer), and amplified (NL104) and filtered (NL 125, band pass 200-

3000 Hz). A Micro 1401 interface and Spike2 software (Cambridge Electronic Design, UK)

were used to acquire the nerve signal. An oscilloscope (Tektronix TDS 210) was used to display

whole nerve activity. The spontaneous afferent nerve discharge and distension-induced activity

Page 81: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

68

were allowed to become stable, and then the intraluminal infusion solution was switched to 30

µmol/L Sp-CAMPS (in Krebs’ solution, 9 ml/h for 30 minutes; Tocris Bioscience, Ellisville,

MO, USA). In a separate set of experiments, co-infusion of Sp-CAMPS with H-89 (12 µmol/L)

was performed.

3.3.6 PKA Activity Assay

PKA activity in duodenal samples taken directly after the clamp studies was measured

with the PepTag Assay Kit (Promega, Madison, WI) with minor modifications. 1 g of frozen

duodenal tissue was homogenized with a motor and pestle in ice-cold PKA extraction buffer

containing 25 mmol/L Tris-HCl (pH 7.4), 0.5 mmol/L EDTA & EGTA, 0.5 mmol/L ethylene

glycol-bis(β -aminoethylether)-N,N,N’,N’-tetraacetic acid, 10 mmol/L β-mercaptoethanol, and

3X Complete Mini EDTA-Free Protease Inhibitor Cocktail Tablet (Roche Diagnostics, Laval,

QC, Canada) and transferred to eppendorf tubes. The homogenates were centrifuged at 12,300

rpm for 5 minutes at 4°C. The supernatant was transferred to new eppendorf tubes and the

protein concentration was measured as described in the General Methods section 2.4. 5 µg of

protein was used for the reaction, which contained the PKA reaction 5X buffer, A1 peptide,

PKA activator 5X solution, Peptide protection solution, the sample and water. The positive

control contained all solutions described with the catalytic subunit of PKA provided by the kit

where the negative control contained no subunit or sample. All reagents excluding the sample or

catalytic subunit was first pipetted and incubated in a 30°C water for 1 min. Then the duodenal

lysates or PKA catalytic subunit (positive control) was added and the reaction was begun for 30

min at 37°C.The reaction was stopped by transferring the tubes to a 95°C heating block for 10

min. Then, 1 µl of 80% glycerol was added to the samples. The samples were then run on a

0.8% agarose gel (0.4 g of agarose dissolved in 50 mL of 50mM Tris-HCl (pH 8.0)) at 100 V for

15 minutes. The gel fluorescence was analyzed with a BioRad Molecular Imager Gel Doc XR+

Page 82: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

69

Imaging System (BioRad, Hercules, CA, USA). Data were analyzed using ImageJ (National

Institutes of Health software).

3.3.7 PCR methods

3.3.7.1 Tissue Preparation and RNA Extraction

An equal number of male SD rats were placed on a standard chow diet and a lard-oil

enriched high fat diet for 3 days ad libitum. Approximately 100 mg of fresh duodenal whole

tissue was collected and utilized. The tissues were homogenized using a mortar and pestle,

which were cooled by liquid nitrogen. Following homogenization, RNA was isolated using the

TRIzol method (Invitrogen-Life Technologies). 1.0 mL of TRIzol reagent was added to each

sample and vortexed to allow for the tissue homogenates to dissolve. Insoluble material from the

homogenate was then removed by centrifugation at 12,000 g for 10 minutes at 4ºC. The RNA-

containing supernatant was collected and allowed to incubate for 5 minutes at room temperature

to permit complete dissociation of the nucleoprotein complex. 0.2 mL of chloroform was added

to the samples, vortexed and incubated for 3 minutes at room temperature. The samples were

centrifuged at 12,000 g for 15 minutes at 4ºC. Following centrifugation, the mixture separated

into a lower red, phenol-chloroform phase, an interphase and a colourless upper aqueous phase.

The upper aqueous phase was collected as RNA remains exclusively in this phase. 0.5 mL of

isopropanol was added to the sample, vortexed and incubated at room temperature for 10

minutes. The samples were centrifuged at 12,000 g for 10 minutes at 4ºC to precipitate the RNA.

After removing the supernatant, 1mL of 75% ethanol was added to wash the RNA pellet. The

mixture was vortexed and centrifuged at 7,400 g for 5 minutes at 4ºC. The supernatant was

removed and the RNA pellet was allowed to dry for 10 minutes. The RNA pellet was

reconstituted in RNase-free water and incubated at 55ºC for 10 minutes. Measurement of the

optical density (OD) was performed to quantify RNA content at 260 and 280 nm using 2 µl of

Page 83: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

70

sample with a NanoDrop 1000 spectrophotomoter (Thermo Fisher Scientific, Mississauga, ON,

Canada). The ratio of 260/280 should be between 1.8 and 2 for RNA. RNA concentration

(µg/ml) was then calculated as:

RNA concentration =  OD260 x dilution factor x 40  

3.3.7.2 cDNA synthesis and PCR

First-strand cDNA was synthesized from 2 µg total RNA using the SuperScript™III

reverse transcriptase protocol (Invitrogen Life Technologies, Carlsbad, CA, USA). The

RNA/primer mixture was prepared in 0.5 mL tubes with the following: 2 µg of total RNA,

Oligo(dT)30 (50 µM), dNTP mix (10 mM), and DEP–treated water. The tubes were incubated at

65 °C for 5 min and immediately transferred to 55 °C. The cDNA synthesis mix was then made

as follows: DEPC-treated water, 10X RT buffer, 25 mM MgCl2, 0.1 M DTT, RNaseOUT™

Recombinase RNase Inhibitor, and SuperScript™III RT. The cDNA Synthesis mix was

prewarmed to 55 °C. The cDNA synthesis mix was added to each sample incubating at 55 °C

and incubated for 50 min total. The reaction was terminated at 85 °C and the tubes were chilled

on ice. After brief centrifugation and collection of the reaction, RNase H was added to each tube

and incubated at 37 °C for 20 min before proceeding to PCR. A PCR mix was prepared (totaling

50 µl) with the following reagents: Phusion High-Fidelity DNA polymerase (Thermo Scientific,

IL, USA), 5X buffer, dNTP (10 mM), primers (13µM), cDNA (aliquots of 5 µL and 1 µL of

cDNA product were used for PCR for CCK1 receptor and β-actin respectively), and water.

PCR amplification was performed with a S1000 Thermal Cycler (Biorad, Hercules, CA, USA)

with an initial cycle of 95 °C for 3 min, followed by 30 cycles each at 95 °C for 30 s

(denaturing), 56 °C for 30 s (annealing), and 72 °C for 40 s (extension). The final extension step

was executed at 72 °C for 7 min. The sequence of the primers for the CCK1 receptor were as

follows: 5′-TGAACTCGGACTGGAAAATGAGAC-3′ for the forward primer and

Page 84: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

71

5′-GCATAGCGTCACTTGGCAACAG-3′ for the reverse primer. The sequence of the primers

for β-actin were as follows: 5'-TGAGACCTTCAACACCCCAGCC-3' for the forward primer

and 5'-GAGTACTTGCGCTCAGGAGGAG-3' for the reverse primer. The expected

amplification product sizes for CCK1 receptor and β-actin were 563 bp and 642bp, respectively.

A negative control reaction that contained all the PCR components, except the target cDNA,

was included in each PCR assay. β-actin was used as a control for PCR efficiency. A 1%

agarose gel was prepared by combining 100 ml of 1 x TBE (Tris, boric acid, and EDTA), 1g of

agarose and ethidium bromide. The PCR final products were electrophoresed at 90V until

separation. A DNA ladder was included in the gel for determination of product size. Gels were

visualized under ultra violet light with a BioRad Molecular Imager Gel Doc XR+ Imaging

System (BioRad, Hercules, CA, USA). The band intensities for CCK1 receptor density was

quantified by densitometry with the Quantity One 1-D Analysis Software (BioRad, Hercules,

CA, USA) and normalized to those of the housekeeping gene, β-actin.

3.3.9 Biochemical Analysis

Please refer to the General Methods section 2.6.1-2.6.3 for details on biochemical

analyses. Plasma glucose concentrations were determined using a GM9 Analox Glucose

Analyzer (Analox Instruments, Lunenbertg, MA). Radioactivity of plasma glucose was

conducted as described. Plasma insulin levels were measured using a radioimmunoassay (Linco

Research, St Charles, MO).

3.3.10 Calculations and Statistical Analysis

Values are represented as the mean ± SEM. The basal conditions were averaged between

t = 60-90 minutes and the clamp conditions were averaged between t = 180-200 minutes. For the

electrophysiological ex vivo recordings of duodenal preparations, the mesenteric afferent nerve

activity was recorded as a mean discharge rate (impulses/sec). ANOVA was used to determine

Page 85: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

72

statistical differences between groups followed by a Tukey’s post hoc test. A probability of P <

0.05 was considered significant.

3.4 Results

3.4.1 Direct activation of PKA lowers glucose production

In order to first evaluate whether activation of PKA within the duodenum regulates

glucose production, we infused the cell-permeable PKA activator Sp-CAMPS directly into the

duodenal lumen of normal rodents during the pancreatic (basal insulin) euglycemic clamp

studies (Figure 3.1 A and B). During the pancreatic clamp studies, where plasma insulin was

maintained at basal levels (t=60-90 min) (Table 3.1) an intraduodenal Sp-CAMPS

administration increased the glucose infusion rate which was needed to maintain euglycemia

(Figure 3.2A). This was due secondarily to an inhibition in the rate of glucose production

(Figure 3.2B and C) while the rate of glucose uptake remained unchanged (Figure 3.2 D).

Importantly, a direct infusion of Sp-CAMPS into the jejunum failed to lower glucose production

(clamp glucose production: 12.2 +/- 2.0 mg/kg/min; n = 5), suggesting that duodenal Sp-

CAMPS administration activated duodenal PKA to lower glucose production. Next, co-infusion

of Sp-CAMPS with two independent cell-permeable PKA inhibitors H89 or Rp-CAMPS was

conducted (Figure 3.1A and B). Inhibition of PKA activation through co-infusion of

intraduodenal Sp-CAMPS with either H89 or Rp-CAMPS abolished the ability of duodenal Sp-

CAMPS to increase the glucose infusion rate (Figure 3.2A) and decrease glucose production

(Figure 3.2B and C). In order to confirm the specificity of these treatments to activate duodenal

PKA, from duodenal tissues taken immediately after the termination of the clamp studies we

assessed PKA activity. The A1 peptide is phosphorylated by PKA and thus a higher the ratio of

phospho(P)-A1/A1 reflects a higher degree of PKA activation. Intraduodenal Sp-CAMPS

induced duodenal PKA activity (Figure 3.2E) and this activation was fully blocked by co-

Page 86: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

73

infusion with Rp-CAMPS (Figure 3.2E). These data suggest that direct activation of duodenal

PKA is sufficient to lower glucose production.

3.4.2 Activation of PKA lowers glucose production via a vagal afferent firing

We next assessed whether duodenal PKA lowers glucose production through activation

of a neuronal network. We first evaluated, in an ex vivo duodenal preparation, the effect of an

intraluminal infusion of Sp-CAMPS on mesenteric neuronal discharge rate (Figure 3.3). In this

regard, a branch of the mesenteric nerves consisting of both vagal and spinal afferents was

dissected and recorded using a suction electrode466. An intraduodenal infusion of Sp-CAMPS

resulted in a gradual rise in the spontaneous discharge rate of the mesenteric nerve (Figure

3.4A) The peak discharge rate during intraduodenal Sp-CAMPS administration was 116.8 +

25.4 imp/s and this reflected a significant increase of 27 + 7 % (Figure 3.4B) compared with the

average basal discharge rate (92.0 + 20.8 imps/s). We next addressed whether this increase in

mesenteric neuronal discharge rate was due to activation of PKA through co-infusion of Sp-

CAMPS with the PKA inhibitor H89. An intraduodenal infusion of H89 alone did not have any

effect on the spontaneous firing rate but abolished the ability of Sp-CAMPS to increase

mesenteric neuronal discharge rate (Figure 3.4C). These results suggest that direct activation of

duodenal PKA increases the spontaneous firing rate of the duodenal mesenteric nerve.

We next investigated whether changes in vagal and/or spinal firing resulted in changes in

the mesenteric neuronal firing. This was through measuring distension-induced duodenal

afferent activity in response to duodenal PKA activation. In brief, ramp distension results in

biphasic increases in duodenal afferent nerve discharge of which the initial phase of the

response (Figure 3.4D; rectangle) is a rapid increase in afferent discharge at the beginning of

distension, reflecting an activation of low threshold mechanoreceptors. The second phase

(Figure 3.4D; arrow) is an accelerated increase in afferent activity when the intraluminal

Page 87: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

74

pressure reaches ~20 mmHg, reflecting an activation of high threshold mechanoreceptors. It is

generally thought that firing of vagal afferents is due to activation of low threshold

mechanoreceptors that encode innocuous (physiological) mechanical stimulation (i.e., gastric

emptying) whereas spinal afferents are mostly triggered by activation of high threshold

mechanoreceptors that encode noxious stimulation (i.e., over distension due to obstruction)467.

We report that intraduodenal administration of Sp-CAMPS inhibited the high threshold

mechanosensory responses (Figure 3.4E). Together with the overall increase in duodenal

mesenteric spontaneous afferent firing induced by duodenal PKA activation, our results strongly

suggest that an intraduodenal Sp-CAMPS administration activates duodenal spontaneous vagal

afferent firing but inhibits the spinal afferent.

We next delineated the functional relevance of the change in duodenal ex vivo vagal

afferent firing induced by duodenal PKA activation by assessing the neuronal network that is

required in glucose regulation induced by duodenal Sp-CAMPS. In this regard, during the clamp

studies we co-infused intraduodenal Sp-CAMPS with the local anesthetic tetracaine (Figure 3.5

A and B). Infusion of tetracaine alone into the duodenum did not affect the glucose kinetics

(Figure 3.6A-D) but abolished the ability of intraduodenal Sp-CAMPS to increase the glucose

infusion rate and lower glucose production independent of changes in plasma insulin and

glucose levels as well as glucose uptake during the clamps (Figure 3.6A-D;Table 3.2). Thus,

duodenal innervation of vagal afferent nerves is required for intraduodenal Sp-CAMPS to lower

glucose production.

3.4.3 Activation of NR1-containing NMDA receptors is required for duodenal PKA to

lower glucose production

The vagal afferent nerves that innervate the small intestine terminate at the level of NTS

within the DVC. To address the requirement of NMDA receptor activation in duodenal Sp-

Page 88: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

75

CAMPS induced suppression of glucose production, we next inhibited NMDA receptor-

mediated neuronal transmission in the DVC via direct NTS-targeted administration of the

NMDA receptor blocker MK-801 (Figure 3.7A and B). A MK-801 infusion into the DVC

alone did not affect glucose kinetics (Figure 3.8A-D) but negated the ability of duodenal Sp-

CAMPS to increase the glucose infusion rate and lower glucose production (Figure 3.8A-C).

This blockade effect of DVC MK-801 occurred independent of changes in the rate of glucose

uptake (Figure 3.8D) and plasma insulin levels (Table 3.2).

The NMDA receptor is composed of the NR1 and NR2 subunits465 and direct activation

of either the NR1 or NR2 subunit within the DVC is sufficient to lower glucose production382.

To address the role of the NR1 subunit in duodenal PKA induced suppression in glucose

production, we knocked down the NR1 subunit expression of the NMDA receptors in the DVC.

To this end, we injected an adenovirus expressing the shRNA of NR1 vs. mismatch (mm)

control into the DVC (Figure 3.7A) and the rats subsequently underwent duodenal infusion and

pancreatic clamp studies (Figure 3.7B). We have previously confirmed the specificity of the

NR1 knock-down within the DVC using the same adenoviral injection protocol382. An

intraduodenal Sp-CAMPS administration increased the glucose infusion rate and lowered

glucose production in DVC mm-injected rats (Figure 3.8A-C). The ability of duodenal Sp-

CAMPS infusion to alter glucose kinetics was negated in DVC shRNA-NR1-injected rats

(Figure 3.8A-C). Together with the pharmacological loss-of-function studies, these results

indicate that activation of the NR1-containing NMDA receptor within the DVC mediates the

vagal afferent neuronal signal(s) ignited by duodenal PKA activation to lower glucose

production.

Page 89: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

76

3.4.4 Duodenal PKA activation requires brain to liver communication to lower glucose

production

We next tested whether brain to liver communication is required for duodenal PKA

activation to lower glucose production, we repeated the intraduodenal Sp-CAMPS infusion

clamp studies in rats that received hepatic vagotomy, a surgical procedure which abolishes brain

to liver communication (Figure 3.7A). An intraduodenal Sp-CAMPS administration failed to

increase the glucose infusion rate and lower glucose production in hepatic vagotomized rats

(Figure 3.8A-C). Together with the duodenal ex vivo data, these in vivo results together indicate

that activation of duodenal PKA is sufficient to increase vagal afferent firing to trigger a gut-

brain-liver axis to lower glucose production.

3.4.5 CCK lowers glucose production via PKA activation

A direct duodenal CCK-8 administration activates CCK1 receptors to trigger a neuronal

network to lower glucose production, an effect that is abolished in rodents fed a high fat diet for

three days384. In order to locate the downstream potential defect(s) of CCK signaling in the

duodenum causing duodenal CCK resistance, we first addressed whether binding of CCK to its

CCK1 receptor results in PKA activation to lower glucose production. An intraduodenal

infusion of CCK-8 with either PKA inhibitor, H89 or Rp-CAMPS, was performed while plasma

insulin and glucose levels were maintained at basal levels during the clamps (Figure 3.9A and

B; Table 3.3). Consistent with previous findings384, intraduodenal CCK-8 increased the

glucose infusion rate required to maintain euglycemia due to an inhibition of glucose production

(Figure 3.10A-C) rather than changes in glucose uptake (Figure 3.10D). Co-infusion of CCK-8

with either H89 or Rp-CAMPS abolished the ability of duodenal CCK-8 to increase the glucose

infusion rate and lower glucose production (Figure 3.10A-C). Importantly, in duodenal tissues

taken immediately after the pancreatic clamp studies, duodenal CCK-8 administration activated

Page 90: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

77

PKA (Figure 3.10E), which was reversed by co-infusion of the PKA inhibitor Rp-CAMPS

(Figure 3.10E). To ensure activation of PKA lies downstream of the CCK1 receptor, we next

co-infused Sp-CAMPS with a CCK1 receptor antagonist MK-329 which did not abolish the

ability of Sp-CAMPS to lower glucose production (Figure 3.10B and C), indicating that PKA

activation lies downstream of CCK. Given these findings, we next tested whether direct

activation of PKA bypasses duodenal CCK resistance to lower glucose production in rats fed

with a high fat-diet (Figure 3.11A).

3.4.6 The CCK1 receptor fails to activate PKA after short term high fat feeding

Rats were fed with a lard-oil enriched diet (Table 2.1) for 3 days and rats that were

hyperphagic underwent the intraduodenal infusion clamp studies (Figure 3.11A). Intraduodenal

CCK-8 failed to increase duodenal PKA activity (Figure 3.12E) and the glucose infusion rate

(Figure 3.12A), and also failed to lower glucose production (Figure 3.12B and C) in high fat-

fed rats. Glucose uptake was comparable among groups (Figure 3.12D). The inability of

duodenal CCK-8 to regulate glucose homeostasis was independent of changes in duodenal

CCK1 receptor expression upon high fat feeding (Figure 3.13F), suggesting that CCK-

resistance lies within the signaling pathway(s). In this regard, direct activation of duodenal PKA

via Sp-CAMPS increased the glucose infusion rate (Figure 3.13A), lowered glucose production

(Figure 3.13B and C) and activated duodenal PKA (Figure 3.13E) in high fat-fed rats. Glucose

uptake was unaltered (Figure 3.13D). These results suggest that direct activation of duodenal

PKA bypasses CCK-resistance to lower glucose production and that duodenal CCK-resistance

likely arises from the inability of the CCK1 receptor-coupled signaling cascade to activate PKA

in high-fat fed rats.

Page 91: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

78

3.5 Discussion

This current study set out to elucidate the duodenal CCK/CCK1 receptor-signaling

cascade that regulates glucose production. We here demonstrated that direct activation of

duodenal PKA was sufficient to lower glucose production in vivo in parallel to an induction of

spontaneous vagal afferent firing in an ex vivo duodenum preparation. The in vivo functional

relevance of the increased vagal afferent firing was illustrated by the following findings when: i)

inhibition of neuronal innervation of the duodenum ii) DVC NR1-containing NMDA receptors

and iii) hepatic vagal transmission all negated the ability of duodenal PKA activation to lower

glucose production. Moreover, in normal rats, duodenal CCK/CCK1 receptor signaling requires

PKA activation to lower glucose production. Excitingly, direct activation of duodenal PKA

bypassed CCK resistance to lower glucose production in rodents fed a high fat diet. The

physiological relevance in glucose regulation of duodenal PKA signaling remains to be

clarified. However, these data collectively illustrate that duodenal PKA activation triggers vagal

afferent firing to activate NR1 containing NMDA receptors to lower glucose production in

normal and high fat fed rodents. Thus, administration of PKA agonists into the duodenum could

potentially help to restore glucose homeostasis in diabetes and obesity.

It has been previously demonstrated that vagal afferents innervating the small intestine

express the CCK1 receptor468. We are currently limited by technology to locate the exact site of

duodenal PKA activation. However our data strongly suggests that activation of the duodenal

CCK1 receptor G-protein coupled PKA signaling triggers vagal afferent firing to lower glucose

production. It remains to be clarified how PKA mediated signaling pathway(s) trigger vagal

afferent firing to inhibit glucose production. Given that the anesthetic tetracaine, a voltage gated

sodium channel inhibitor, abolished the ability of duodenal PKA activation to lower glucose

production, voltage gated sodium channels represent a potential downstream mediator.

Supporting this finding, PKA activation potentiates sodium current in the brain469 and argues for

Page 92: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

79

a role of duodenal voltage-gated sodium channels in mediating duodenal signal(s) to regulate

glucose production.

In addition to the findings described above, we also demonstrated that activation of

duodenal PKA inhibits mechanosensory spinal afferent firing. Although the underlying

mechanisms in the regulation of pain remain elusive, studies aimed to dissect the neuronal and

signaling mechanisms that control pain tend to focus on the control of spinal afferent firing as

changes in spinal afferent firing directly modulate pain470. For the first time, our study indicates,

that duodenal PKA signaling can regulate spinal afferent firing. Although this hypothesis

remains to be validated, an important potential implication arises in the context of the current

study, as direct delivery of a PKA agonist into the duodenum would lower glucose production in

the absence of pain induction.

We further discovered that direct activation of duodenal PKA can lower glucose

production in high fat diet fed rodents to bypass CCK resistance. This suggests that a duodenal

CCK-8 administration fails to activate the CCK-1 receptor and subsequent downstream

mediators, such as PKA, through an inability of G-protein coupled signaling to activate AC and

increase cAMP formation. This is consistent with the findings that high fat feeding reduces the

activity of AC and subsequent cAMP formation in the liver471. In addition to the PKA pathway,

the duodenal CCK/CCK1 receptor activates a PLC-dependent signaling pathway to regulate

pancreatic secretions461–464. The role of PLC in mediating the duodenal CCK effect on glucose

production remains to be clarified but our preliminary data suggests an involvement of duodenal

PLC signaling since an intraduodenal co-infusion of the PLC inhibitor U 73122 (200 µM) with

CCK-8 negated the ability of CCK-8 to inhibit glucose production. The rate of glucose

production during the clamp was 11.1 +/- 1.5 mg/kg/min (CCK-8 + U73122; n= 6) vs. 10.8 +/-

0.7 (U73122 alone; n=5) or 6.0 +/- 0.4 (CCK-8 alone; n=8). Finally, given that duodenal protein

kinase C-δ signaling is necessary for duodenal lipid sensing383 and sufficient to trigger CCK1

Page 93: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

80

receptors391 to regulate glucose production, we propose duodenal lipids activate a PKC-δ ->

CCK1 receptor -> cAMP-PKA dependent sequential signaling cascade to trigger vagal afferent

firing and inhibit glucose production in normal rats. Importantly, direct activation of

downstream signaling of CCK1 receptor (i.e., PKA), but not upstream (i.e., PKC-δ391), bypasses

duodenal CCK-resistance to inhibit glucose production in high-fat fed rats.

In summary, we have demonstrated, for the first time to our knowledge, that activation

of duodenal PKA ignites vagal afferent firing and triggers a neuronal network to lower glucose

production. In addition, activation of duodenal PKA is required for CCK to lower glucose

production in normal rats and bypasses CCK-resistance in high-fat fed rats to lower glucose

production. These data highlight a previously unappreciated role of duodenal PKA signaling in

neural regulation of glucose homeostasis.

As discussed in the introduction, CCK and leptin may interact to regulate feeding. Given

that CCK regulates glucose production, the purpose of Study 2 of this thesis was to address

whether intestinal leptin action, like CCK, also regulates glucose production through a neuronal

network.

Page 94: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

81

Figure 3.1 Schematic representation of working hypothesis – duodenal Sp-CAMPS activates PKA to lower glucose production, which is abolished upon co-infusion of Sp-CAMPS and H-89 or Rp-CAMPS, and experimental design.

A) Proposed model for duodenal PKA to lower glucose production. Infusion of Sp-CAMPS (PKA agonist) activates duodenal PKA and such activation is prevented upon co-infusion with either PKA inhibitor H-89 and Rp-CAMPS. B) Schematic representation of experimental design: on Day 1, intravenous and duodenal catheters were implanted in male SD rats (280-300g). Rats were given 4 to 5 days of recovery until the pancreatic clamp studies where duodenal infusions of saline, Sp-CAMPS ± H-89 or Rp-CAMPS were administered.

Page 95: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

82

Figure 3.2 Duodenal PKA activation lowers glucose production.

(A and B) During the pancreatic clamp (t = 180-200) an intraduodenal Sp-CAMPS infusion (30 µmol/L) increased the glucose infusion rate (A, *P < 0.05 vs other groups) and decreased glucose production (B, *P < 0.05 vs other groups). Co-infusion of Sp-CAMPS with H-89 or Sp-CAMPS abolished the effects of Sp-CAMPS on A) the glucose infusion rate and B) glucose production. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (*P < 0.05 vs other groups). D) The rate of glucose uptake remained unchanged amongst all groups. E) Duodenal PKA activity assessed with the PepTag assay. Duodenal Sp-CAMPS infusion significantly increased the amount of phosphorylated A1 peptide versus nonphosphorylated A1 peptide (*P < 0.01 vs other groups). SAL, n = 10; Sp-CAMPS n = 9; H-89 n = 5; Sp-CAMPS + H-89 n = 6; Rp-CAMPS n = 5; Sp-CAMPS + Rp-CAMPS n = 5. Values are shown ± SEM.

Page 96: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

83

Figure 3.3 Schematic representation of working hypothesis – duodenal PKA activation increases vagal afferent firing

Proposed model for duodenal PKA activation to increase vagal afferent firing. Infusion of Sp-CAMPS into the duodenum increases the spontaneous discharge rate of the mesenteric nerve which is inhibited upon co-administration with H-89.

Page 97: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

84

Figure 3.4 Direct activation of duodenal PKA increases the spontaneous discharge rate of the mesenteric nerve and inhibits spinal afferent firing of the duodenum.

A) An intraluminal infusion of Sp-CAMPS increases the spontaneous discharge rate of the duodenal mesenteric nerve. The top panel represents nerve activity and the bottom panel represents discharge frequency. B) An intraluminal infusion of Sp-CAMPS increased the spontaneous discharge rate of the mesenteric nerve, represented as normalized nerve activity (*P < 0.05 vs control). C) An intraluminal infusion of Sp-CAMPS with H-89 abolished the increase in afferent discharge. D) Distension-evoked biphasic activation of the duodenal afferent nerves ± Sp-CAMPS administration. Pressure increase and discharge frequency are shown in top and bottom panels, respectively. The rectangles indicate activation of low-threshold mechanoreceptors and the arrows indicate activation of high-threshold mechanoreceptors. Intraduodenal Sp-CAMPS administration inhibited high-threshold mechanosensory responses. E) Sp-CAMPS infusion inhibited the high-threshold mechanosensory responses, represented as a change in discharge rate in comparison to the basal discharge rate (**P < 0.05 vs control). N = 6 per group. Values are shown ± SEM.

Page 98: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

85

Figure 3.5 Schematic representation of working hypothesis – duodenal PKA activation triggers a neuronal network to lower glucose production and experimental design.

A) Proposed model for a neuronal network activated by duodenal PKA activation. The local anesthetic tetracaine abolishes neuronal innervation of the duodenum. B) Schematic representation of experimental design. Tetracaine was co-infused with Sp-CAMPS during the pancreatic clamp.

Page 99: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

86

Figure 3.6 Duodenal PKA activation lowers glucose production through a neuronal network.

(A and B) An intraduodenal infusion of Sp-CAMPS increased the glucose infusion rate (A, *P < 0.01 vs other groups) and decreased glucose production (B, *P < 0.01 vs other groups). This was abolished upon co-infusion with tetracaine. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (*P < 0.001 vs other groups). D) The rate of glucose uptake remained unchanged in all groups. SAL, n = 10; Sp-CAMPS n = 9; tetracaine, n = 5; Sp-CAMPS + tetracaine, n = 5. Values are shown ± SEM.

Page 100: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

87

Figure 3.7 Schematic representation of working hypothesis – duodenal PKA activation lowers glucose production through a gut-brain-liver neuronal axis and experimental design.

A) Proposed model for PKA induced activation of a gut-brain-liver axis. MK-801, a potent NMDA receptor antagonist, blocks activation of the receptors. A separate group of rats received a viral injection of an adenovirus expressing short hairpin RNA-NR1 versus a mismatch control to knockdown expression of the NR1 subunit of the NMDA receptor. Another group of rats received hepatic vagotomy surgery, which abolishes communication between the brain and liver. An intraduodenal Sp-CAMPS infusion failed to lower glucose production in the presence of a MK-801 infusion in the DVC, rats injected with shRNA NR1, or rats subjected to hepatic vagotomy. B) Experimental protocol. Stereotaxic surgeries were performed on male SD rats (~250-300g) 7 days prior to duodenal and vascular cannulations. A subgroup of rats received an adenovirus injection at the same time of the stereotaxic surgery. During the clamp studies, an intraduodenal Sp-CAMPS infusion was given ± MK-801 infusion.

Page 101: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

88

Figure 3.8 Duodenal PKA activation lowers glucose production through activation of the DVC NR1- containing NMDA receptor and hepatic innervation.

(A and B) An intraduodenal Sp-CAMPS infusion increased the glucose infusion rate (A, *P < 0.001, # P < .001 vs other groups) and decreased glucose production (B, *P < 0.001, #P < 0.001 vs other groups). Rats that received a DVC MK-801 administration, hepatic vagotomy surgery or injection of shRNA-NR1 failed to respond to a duodenal Sp-CAMPS infusion. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (*P < 0.001, # P < 0.01 vs other groups). D) The rate of glucose uptake remained unchanged in all groups. SAL, n = 10; Sp-CAMPS; DVC-MK-801 n = 5; Sp-CAMPS + DVC-MK-801 n = 5; DVC-shRNA NR1 n = 7; DVC-mistmatch n = 5; HVAG n = 6; Sp-CAMPS + HVAG n = 5. Values are shown as mean ± SEM. HVAG: Hepatic vagotomy

Page 102: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

89

Figure 3.9 Schematic representation of working hypothesis – Duodenal CCK requires PKA activation to lower glucose production and experimental design.

A) Proposed model for the necessity of PKA activation for duodenal CCK to lower glucose production. H-89 and Rp-CAMPS are PKA inhibitors, and MK-329 is a CCK1 receptor inhibitor. Intraduodenal CCK fails to lower glucose production upon co-infusion with either PKA inhibitor H-89 or Rp-CAMPS. PKA activation lies downstream of the CCK1 receptor as a Sp-CAMPS infusion lowers glucose production in the presence of the CCK1 receptor MK-329. B) Experimental protocol. 4 to 5 days after duodenal and vascular cannulation, the clamp studies were conducted where CCK-8 ± H-89 or Rp-CAMPS and Sp-CAMPS ± MK-329 were given.

Page 103: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

90

Figure 3.10 Duodenal CCK requires PKA activation to lower glucose production.

(A and B) An intraduodenal CCK-8 infusion increased the glucose infusion rate (A, *P < 0.01 versus other groups) and decreased glucose production (B, *P < 0.05 versus other groups). In contrast, coinfusion with either H-89 or Rp-CAMPS abolished the effects of CCK-8. A Sp-CAMPS infusion increased the glucose infusion rate (A, *P < 0.01 versus other groups) and decreased glucose production (B, *P < 0.05 versus other groups) in the presence of MK-329. C) Suppression of glucose production during the clamp period expressed as the percentage decrease from basal (*P < 0.01 versus all groups). E) A duodenal CCK-8 infusion significantly increased the amount of phosphorylated A1 peptide versus nonphosphorylated A1 peptide (*P < 0.05 versus all groups). SAL n = 10; CCK-8 n = 8; H-89 n = 5; CCK-8 + H-89 n = ; Rp-CAMPS n = 5; CCK-8 + Rp-CAMPS n = 5. Values are shown as mean ±SEM.

Page 104: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

91

Figure 3.11 Schematic representation of working hypothesis – Duodenal CCK fails to suppress glucose production upon high fat feeding, which is rescued upon PKA activation and experimental design

A) Proposed model to determine whether duodenal CCK-8 can suppress glucose production in response to high fat feeding for 3 days. Rats were placed on a lard-oil enriched high fat diet for 3 days and then the clamp studies were performed. Duodenal CCK-resistance is bypassed upon PKA activation. B) Experimental protocol. Rats were placed on regular chow for 4 days and then switched to a lard-oil enriched high fat diet for 3 days until the pancreatic clamp study where an intraduodenal infusion of CCK-8 or Sp-CAMPS was given.

Page 105: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

92

Figure 3.12 Duodenal CCK fails to activate duodenal PKA and lower glucose production after three days of high fat feeding.

A) Intraduodenal CCK-8 increased the glucose infusion rate (A, * P < 0.05, versus other groups) and decreased glucose production (B, *P < 0.05 versus other groups) in regular chow fed rats. After high fat feeding for 3 days, rats failed to respond to intraduodenal CCK-8. C) Suppression of glucose production during the clamp period expressed as a percentage decrease from basal (C, *P < 0.01 versus other groups). D) Glucose uptake remained unchanged among the groups. E) PKA activation in tissues taken after the clamp studies. Intraduodenal infusion of CCK-8 in HFD rats failed to increase PKA activity. SAL RC n = 10; SAL HFD n = 5; CCK-8 RC n = 8; CCK-8 HFD n = 6. Values are shown as mean ± SEM

Page 106: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

93

Figure 3.13 Duodenal Sp-CAMPS activates duodenal PKA activity and lowers glucose production in high fat diet fed rats.

(A and B) Intraduodenal Sp-CAMPS increased the glucose infusion rate (A, *P < 0.05 versus other groups) and decreased glucose production (B, *P < 0.05 versus other groups) in rats fed with RC. The glucose infusion rate was increased (A, #P < 0.01 versus other groups) and decreased glucose production (a, #p < 0.01 vs. other groups) in rats fed HFD. C) Suppression of glucose production during the clamp period expressed as a percentage decrease from basal (C, *P < 0.05 versus other groups, #P < 0.05). E) Duodenal Sp-CAMPS infusion in rats fed a HFD significantly increased the amount of phosphorylated A1 peptide versus nonphosphorylated A1 peptide (E, *p < 0.01 versus HFD SAL). F) CCK1 receptor expression was comparable in both regular chow and high fat fed rats. SAL RC n = 10; SAL HFD n = 5; Sp-CAMPS RC n = 9; Sp-CAMPS HFD n = 9. Values are shown as mean ± SEM.

Page 107: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

94

Table 3.1 Plasma insulin and glucose concentrations of the groups receiving an intraduodenal infusion during basal and clamp conditions.

Values are expressed as means ± SEM. (Basal: 60-90 min; Clamp: 180-200 min).

Page 108: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

95

Table 3.2. Plasma insulin and glucose concentrations of the groups receiving both an intraduodenal infusion and DVC infusion during basal and clamp conditions

Values are expressed as means ± SEM. (Basal: 60-90 min; Clamp: 180-200 min). HVAG, hepatic vagotomy.

Page 109: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

96

Table 3.3 Plasma insulin and glucose concentrations of the groups receiving an intraduodenal infusion during basal and clamp conditions.

Values are expressed as means ± SEM. (Basal: 60-90 min; Clamp: 180-200 min).

Page 110: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

97

Chapter 4Study 2

Jejunal Leptin-PI3K signaling lowers glucose production Modified From: Rasmussen, BA*, Breen, DM*, Duca, FA, Côté, CD, Zadeh Tahmasebi, M, Filippi, BM, and Lam, TK. (2014) Jejunal leptin-PI3K signaling lowers glucose production. Cell Metabolism 19, 1-7 *Equal contribution Permission to reproduce portions of the above manuscript has been obtained from the copyright owner: Elsevier Limited

Page 111: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

98

4.1 Abstract

Background and Aims: The fat derived hormone leptin binds to its hypothalamic receptors to

regulate glucose homeostasis. Leptin is also synthesized in the stomach and binds to its

receptors expressed in the intestine. Given that recent studies report jejunal nutrient sensing is

necessary for DJB to lower glucose production and plasma glucose levels in uncontrolled

diabetic rodents with insulin-deficiency, we sought to determine whether intestinal leptin

regulates glucose production through similar mechanisms as the brain in normal, and disease

models and whether jejunal leptin action mediates the glucose-lowering effect induced by DJB

in insulin-deficient uncontrolled diabetes. Methods: In rats and mice, we administered leptin

into the jejunum for 50 min and evaluated changes in glucose production during pancreatic

clamps in vivo. Molecular and chemical loss-of-function approaches targeting intestinal leptin

receptor-mediated signaling were utilized to assess the underlying mechanisms involved in

normal and diabetic (with or without DJB) rodents. Results: Intrajejunal leptin infusion

activated jejunal PI3K and STAT3 and lowered glucose production in normal rats and mice

independent of changes in circulating leptin and insulin levels. The glucose production-lowering

effect induced by jejunal leptin was negated in leptin receptor deficient fak/fak rats and db/db

mice, or upon co-infusion with a leptin receptor antagonist. Interestingly, blockade of jejunal

PI3K and not STAT-3 signaling negated jejunal leptin to lower glucose production in normal

rats, while the metabolic effect of leptin was also seen in insulin deficient STZ-induced

uncontrolled diabetic (independent of changes in glucagon levels) and insulin resistant HFD

rodents. Lastly, blockade of jejunal leptin action disrupted glucose homeostasis during refeeding

in uncontrolled diabetic rodents that received DJB. Conclusions: These data unveil a novel

Page 112: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

99

glucoregulatory site of leptin action and suggest that enhancing leptin-PI3K signaling in the

jejunum lowers plasma glucose concentrations in normal and diabetic conditions.

4.2 Introduction

Since the discovery of leptin125, there has been a large effort by scientists to evaluate the

physiological impact of hypothalamic leptin signaling9. Indeed, the brain plays an important role

in mediating the action of leptin by binding to the hypothalamic Leprb and activating

downstream signaling molecules, STAT3 and/or PI3K to regulate energy balance9,472. In

addition to regulating energy balance, activation of hypothalamic PI3K and STAT3 via a central

leptin infusion improves insulin sensitivity473 and lowers glucose production474 in high-fat fed

rodents. Central leptin also lowers plasma glucose levels in non-obese STZ-induced insulin-

deficient uncontrolled diabetic rodents by inhibiting glucose production in association with a

drop in plasma glucagon levels475,476. Thus it is evident that since its discovery, many questions

in regards to hypothalamic leptin action have been uncovered, but much work still remains to

uncover the neurocircuitry and metabolic impact of hypothalamic leptin action. Moreover,

whether leptin action regulates metabolism in extra-hypothalamic sites remains in question, but

studies are beginning to uncover such sites as hindbrain leptin signaling lowers food intake477.

In addition to adipocytes, it is believed that leptin is also produced by gastric chief

cells127,144 and acts on the Leprb expressed in the intestine and/or on vagal afferents that

innervate the intestine149–151 to regulate various intestinal processes. For example, gastric leptin

is secreted in response to nutrient ingestion478 and has been shown to work in collaboration with

other gut peptides to modulate feeding479. In addition to food intake regulation, leptin also helps

to maintain the intestinal environment as mice480 and humans481 with mutations in the leptin

receptor have higher susceptibility to intestinal infection. Furthermore, leptin regulates intestinal

lipid482 and carbohydrate483 absorption and increases neuronal activity of the NTS484. Given that

Page 113: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

100

STAT3 and PI3K are expressed in the intestine and/or on the vagal terminals that innervate the

intestine151,157,485, and that intestinal leptin receptor signaling regulates various intestinal

functions, intestinal leptin receptor signaling may also regulate glucose homeostasis. However,

such a hypothesis has not yet been tested in vivo.

Given that hypothalamic leptin triggers a neurocircuitry to control glucose

homeostasis25,486 while in parallel nutrient sensing in the small intestine triggers a gut-brain axis

to regulate glucose homeostasis402,487 we hypothesize that intestinal leptin activates a leptin

receptor-PI3K and/or STAT3-dependent pathway to regulate glucose homeostasis through a

neuronal network. In addition, jejunal nutrient sensing mechanisms are required for DJB surgery

to lower plasma glucose levels and glucose production in non-obese uncontrolled diabetic

rodents with insulin deficiency402. Given that hypothalamic brain leptin action has a similar

effect in uncontrolled diabetes475,476 we then evaluated whether the glucoregulatory control of

intestinal leptin action is intact in uncontrolled diabetic or high-fat fed rodents and necessary for

the rapid anti-diabetic effect of DJB.

4.3 Materials and Methods

4.3.1 Animal Preparation

Normal SD rats (280-300g) were maintained as described in General Methods section

2.1. 18 week old (~25-30g) C57BL/6J mice were obtained from Jackson laboratories (Bar

Harbor, Maine, USA). Mice were housed in groups of four and maintained on a standard 12-12h

light dark cycle, and had ad libitum access to water and rodent chow (Harlan Teklad 6%

mouse/Rat diet; 49% carbohydrate, 33% protein and 18% fat; total calories provided by

digestible nutrients: 3.1 kcal/g). Mice were given at least 5 days to acclimatize upon arrival

before surgeries were performed.

Page 114: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

101

4.3.1.1 Koletsky Rat

The Koletsky rat (fak/fak) was used as a model deficient of the long form leptin

receptors. The obese phenotype results from a spontaneous autosomal recessive nonsense

mutation on chromosome 5 producing a mutated leptin receptor488,489. Both lean (fa/fa) and

obese Koletsky rats (fak/fak) were obtained from Charles River at the age of 8 weeks. Koletsky

(fak/fak) rats weigh nearly the same as their lean littermates until ~4-6 weeks of age (~250-300g)

and then become hyperphagic and rapidly gain weight and become obese. Young adult (+6

weeks of age) Koletsky (fak/fak) rats are also hypertensive, hyperinsulinemic, hyperlipidemic,

and display only a marginal elevation in post-prandial glucose levels (6.2 vs. 5.2 mM) but with

normal fasting glucose levels, indicating only mild glucose intolerance490,491. Lean and obese

Koletsky rats were monitored at 4:00pm daily for body weight and food intake. Due to the fact

that obese Koletsky rats are hyperphagic, food intake was restricted to just below the average

amount of food consumed by the lean Koletsky rats as described473 in order to maintain a body

weight of ~300 g that was comparable to the lean control and male Sprague-Dawley rats. After

5 weeks, intravenous and jejunal cannulation surgeries were performed.

4.3.1.2 db/db mouse

The obese leptin receptor deficient male db/db mice from Jackson Laboratories were

used as an additional model of long form leptin receptor deficiency. Diabetes that results in

these mice arises from a recessive, autosomal single-gene mutation on chromosome 4, with

complete penetrance492–494. These mice become obese around 3-4 weeks, with elevations of

blood glucose levels at 4 to 8 weeks. Thus, affected mice are polydipsic, polyuric and

hyperphagic495. db/db mice were obtained at ~ 6-7 weeks of age. In order to age and weight

match the lean 18 week old C57BL/6J control mice (25-30g) to the obese db/db mice, the obese

db/db mice were monitored for body weight and food intake daily until achieving a body weight

Page 115: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

102

of ~30 g (~ extra 12 weeks). Since obese db/db mice are hyperphagic, their food intake was

restricted daily in order to maintain body weight near ~30 g. At 18 weeks of age, jugular vein

and jejunal cannulation surgeries were performed.

4.3.1.3 High fat diet feeding

A subgroup of rats were fed a lard oil enriched high fat diet for three days. Rats that

were hyperphagic underwent the clamp studies. Please refer to General Methods 2.1.1 for details

on high fat feeding.

4.3.1.4 Streptozotocin induced uncontrolled diabetes

A subgroup of SD rats were injected with 65 mg/kg STZ (Sigma-Aldrich, St. Louis,

MO, USA). STZ is a diabetogenic agent that is cytotoxic to pancreatic β cells and is used to

induce uncontrolled diabetes in rodents496. Through the glucose transporter 2 (GLUT2)497 the

deoxyglucose moiety of STZ enters the pancreatic β cells and its cytotoxic effect are through its

nitrosurea moiety. STZ was first weighed out and transferred to a light protective conical tube.

Before injection, the powder was dissolved in 0.9% saline and immediately injected i.p. to

induce diabetes in rats. STZ was administered 5–6 d before sham or DJB surgery or 4 days

before jejunal and vascular surgeries. Injected rats had ad libitum access to food and water. The

rats were monitored daily for blood glucose levels with a glucometer (Contour Blood Glucose

Meter, Bayer Inc., Toronto, ON, Canada) to ensure they were hyperglycemic. We included only

rats that were hyperglycemic (plasma glucose levels > 300 mg/dl) in the study.

Page 116: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

103

4.3.2 Animal Surgeries

4.3.2.1 Intestinal and vascular cannulation

4.3.2.1.1 Rats

3-4 days before the clamp studies, SD and Koletsky rats were anesthetized and a jejunal

catheter was inserted 8-10 cm from the Ligament of Treitz. A subgroup of rats underwent

duodenal catheter placement (0.5 cm proximal to the pyloric sphincter.). After the intestinal

cannulation, the jugular vein and carotid artery were cannulated. Refer to General Methods

Section 2.2.1 and 2.2.2 for details regarding these surgical procedures in rats.

4.3.2.1.2 Mice

The surgical procedures conducted in mice were similar to that described for rats in

General Methods Section 2.2. 2-3 days before the clamp studies, C57BL/6 mice or db/db mice

were anesthetized with an i.p. cocktail of (60-90 mg/kg) ketamine (Ketalean; Bimeda-MTC,

Cambridge, Ontario) and (8-10mg/kg) Xylazine (Rompun; Bayer). Exposure of the

gastrointestinal tract within the peritoneum was conducted through a laparotomy incision made

on the ventral midline as well as the abdominal muscle wall. After identifying the pyloric

sphincter, the jejunum was identified to be 4-6 cm from the Ligament of Treitz. With a 25-gauge

needle, a small hole was made on the ventral aspect of the jejunum (in a region with the least

vascularization to minimize bleeding) to allow insertion of an intestinal catheter made of

polyethylene tubing (PE 10, Clay Adams, Boston, MA) with a 0.1 cm extension of smaller

silicone tubing (0.012 in ID, 0.025 in. OD; Sil-Tec, Technical Products, USA). To ensure the

cannula was placed in the lumen of the jejunum, the cannula was flushed with saline. It was then

anchored to the outer serosal surface of the jejunum with 3M adhesives (Vetbond) and a 0.2 cm2

and piece of Marlex mesh sewn to surface with a 6-0 silk suture. Through the laparotomic

incision, the proximal portion of the catheter excited the abdominal cavity and the abdominal

Page 117: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

104

wall was closed with a 6-0 silk suture. At the back of the neck, a 1 cm midline incision was

made in the skin, rostral to the interscapular area, and the cannula was tunneled subcutaneously

to exit the incision. This 1 cm incision was sewn closed with 6-0 silk sutures and the proximal

portion of the cannula was closed with a knot until the day of the experiment. For jugular vein

cannulation, an indwelling catheter made with polyethylene tubing (PE 10, Clay Adams,

Boston, MA) with a cuff extension (10 mm, internal diameter of 0.012 inches) of Silastic tubing

(Dow Corning, Midland. MI) was inserted for infusion purposes. Briefly, after blunt dissection

through the muscle layer, the jugular vein was teased out and two 7-0 silk sutures were used to

prevent blood flow. After a small incision into the vessel wall, the catheter was inserted. After

insertion, the catheters were tunneled subcutaneously and filled with a 0.2% heparin mixture to

maintain patency of the cannula, which was tunneled with an 18 G needle. The cannula was

closed through knotting until the day of the procedure.

4.3.2.2 Duodenal-jejunal bypass surgery

DJB surgery was conducted in Study 2 as previously described in STZ (65 mg/kg)

injected rats402,440. The rats were fasted the night before the surgery to ensure no food remained

in the digestive tract. A midline laparotomy incision was made on the ventral flank of the rat to

expose the gastrointestinal tract. After locating the stomach and proximal duodenum, blood

vessels innervating both areas were sutured with 4-0 silk sutures to ensure no bleeding occurred

during the surgery. The stomach was clamped proximal to the pyloric sphincter with a metal

curved clamp and blunted small end scissors were used to separate the duodenum containing the

gastric sphincter from the stomach. The gastric sphincter duodenal stump was closed with 6-0

silk sutures using the purse-suture technique. 15 cm from the pyloric sphincter, a transection

was made to separate the distal duodenum/most proximal portion of the jejunum from the

remaining jejunum and digestive system. The distal section of the jejunum was then

Page 118: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

105

anastomosed to the stomach (lumen to lumen) with a 6-0 suture. 12 cm distal from the new

jejunum/stomach connection, a small 1 cm hole was cut using blunted small scissors where the

distal duodenum/most proximal jejunum section was anastomosed. The gastrointestinal tract

was returned to the abdomen and wet with saline. The sham surgery consists of the same

procedure except that all the transactions and cuts were sutured back together. The rodents were

monitored daily after surgery for recovery via food intake and body weight. Blood glucose

concentrations were also monitored each day after surgery in both sham and DJB rats (please

see the General Methods section 2.6.1 for plasma glucose measurements).

4.3.3 Intraintestinal infusions and treatments

The following substances were infused through a jejunal or duodenal catheter as described

during the pancreatic clamp from t = 150-200 at a rate of 0.01 ml/min in rats or from t = 120-

170 at 2 µl/min in mice:

(1) saline

(2) leptin (6.7 ng/min; R & D systems, Minneapolis, MN, USA)

(3) soluble leptin receptor (SLR) (binds to leptin to prevent binding to the Leprb 1 µg/min; R

& D systems, Minneapolis, MN, USA)

(4) STAT3 PI (STAT3 peptide inhibitor; 15 pmol/min; Calbiochem, Millipore, Billerica,

MA, USA)

(5) wortmannin (PI3K antagonist; 0.002 nmol/min, Sigma-Aldrich, St. Louis, MO, USA)

(6) LY294002 (PI3K antagonist; 0.2 nmol/min Sigma-Aldrich, St. Louis, MO, USA)

(7) tetracaine (local anesthetic; 0.01 mg/min Sigma-Aldrich, St. Louis, MO, USA)

Solution #2-4 was dissolved in saline while solution #5-7 was dissolved in 5% dimethyl

sulfoxide (DMSO). In mice, saline or leptin (6.7 ng/min) was infused intrajejunally. The dose

chosen for leptin (6.7 ng/min) for the intraintestinal infusions was selected based on the gastric

Page 119: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

106

emptying rate of leptin in rats143 and also based on the amount of leptin that was administered

centrally in rats that lowered glucose production474. In a separate set of experiments in both rats

and mice, intravenous leptin was infused at the same rate (0.01 ml/min for rats; 2 µl/min for

mice), duration (50 min) and dose (6.7 ng/min) as performed for the intrajejunal leptin infusions

in conjunction with receiving intrajejunal saline infusion (0.01 ml/min for rats; 2 ul/min for

mice).

4.3.4 Pancreatic (Basal Insulin) Euglycemic Clamp Technique

4.3.4.1 Pancreatic (Basal Insulin) Euglycemic Clamp Technique in Rats

Please refer to the General Methods section 2.3 for a detailed description of the clamp

procedure. After an overnight food restriction, rats received a primed-continuous constant

infusion of [3–3H] glucose, which was given throughout the experiment (t = 200) to reach

steady state. The pancreatic clamp was then initiated at t = 90 where insulin (1.2 mU/kg/min)

and somatostatin (3 µg/kg/min) were infused at a constant rate. Blood samples were taken to

determine if a variable 25% glucose infusion was needed to maintain euglycemia. At t = 150, a

jejunal infusion (please refer to 4.3.3) at 0.01 ml/min was conducted and maintained until the

end of the experiment (t = 200). In a subgroup a rats, a duodenal infusion of leptin was

performed at 0.01 ml/min. In a separate set of experiments, in addition to a jejunal saline

infusion, an intravenous leptin infusion was performed from t = 150 to t = 200 at the same equal

dose and duration as the intrajejunal leptin infusion.

4.3.4.2 Pancreatic (Basal Insulin) Euglycemic Clamp Technique in Mice

After an overnight food restriction, a primed-continuous intravenous infusion of [3–3H]-

glucose (1 µCi bolus, 0.1 µCi/min; Perkin Elmer) was initiated at the beginning of the

experiment (t = 0 min) and maintained until completion of the study (t = 170) to assess glucose

kinetics under steady state conditions using the tracer dilution methodology. A pancreatic (basal

Page 120: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

107

insulin)-euglycemic clamp was started through a constant infusion of insulin (1.4 mU/kg/min)

and somatostatin (8.3 µg/kg/min) from t = 60 until t = 170. Every 10 minutes, via tail sampling,

blood glucose readings were conducted (please see the General Methods section 2.5.1 for

plasma glucose measurements) and a variable infusion of 10% glucose solution was started and

periodically adjusted (every 10 min from t = 60–170 min) to maintain plasma glucose levels

similar to the basal state (t = 50 and 60 min). Jejunal infusions (2 µl/min) were initiated at 120

min and continued for the remaining 50 min until t = 170. In a separate set of experiments, in

addition to a jejunal saline infusion, an intravenous leptin infusion was conducted at equal dose

and duration jejunal leptin infusion from t = 120 to t = 170 min. Plasma samples for the

determination of [3–3H] glucose specific activity (please see the General Methods section 2.5.2)

were obtained at 10–min intervals during the basal period (50 and 60 min) and at the end of the

jejunal infusion period (150-170 min). At the end of the experiment, mice were anesthetized and

tissue samples were removed and immediately immersed in liquid nitrogen. All tissue samples

were stored at –80 ºC until use.

4.3.5 Rat [3–3H] glucose infusion protocol (non-clamped conditions)

These studies were performed in a group of STZ-injected rats 9-10 days after the STZ

injection (65 mg/kg) and 4 days after jejunal and vascular cannulation surgeries. Only if the rats

were hyperglycemic (plasma glucose concentrations > 300 mg/dl) were they then included in

the subsequent infusion studies. Rats were restricted to ~56 kcal the night before the experiment.

The total experimental time for the in vivo infusion experiments was 140 minutes. At t = 0, a

primed-continuous infusion of [3–3H] glucose (40 µCi bolus; 0.4 µCi/min) was initiated and

maintained until the end of the experiment (t = 140 min) to assess glucose kinetics under steady

state conditions using the tracer dilution methodology. At t = 90 min, an intrajejunal infusion of

saline or leptin was initiated and continued for the remaining 50 min. In a separate set of

Page 121: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

108

experiments, intravenous leptin was infused at the same dose and duration as intrajejunal leptin

infusion and was initiated at 90 min and continued for the remaining 50 min of the experiment.

Plasma samples for the determination of [3–3H] glucose specific activity (please see the General

Methods section 2.5.2) were obtained at 10–min intervals during the basal period (60-90 min)

and at the end of the jejunal infusion period (130 and 140 min). At the end of the experiments,

rats were anesthetized and tissue samples were removed and immediately immersed in liquid

nitrogen. All tissue samples were stored at –80 ºC until use.

4.3.6 Fasting and refeeding protocol

Fasting and refeeding experiments were conducted in the STZ-injected rats that received

either SHAM or DJB surgery in conjunction with jejunal catheter placement. The experiment

took place 2 days after the surgery. The night before the experiment (5:00pm), the rats were

fasted for 24 hours. At 4:50pm (t = -10), the day of the experiment, baseline glucose

measurements (please see General Methods section 2.5.1 for details regarding plasma glucose

readings) were taken via tail sampling. Then, a continuous intrajejunal infusion (Harvard

Apparatus PHD 2000 infusion pumps) of either (i) saline or (ii) SLR (1 µg/min; the same dose

as given in the clamp studies) was initiated and lasted throughout the course of the experiment

until t = 50 to match the treatment during the clamp studies. At t = 0, rats were allowed to

consume a regular chow diet ad libitum. Blood glucose levels and food intake were measured

throughout the course of the experiment in 10-minute intervals until completion (t = 50).

4.3.7 Gut tissue collection and preparation for western blotting and enzymatic activity

assay

Separation of the jejunal or duodenal mucosal layer (~100 mg) from the jejunal or

duodenal smooth muscle layer (~150 mg) was conducted immediately after removal from

anesthetized animals at the termination of the clamp studies. The separation was done in a petri

Page 122: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

109

dish filled with 0.9% saline on ice with a spatula. The separated layers were transferred to

separate eppendorf tubes and stored at -80°C until use. The tissues were transferred to ice the

day of the western blot or enzymatic activity assay and lysed on ice with a handheld blender in

6.3 µl per 1mg of tissue of a buffer containing: 50 mM Tris–HCl (pH 7.5), 1 mM EGTA, 1 mM

EDTA, 1 % (w/v) Nonidet P40, 1 mM sodium orthovanadate, 50 mM sodium fluoride, 5 mM

sodium pyrophosphate, 0.27 M sucrose, 1 µM Dithiotritolo (DTT) and protease inhibitor

cocktail (Roche Diagnostics, Laval, QC, Canada). After homogenization, the tissues were spun

at 12,000 rpm for 15 minutes at 4ºC. The supernatant was transferred to new eppendorf tubes

and the protein concentration of each homogenized tissue was determined with the Pierce 660

nm protein assay as described in the General Methods section 2.4.

4.3.8 Western blotting

Intestinal tissues were removed, separated into the mucosal and smooth muscle layer,

homogenized and processed as described in section 4.3.6 above. 100 µg of protein was thawed,

vortexed and then subjected to sodium dodecyl sulfate- polyacrylamide gel electrophoresis on

an 8% polyacrylamide gel for 90 minutes at 100V. After electrophoresis separation, in transfer

buffer the protein was transferred to nitrocellulose membranes. The membranes were incubated

for 1 hour at room temperature with Tris buffered saline-Tween (TBS-T) containing 5% (w/v)

BSA. The membranes were then immunoblotted in the same buffer for 16 hours at 4 °C with the

indicated primary antibodies (diluted to 1:1000 for pSTAT3 and total STAT3; Cell Signaling

Technology, Danvers, MA, USA). The blots were then washed 5 times with TBS-T for 30

minutes at room temperature to remove the primary antibody and incubated with secondary

horseradish peroxidase (HRP)-conjugated rabbit IgG antibody (Cell Signaling Technology,

Danvers, MA, USA) for P-STAT3 and total STAT3 (diluted 1:4000) in 5% skim milk for 1 hour

at room temperature. After repeating the washing steps, the signal was detected with the

Page 123: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

110

enhanced chemiluminescence reagent (Thermo Scientific, IL, USA). Immunoblots were

exposed to x-ray film and developed using a film automatic processor (SRX-101; Konica

Minolta Medical), and films were scanned with the GS-800 Calibrated Densitometer (BioRad,

Hercules, CA, USA). The phosphorylation level of STAT3 was quantified by densitometry with

the Quantity One 1-D Analysis Software (BioRad, Hercules, CA, USA) and normalized for the

corresponding total protein level. As the ability of leptin to stimulate STAT3 phosphorylation

was found to be the same in both the mucosal and smooth muscle layer of jejunum and

duodenum, we have simply presented leptin-induced STAT3 phosphorylation in the jejunal and

duodenal mucosa.

4.3.9 RNA extraction, reverse transcription and PCR methods

4.3.9.1 RNA Extraction

The total RNA from rat duodenal and jejunal mucosa was extracted by using the

PureLink RNA Mini Kit from Ambion. First, a RNase free work area was ensured. Of note,

RNase free tubes and pipette tips were used throughout the extraction. Briefly, ~70 mg was

homogenized in lysis buffer (provided with the kit) containing 1% 2-mercaptoethanol using the

rotor-stator homogenizer. The lysate was then centrifuged at 12,000 X g for 2 minutes.

Following centrifugation, one volume of 70% ethanol was added to the tissue homogenate and

mixed. 700 µl of the sample was transferred to the spin cartridge and centrifuged at 12,000 x g

for 15 seconds at room temperature. The flow-through was then discarded and this process was

repeated 3 times. 700 µl of wash Buffer I followed by 500 µl of wash buffer II was added

individually and the same process was conducted as described for the 70% ethanol. The

membrane with the attached RNA was allowed to dry for 1-2 minutes. Recovery was conducted

through the addition of RNase-Free water to the spin cartridge and allowed to incubate at room

temperature for 1 minute. After centrifugation, the purified RNA was stored at –80 ºC until use.

Page 124: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

111

Measurement of the optical density (OD) was performed to quantify RNA content at 260 and

280 nm using 2 µl of sample with a NanoDrop 1000 spectrophotometer (Thermo Fisher

Scientific, Mississauga, ON, Canada). The ratio of 260/280 should be between 1.8 and 2 for

RNA. RNA concentration (µg/ml) was then calculated as:

RNA concentration  =  OD260 x dilution factor x 40

4.3.9.2 Reverse transcription and polymerase chain reaction

The reverse transcription and subsequent PCR were preformed with the QIAGEN

OneStep RT-PCR kit that allowed performing both the reactions in a single PCR programme.

After thawing of all reactants, a master mix was made on ice containing the Omniscript and

Sensiscript Reverse Transcriptase and the HotStarTaq DNA Polymerase (provided by the kit), a

dNTP mix with a final concentration of 400 µM each, the Q-reagent (provided by the kit) and

the primers (0.6 µM each). A total amount of 1µg of RNA was incubated in this reaction

mixture. In controls, reverse transcriptase was omitted. The PCR reaction was performed in a 50

µl volume using a S1000 Thermal Cycler (Biorad, Hercules, CA, USA) as follow: 30’ at 50°C

(reverse transcription), 15’ at 95°C (initial PCR activation), three step cycling (40 cycles) of 45’

at 94°C, 1’ at 50°C and 1’ at 72°C and a final extension for 10’ at 72°C. The sequence of the

forward primer used was 5’- ATGAAGTGGCTTAGAATCCCTTCG-3’ and that of the reverse

was 5’-ATATCACTGATTCTGCATGCT-3’ (ACGT Corporation, Toronto, ON, Canada) as

previously described for the long form leptin receptor149.

A 1.3% agarose gel was prepared by combining 100 ml of 1 x TBE (Tris, boric acid, and

EDTA), 1.3g of agarose and 4 µl of RedSafe Nucleic Acid Staining Solution (INtRON

Biotechnology,). 25 µl of the PCR product containing 5X Gel Loading Dye (New England

Biolabs) was then run through the gel from the negative to positive electrode at ~90 V. DNA

ladders were included in the gel for determination of product size. Bands were visualized under

Page 125: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

112

ultra violet light with a BioRad Molecular Imager Gel Doc XR+ Imaging System (BioRad,

Hercules, CA, USA).

4.3.10 PI3K Activity Assay

4.3.10.1 Immunoprecipitation of PI3K and Assay Reaction

PI3K activity was measured with the PI3K activity ELISA: Pico kit (echelon, K-1000s;

Salt Lake City, UT, USA). On day 1 of the experiment, PI3K was immunoprecipitated from 500

µg of protein, prepared as described above in 4.3.7, with 5 µl of an anti-p89 PI3K subunit

antibody (Millipore; Billerica, MA, USA). A negative control was included with no addition of

the antibody. The lysate was incubated for 1 hour at 4°C in a rotating wheel (Mini Labroller,

Diamed lab supplies, Edison, NJ, USA) with the antibody. Next, 25 µl of 25% protein A/G

beads (Santa Cruz, CA, USA) were added to the lysate and was incubated for an additional 2

hours at 4°C in a rotating wheel. The beads were then spun at 8000 rpm for 1 min at 4°C and the

supernatant was kept and frozen for later use if needed. The remaining pellet underwent a

washing protocol: washed 3 times with 500 µl of the lysis buffer (please refer to 4.3.7) + 0.1M

NaCl, three times with 500 µl of 0.1 M Tris-HCl pH 7.4, 5 mM LiCl and 0.1 mM sodium

orthovanadate and twice with 500 µl of 10 mM Tris-HCl pH 7.4, 150 mM NaCl, 5 mM EDTA

and 0.1 mM sodium orthovanadate. The KBZ reaction buffer was then prepared by combining

the KBZ buffer with 1M DTT, 10mM ATP and ultrapure water, according to the number of

samples in the assay. The last wash was then aspirated and the beads were re-suspended in 30 µl

of the KBZ reaction buffer. Then, a 100 µM PI(3,4,5)P2 (PIP2) working solution was prepared

with the addition of distilled water to the substrate vial. 30 µl of the PIP2 substrate was also

added to the beads. A positive control was added by combining 5 µl of purified PI3K with KBZ

reaction buffer and PIP2 substrate. The reaction was conducted through incubation of the

reaction containing eppendorf tubes at 37°C for 4 hours. After 4 hours, the reaction was stopped

Page 126: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

113

through addition of 90 µl of the Kinase Stop Solution provided by the kit. The tubes were then

centrifuged at 8000 rpm for 2 min at 4°C. The supernatant was transferred to clean tubes and

stored at -20°C.

4.3.10.2 ELISA Incubation and Detection

The following were prepared on ice: a standard curve buffer (KBZ reaction buffer +

PIP2 substrate + K-EDTA), PIP3 standard stock at 3.6 µM (distilled water into PIP3 vial), and

TBS-T buffer (distilled water + 10x TBS-T buffer). The stopped kinase reactions described

above were thawed on ice and PIP3 standards and controls for the ELISA for prepared (360 n,

120 nM, 40 nM, 13.3 nM, 4.4 nM, no enzyme control, no lipid control). 60 µl of the standards,

no enzyme control and no lipid control, and stopped kinase reactions were transferred to the

incubation plate provided by the kit. Then, 60 µl of the PIP3 detection buffer was added to each

well where the plate was sealed and incubated for 1 hour at room temperature on a plate shaker.

100 µl from each well was transferred to the detection plate provided by the kit. The detection

plate was sealed and incubated for 1 hour at room temperature on a plate shaker. After

aspiration, the wells were washed 3 times with 200 µl of TBS-T and 100 µl of secondary

detector solution was added to each well and the plate was incubated for 30 min. Each well was

aspirated and 300 µl of TMB solution was added. Color was allowed to develop for 15 minutes

in a dark space. The reaction was stopped by adding 50 µl of 1N H2SO4 stop solution. The plate

was transferred to a spectrophotometer and the absorbance was read at 450nm. The statistical

software program Prism (GraphPad Software Inc., CA, USA) was used for data analysis. Data

was presented as fold increase over the saline treated samples.

4.3.11 Biochemical Analysis

Please refer to the General Methods section 2.6.1-2.6.3 for details on biochemical

analyses. Plasma glucose concentrations were determined using a GM9 Analox Glucose

Page 127: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

114

Analyzer (Analox Instruments, Lunenbertg, MA). Radioactivity of plasma glucose was

conducted as described. Plasma insulin levels were measured using a radioimmunoassay (Linco

Research, St Charles, MO).

4.3.11.1 Plasma Leptin

Peripheral and portal plasma leptin levels were measured using a rat Leptin RIA kit

(Linco Research, St. Charles, MO). This RIA kit follows the same principle as that described in

the 2.5.3 Plasma insulin section. Briefly, leptin from peripheral and portal plasma samples or

standards compete for binding to a guinea pig anti-rat leptin antibody against 125I-labeled leptin.

The amount of radiolabeled 125I-labeled leptin binds in reverse proportion to the known

standards and the amount of leptin in the plasma sample. Separation of the 125I-labeled leptin

and unbound fractions is conducted through the use of a double antibody solid phase.

Specifically, a three day protocol as per the supplier’s instructions was used. 50 µl of

peripheral and portal plasma samples and standards in a range of concentrations (0.78, 1.56,

3.125, 6.25, 12.5, 25 ng/ml) were prepared and 50 µl of the anti-leptin antibody was added. The

tubes were vortexed and allowed to incubate at room temperature overnight. After the first day,

50 µl of 125I-labeled leptin was added and samples were vortexed and allowed to incubate

overnight at room temperature. 500 µl of precipitating reagent was added followed by vortexing

and incubation at 4°C for 20 minutes. The samples were then centrifuged to pellet the bound

leptin and the radioactivity of this pellet was counted by a gamma counter (Perkin Elmer 1470).

The final concentration of leptin in the samples was conducted through construction of a

standard curve and interpolation.

4.3.12 Calculations and Statistical Analysis

Data are represented as mean ± SEM. Data between t = 60-90 (rats) and t = 50-60 (mice)

were averaged for basal conditions and data between t = 150-200 (rats) and t = 120-170 (mice)

Page 128: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

115

were averaged for clamp conditions. Statistical difference between two groups was determined

via unpaired Student’s t-test. When comparisons were made across more than two groups,

ANOVA was performed, and if significant, this was followed by Tukey’s post-hoc test, which

enabled comparisons of all treatment groups.

4.4 Results

4.4.1 Jejunal leptin requires jejunal leptin receptor activation to lower glucose production

We first assessed whether Leprb is expressed in duodenal and jejunal tissues via PCR

technology. Consistent with previous reports97,103,410,411,416, PCR analyses revealed Leprb

expression in both the duodenum and jejunum of normal rats (Figure 4.2). To assess whether

stimulation of the Leprb in the small intestine, which is classically known to mediate the

metabolic effects of leptin, possesses the ability to regulate glucose production, we subjected

fully recovered conscious healthy rats to a pancreatic (basal insulin) clamp when leptin was

administered directly into the duodenum or jejunum during the final 50 min of the experiment

(Table 4.1; The infusion-clamp experiments lasted a total of 200 min; basal period is averaged

from 60-90 min and clamp period is averaged from 180-200 min) (Figure 4.1). Interestingly,

despite the presence of the Leprb in the duodenum, an intraduodenal leptin infusion did not

affect glucose metabolism as evident by the fact that the glucose infusion rate (Figure 4.3A),

glucose production (Figure 4.3B and C) and glucose uptake (Figure 4.3D) remained

comparable to the vehicle control. In contrast, a continuous intrajejunal leptin infusion for 50

min led to a higher glucose infusion rate (Figure 4.3A) and lower glucose production (Figure

4.3B and C) compared to saline. No changes in glucose uptake (Figure 4.3D) and plasma

insulin and glucose levels (Table 4.1) were detected.

Next, we wanted to confirm that this effect of leptin is specific to the jejunum, as it could

be argued that the decrease in glucose production induced by intrajejunal leptin administration is

Page 129: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

116

due to a leakage of leptin into the portal and subsequent peripheral circulation resulting in

activation of extra-intestinal leptin receptors (i.e. in the hypothalamus). We measured leptin

levels in plasma samples taken from both portal and peripheral blood at the end of the 50 min

gut infusion period. Importantly, an intrajejunal infusion of leptin for 50 min did not elevate

plasma leptin levels during the clamps (averaged from 180-200 min) (Figure 4.4A) and did not

alter portal leptin levels at the end of the experiments (Figure 4.4A). To alternatively ensure

changes of glucose production in response to an intrajejunal leptin infusion occurred

independent of changes in circulating leptin levels, we performed an intravenous administration

of leptin at an equal dose and duration as the intrajejunal leptin infusion. An intravenous leptin

administration for 50 min given at the equal dose as the intrajejunal leptin infusion did not alter

glucose metabolism (Figure 4.4B-D). Thus, an intrajejunal (but not intraduodenal) infusion of

leptin lowers glucose production independently of changes in plasma leptin, insulin and glucose

levels. Subsequent clamp studies were conducted to delineate the downstream effectors involved

in leptin’s effects in the jejunum.

To determine whether the binding of leptin to the Leprb in the jejunum is responsible for

lowering glucose production, we co-infused leptin with a SLR previously shown to rapidly bind

to leptin and antagonize its effects499. An intrajejunal infusion of the SLR alone for 50 min did

not alter glucose metabolism (Figure 4.5A-D) but fully negated the effect of leptin (Figure

4.5A-D). To further evaluate that the Leprb is required for jejunal leptin-sensing to lower

glucose production, we performed the same set of jejunal leptin infusion experiments in a Leprb

deficient rodent model, the obese (fak/fak) Koletsky rat versus the lean Fa/Fa control. First, an

intrajejunal leptin 50 min infusion led to a higher glucose infusion rate (Figure 4.6A) and lower

glucose production (Figure 4.6B and C) with no changes in glucose uptake (Figure 4.6D)

compared to saline in lean Fa/Fa rats. When placed on a regular chow diet ad lib, fak/fak rats are

hyperphagic and rapidly increase body weight compared to their lean Fa/Fa littermates500. To

Page 130: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

117

match their body weight to the Fa/Fa control rats, we limited the food intake of the fak/fak rats

similar to that described473 and assessed jejunal leptin action in weight- and age-matched leptin

receptor deficient fak/fak rats. An intrajejunal leptin infusion failed to alter glucose metabolism

compared to saline in the leptin receptor deficient fak/fak rats (Figure 4.6A-D).

We next tested jejunal leptin action in the leptin receptor-deficient obese db/db mice.

We first developed a murine intrajejunal infusion model to directly activate jejunal sensing

mechanisms as performed in the rat, by implanting a catheter directly into the jejunum of mice.

We then subjected fully recovered conscious mice to a pancreatic (basal insulin) clamp while

leptin was administered continuously and directly into the jejunum for 50 min. Similar to what

was discovered in rats, an intrajejunal leptin infusion in healthy C57Bl6/J mice led to a higher

glucose infusion rate (Figure 4.7A) and lower glucose production (Figure 4.7B and C) with no

changes in glucose uptake (Figure 4.7D) compared to saline. Importantly, this glucose lowering

effect was independent of a rise in plasma leptin levels since intravenous leptin infusion

administered at an equal dose and duration as intrajejunal leptin infusion did not alter glucose

metabolism in healthy C57Bl6/J mice (Figure 4.8A-D). Importantly, intrajejunal leptin infusion

was unable to alter the glucose infusion rate (Figure 4.7A) and glucose production (Figure 4.7B

and C) in the weight- and age-matched leptin receptor deficient db/db mice, confirming our

observations in the obese Koletsky rats. These leptin receptor loss-of-function experiments

demonstrate that activation of jejunal leptin receptors by preabsorptive leptin lowers glucose

production in rats and mice.

4.4.2 A STAT3-independent and PI3K-dependent signaling pathway is required for

jejunal leptin to lower glucose production via a neuronal network

Although STAT3474 and PI3K473 are required for central leptin to regulate glucose, and

both signaling molecules are expressed in the intestine and/or in vagal afferent nerves151,157,485,

Page 131: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

118

the functional impact of the intestinal leptin-STAT3/PI3K signaling is unknown. To address the

role of jejunal STAT3, we co-infused the STAT3 peptide inhibitor (STAT3 PI) with leptin

directly in the jejunum for 50 min. In contrast to the hypothalamus474, STAT3 PI failed to negate

the effect of jejunal leptin compared to saline in rats (Figure 4.9A-D). A higher jejunal STAT3

phosphorylation (Y705) with leptin compared to saline was detected (Figure 4.10A) in the

tissues taken immediately after the clamp studies. Co-infusion of leptin with STAT3 PI negated

the elevation of STAT3 phosphorylation (Y705) (Figure 4.10A) but did not prevent the gluco-

regulatory effect (Figure 4.9A-D). Thus, jejunal leptin activates local STAT3 but such

activation is not required for the gluco-regulatory impact of jejunal leptin.

To investigate the role of jejunal PI3K, we co-infused two independent PI3K inhibitors,

LY294002 or wortmannin, with leptin into the jejunum for 50 min. An intrajejunal infusion of

LY294002 or wortmannin alone did not affect whole-body glucose metabolism (Figure 4.9A-

D) but fully abolished the effect of leptin (Figure 4.9A-D). PI3K activity was assessed from

jejunal tissues taken immediately after the clamp studies. An intrajejunal leptin infusion led to a

higher jejunal PI3K activity compared to saline (Figure 4.10B), and this activation was negated

by co-infusion with LY294002 (Figure 4.10B). In the jejunal tissues that were obtained

immediately after an intrajejunal leptin plus SLR infusion where the SLR negated the effect of

leptin (Figure 4.5A-D), an intrajejunal leptin administration failed to activate PI3K as well

(Figure 4.10B), confirming jejunal PI3K is a target of the leptin receptor. Of note, an

intrajejunal leptin administration did not activate duodenal PI3K activity (Figure 4.10C) or

hypothalamic STAT3 (Figure 4.10D) in the same rats where jejunal PI3K was activated

(Figure 4.10C). Further, an intraduodenal leptin infusion activated duodenal STAT3 (Figure

4.10E) but not duodenal PI3K (Figure 4.10F). Although it is tempting to speculate that the

inability of intraduodenal leptin infusion to lower glucose production (Figure 4.3A-D) is due to

an absence of PI3K activation, future studies are needed to address this possibility as well as the

Page 132: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

119

reasons behind the inability of duodenal leptin to activate PI3K. Nonetheless, selective

activation of the jejunal leptin receptors by presabsorptive leptin triggers jejunal PI3K to lower

glucose production.

Given that jejunal nutrient-sensing ignites afferent neuronal signals to lower glucose

production402, a neuronal network is a potential downstream effector of jejunal leptin signaling.

We infused the topical anesthetic tetracaine locally into the jejunum for 50 min to inhibit

neurotransmission of the nerves that innervate the jejunum402 while leptin was infused. An

intrajejunal infusion of tetracaine alone did not affect whole-body glucose metabolism (Figure

4.11A-D) but was sufficient to fully reverse the ability of intrajejunal leptin administration to

increase the exogenous infusion rate (Figure 4.11A) and lower glucose production (Figure

4.11B and C) during the clamp while glucose uptake (Figure 4.11D) remained comparable

within groups. Thus, neuronal transmission is required for jejunal leptin to lower glucose

production.

4.4.3 Jejunal leptin’s action remain intact in high fat fed or diabetic rats

Recent studies highlight leptin as an anti-diabetic therapy501,502 in which leptin’s effect is

attributed to the brain475,476. In parallel, direct administration of i.c.v leptin is effective to lower

glucose production in 3d high-fat fed rats474. We have previously characterized this 3 day high

fat fed model as having duodenal CCK resistance384,487 as well as insulin resistance393. Thus, we

tested the effectiveness of jejunal leptin action in this same 3d high-fat fed model. An

intrajejunal leptin infusion for 50 min led to a higher glucose infusion rate (Figure 4.12A) and

lowered glucose production (Figure 4.12B and C) compared to saline with no changes in

glucose uptake (Figure 4.12D) in high-fat fed rats. The gluco-regulatory effect of jejunal leptin

action in high-fat feeding was comparable to that observed in regular chow fed rats (Figure

4.12A-D). We again tested whether this effect of jejunal leptin in high fat diet rats is

Page 133: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

120

independent of a rise in circulating leptin levels. Indeed, an intravenous leptin infusion

administered at an equal dose and duration as an intrajejunal leptin infusion did not alter glucose

metabolism in 3d high-fat fed rats (Figure 4.13A-C). These findings indicate that the ability of

jejunal leptin to lower glucose production in this model is specific to the jejunum in the current

experimental conditions. These findings suggest that agonism of jejunal leptin signaling may

represent a novel therapeutic approach for obesity and diabetes.

We next investigated jejunal leptin action in non-obese insulin-deficient uncontrolled

diabetic rats. We injected the rats with STZ to elevate plasma glucose concentrations and

glucose production to ~340 mg dl-1 (Figure 4.14A) and ~23 mg kg-1 min-1 (Figure 4.14B)

respectively, and reduce insulin concentrations by ~80% (Figure 4.14D). An intrajejunal

infusion of leptin for 50 min was effective to induce a reduction in plasma glucose levels

(Figure 4.14A) and glucose production (Figure 4.14B) in non-clamped conditions in

comparison to saline. Consistent with our findings in the various models used in this study, an

intravenous leptin infusion at the same dose and duration did not lower plasma glucose levels or

glucose production in the same diabetic model (Figure 4.15 A and B). These findings indicate

that the ability of jejunal leptin to lower plasma glucose levels and glucose production in this

non-obese insulin-deficient uncontrolled diabetic rodent model is specific to the jejunum. This

glucose- and glucose production-lowering effect of jejunal leptin was independent of changes in

circulating glucagon (Figure 4.14C) and insulin (Figure 4.14D) levels as well. We reason that

if jejunal leptin action (like jejunal nutrient sensing402) lowers plasma glucose levels and glucose

production in insulin-deficient uncontrolled diabetic rats, jejunal leptin action (like jejunal

nutrient sensing) may mediate the rapid early anti-diabetic effects of DJB surgery.

Page 134: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

121

4.4.4 The antidiabetic effect of DJB surgery is mediated by jejunal leptin action

We next investigated whether gastric leptin action in the jejunum contributes to the

glucose-lowering effect of DJB surgery in non-obese STZ-induced uncontrolled diabetes. In

order to test this hypothesis, we inhibited jejunal leptin action in uncontrolled diabetic rats that

received DJB while monitoring plasma glucose levels during refeeding. The use of refeeding as

our experimental design is based on the fact that refeeding will cause secretion of leptin from

the gastric chief cells into the lumen127,143 and makes its way to the jejunum (as the duodenum is

bypassed) to subsequently bind to the Leprb in the jejunum144 and possibly lower plasma

glucose levels in diabetic rodents that have received DJB surgery. First, DJB or sham surgery

was performed as described440 in STZ-induced uncontrolled diabetic rats (Figure 4.16).

Consistent with our previous study402, DJB exerted a rapid reduction of fed plasma glucose

levels compared to sham in STZ-induced diabetic rats within 2 d after surgery (Figure 4.17A).

This effect was not associated with changes in plasma insulin (Figure 4.17B) or glucagon

(Figure 4.17C) levels nor changes in food intake (Before surgery STZ-SHAM 27g ± 0.7 and

STZ-DJB 31g ± 1.3, 2d after surgery STZ-SHAM 4g ± 1.3 and STZ-DJB 5g ± 1.4) or body

weight (Before surgery STZ-SHAM 301g ± 11.2 and STZ-DJB 314g ± 4.3, 2d after surgery

STZ-SHAM 291g ± 14.5 and STZ-DJB 305g ± 8.8).

After confirming our model, we then tested whether jejunal leptin sensing mediates this

early improvement of glycemia induced by DJB in uncontrolled diabetes. We inserted a jejunal

catheter, targeting the same location where the jejunal leptin receptor antagonist SLR infusion

negated the effect of leptin (Figure 4.3A-D), in conjunction with DJB in STZ-diabetic rats

(Figure 4.16). To this end, we then carried out a fasting-refeeding experiment in these rodents

to promote gastric leptin secretion127 while infusing the SLR directly into the jejunum to disrupt

jejunal leptin signaling (Figure 4.16) in STZ-diabetic rats 2 d after DJB. We monitored plasma

Page 135: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

122

glucose levels for 50 min during refeeding in an attempt to match the duration of the intrajejunal

leptin infusion during the clamp studies (Figure 4.3A-D). Consistent with a glucose-lowering

effect of DJB in STZ-induced diabetic rats, the glucose control stimulated by jejunal leptin

sensing during refeeding was intact in STZ-DJB diabetic rats that received intrajejunal saline

infusion as compared to STZ-SHAM (Figure 4.17D, top panel). This glucose control was

independent of changes in food intake (Figure 4.17E). In contrast, an intrajejunal infusion of

the SLR into STZ-DJB rats disrupted the glucose control observed in the STZ-DJB intrajejunal

saline infused rats during refeeding, resulting in elevated plasma glucose concentrations (Figure

4.17D, bottom panel). This marked elevation of plasma glucose levels seen with intrajejunal

SLR infusion occurred independently of changes in food intake (Figure 4.17E) but did not

reach that of STZ-SHAM jejunal saline infused rats (Figure 4.17D, top panel). Nonetheless,

these findings illustrate that gastric leptin action in the jejunum contributes to the rapid (2 d)

glucose-lowering effect induced by DJB in uncontrolled diabetes.

4.5 Discussion

Previous studies focus on the brain as a primary tissue mediating leptin’s effects on the

regulation of glucose homeostasis158. Our discovery revises the traditional view of leptin action

and suggests that, in addition to the brain, leptin triggers a jejunal signaling pathway to lower

glucose production. The physiological relevance of intestinal leptin action remains to be

clarified as (i) jejunal but not duodenal leptin action lowers glucose production and (ii) short-

term inhibition of jejunal leptin receptor-mediated action (via 50 min intrajejunal SLR or PI3K

inhibitors infusion) per se did not alter glucose production. These unknowns however are

balanced with current findings reporting that selective activation of jejunal leptin-PI3K lowers

glucose production in rats and mice.

Page 136: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

123

The current study demonstrates that activation of the jejunal leptin receptor is required

for preabsorptive leptin to activate PI3K to lower glucose production, which was confirmed

through the use of two molecular knockout and chemical inhibitory approaches. STAT5 and

SOCS3 have also been demonstrated to be downstream signaling molecules of the leptin

receptor in the hypothalamus136,503 and both are expressed in the intestine151,156. It remains to be

assessed whether these are potential targets of jejunal leptin action. We also made the interesting

discovery that jejunal-leptin PI3K, and not STAT3 signaling lowers glucose production. Thus,

the role of intestinal STAT3 activation warrants future investigation. In regards to a potential

effector of jejunal PI3K activation, voltage gated sodium channels remain a possibility as co-

infusion of leptin with the anesthetic tetracaine (an inhibitor of voltage gated sodium channels)

abolished leptin’s glucose production suppression effects. In line with this hypothesis, one study

suggests that PI3K alters sodium conductance504. To our knowledge, no similar studies exist for

STAT3. In addition, the current study at best narrows down the site of leptin-PI3K signaling to

the jejunum (i.e., jejunal mucosa and/or the vagal nerves that innervate the jejunum). The exact

site in the GI tract (i.e. cell type involvement) remains to be addressed. Nonetheless, it is clear

that neuronal innervation is required for the gluco-regulatory effect of jejunal leptin.

Previous studies have demonstrated that intestinal peptide hormones, like leptin, regulate

glucose homeostasis via a neuronal axis. For example, the peptide hormone CCK is secreted in

the duodenum upon lipid ingestion and activates PKA to regulate glucose production through a

neuronal network384,487. In addition, selective inhibition of intestinal DPP-IV, the enzyme which

rapidly degrades GLP-1, leads to activation of vagal afferents to improve glucose tolerance in

diet-induced obese rodents285. These findings, together with the discovery of leptin in the

current study, indicate that peptide hormones bind to their receptors expressed in the GI tract to

trigger the CNS to regulate glucose homeostasis. Interestingly, leptin in the brain has been

shown to enhance the action of intestinal CCK to decrease feeding9,477 and possibly at the level

Page 137: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

124

of the intestine157,505 where both the CCK-1 receptor155 and the Leprb149 are expressed. Thus,

jejunal leptin and CCK may act additively or synergistically to regulate glucose production,

although it would be important to first assess whether CCK action in the jejunum (like in the

duodenum) regulates glucose production.

Given that a jejunal leptin infusion lowers glucose production in high-fat fed rats, jejunal

leptin signaling may have therapeutic relevance. It has been previously demonstrated that leptin

resistance occurs after 3 days of high fat feeding392, but a direct infusion of leptin into the

brain474 and jejunum still lower glucose production. However, it remains to be assessed whether

jejunal leptin action through a neuronal network remains intact in chronic obese models.

Moreover, both central and peripheral leptin administration in STZ-induced insulin deficient

uncontrolled diabetic rodents normalizes plasma glucose concentrations in association with

lowering hyperglucagonemia475,476,501. We here demonstrate that jejunal leptin in this same

rodent model lowers (but does not normalize) plasma glucose concentrations through an

inhibition of glucose production, which was independent of changes in circulating glucagon

levels. However, it should be noted that glucagon levels were not elevated in this model to begin

with. This is consistent with the previous findings that a jejunal glucose and lipid infusion

lowered glucose levels in this same model, also independent of changes in glucagon levels. In a

more chronic model of uncontrolled diabetes402, the involvement of glucagon action in

mediating jejunal leptin’s effects may be more apparent.

Upon intake of nutrients into the gastrointestinal tract, there is release of different

peptides hormones364,506,507 including gastric leptin127. There is still much debate over the role of

gut peptides involvement in the glucose lowering effect of DJB with studies demonstrating

conflicting results64,402,445. Nonetheless, the involvement of bile acids is also becoming apparent

and it is suggested that they may also contribute to these beneficial effects450. Given that after

DJB surgery the route of delivery of nutrients from the stomach is redirected into the jejunum,

Page 138: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

125

this would stimulate gastric leptin release, which would make its way to the jejunum to possibly

activate jejunal leptin receptors to mediate the rapid anti-diabetic effect of DJB. We here

discovered that upon blocking leptin receptor signaling through infusion of a SLR during

refeeding in uncontrolled diabetic rodents with DJB disrupted the glucose control albeit this

blockade did not elevate glucose levels to the same extent as seen in uncontrolled diabetic

rodents who had received sham surgery. This suggests that leptin does not work alone to lower

glucose concentrations after DJB surgery but requires additional mechanisms. It is worth

examining whether jejunal leptin converges with nutrient sensing mechanisms as this may

uncover the additional mechanisms that are required. In addition to the uncontrolled diabetic

rodent model used in this study, it remains to be clarified whether jejunal leptin action mediates

the anti-diabetic effects of other types of bariatric surgery in obese and diabetic models.

In conclusion, we here unveil that leptin action in the jejunum activates a jejunal leptin-

receptor-PI3K dependent pathway to lower glucose production. This effect of jejunal leptin

remains intact in high-fat fed or uncontrolled diabetic rodents and mediates the rapid anti-

diabetic effect of DJB surgery. Taken together, these findings suggest that jejunal leptin

signaling may be targeted as a novel therapeutic strategy to lower glucose levels in diabetes.

Page 139: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

126

Figure 4.1 Schematic representation of the working hypothesis – Gastric leptin activates the intestinal long form leptin receptor to activate a PI3K-dependent and STAT-3 independent signaling axis to lower glucose production through a neuronal network.

Proposed model for intestinal leptin to lower glucose production. The soluble leptin receptor (SLR) is a leptin receptor inhibitor, which binds leptin and prevents its binding to the receptor. Koletsky rats and db/db mice are long form leptin receptor deficient rodents. Signal transducer and activator of transcription 3 peptide inhibitor (STAT3 PI) is a STAT3 inhibitor and LY294002 and wortmannin are PI3K inhibitors. Tetracaine is a local anesthetic that prevents neuronal activation. An intestinal leptin infusion fails to lower glucose production upon blockade of leptin receptor, PI3K and vagal afferent activation.

Page 140: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

127

Figure 4.2 Leptin receptor expression in intestinal tissue.

PCR analysis of the long form leptin receptor in both duodenal and jejunal mucosa tissue. –RT: negative control run in the absence of reverse transcriptase. A 375bp project was amplified by primers for the long form leptin receptor.

Page 141: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

128

Figure 4.3 Jejunal leptin administration lowers glucose production.

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups). C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (**P < 0.01 vs other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 5–7 rats per group; n = 7–8 mice per group.

Page 142: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

129

Figure 4.4 Jejunal leptin lowers glucose production independent of changes in portal and circulating leptin levels.

A) Plasma leptin levels before (basal) and at the end of the camp (t = 180-200) during a jejunal saline or leptin infusion. Portal leptin levels were taken at the termination of the clamp studies (t = 200). (B and C) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups). An intravenous leptin infusion at the same dose and duration did not affect the B) glucose infusion rate or C) glucose production. D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean + s.e.m. n = 5–7 rats per group.

Page 143: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

130

Figure 4.5 Leptin activates leptin receptors to lower glucose production in rats (chemical approach).

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups). Co-infusion of the SLR negated the ability of jejunal leptin to A) increase the glucose infusion rate and B) lower glucose production. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (**P < 0.01 vs. other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 5–7 rats per group. SLR, soluble leptin receptor.

Page 144: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

131

Figure 4.6 Leptin activates leptin receptors to lower glucose production in lean fa/fa rats but not in fak/fak (Koletsky) long form leptin receptor deficient rats (molecular approach).

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups) in lean fa/fa rats but not fak/fak rats. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (**P < 0.01 vs. other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 5–7 rats per group.

Page 145: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

132

Figure 4.7 Jejunal leptin activates leptin receptors to lower glucose production in C57BL/6 but not db/db mice (molecular approach).

(A and B) During the pancreatic clamp (t = 150-170) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, ***P < 0.001 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups) in C57BL/6 but not db/db mice. C) Suppression of glucose production during the clamp period (t = 150-170) expressed as the percent reduction from the basal state (t = 50-60) glucose production (**P < 0.01 vs other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 7-8 mice per group.

Page 146: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

133

Figure 4.8 Jejunal leptin lowers glucose production in C57BL/6 independent of changes in circulating leptin levels.

(A and B) During the pancreatic clamp (t = 150-170) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, ***P < 0.001 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups). An intravenous leptin infusion at the same dose and duration did not affect the A) glucose infusion rate or B) glucose production. D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean + s.e.m. n = 7-8 mice per group.

Page 147: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

134

Figure 4.9 Jejunal leptin lowers glucose production through a STAT3-independent and PI3K dependent pathway.

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups). Co-infusion of LY294002 or wortmannin negated the ability of jejunal leptin to A) increase the glucose infusion rate and B) lower glucose production. A STAT3 PI co-infusion with leptin did not abolish leptin’s effects. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (**P < 0.01 vs. other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 5–7 rats per group. STAT3 PI, signal transducer and activator of transcription 3 peptide inhibitor LY, LY294002, Wort, wortmannin

Page 148: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

135

Figure 4.10 Jejunal and duodenal leptin activate intestinal STAT3, and only jejunal leptin activates jejunal PI3K.

A) The level of phosphorylation of STAT3 analyzed by western blot analysis and expressed as a fold increase over saline in the jejunum obtained from rats at the end of the clamp studies (*P < 0.05 vs. other groups). B) PI3K activity measured in the jejunum at the end of the clamp expressed as a fold increase over a jejunal saline infusion (*P < 0.05 vs. other groups). The increase in PI3K activity was abolished upon co-infusion with LY or the SLR. C) PI3K activity in the jejunum or duodenum at the end of the clamp with jejunal saline or jejunal leptin infusion expressed as fold increase over saline. D) The level of STAT3 phosphorylation/total STAT3 in the hypothalamus obtained at the end of the clamp studies in rats that received a jejunal saline or leptin infusion. Analyzed by western blot and expressed as fold increase over saline. (E and F) The level of phosphorylation of STAT3/total STAT3 (E, *P < 0.05 vs. saline) analyzed by western blot and expressed as fold increase over saline and F) PI3K activity in duodenal tissues obtained at the end of the rat clamp studies that received a duodenal saline or leptin infusion. Values are shown as mean + s.e.m. n = 5–7 rats per group. STAT3 PI, signal transducer and activator of transcription 3 peptide inhibitor LY, LY294002, SLR, soluble leptin receptor.

Page 149: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

136

Figure 4.11 Jejunal leptin lowers glucose production through a neuronal network.

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups). Co-infusion of tetracaine negated the ability of jejunal leptin to A) increase the glucose infusion rate and B) lower glucose production. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (**P < 0.01 vs. other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 5–7 rats per group.

Page 150: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

137

Figure 4.12 Jejunal leptin lowers glucose production in high fat diet fed rats.

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, **P < 0.01 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups) in both regular chow and high fat diet fed rats. C) Suppression of glucose production during the clamp period (t = 180-200) expressed as the percent reduction from the basal state (t = 60-90) glucose production (**P < 0.01 vs. other groups). D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean ± s.e.m. n = 5–7 rats per group.

Page 151: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

138

Figure 4.13 Jejunal leptin lowers glucose production in high fat diet fed rodents independent of a rise in plasma leptin levels.

(A and B) During the pancreatic clamp (t = 180-200) an intrajejunal leptin infusion (6.7 ng/min) increased the glucose infusion rate (A, ***P < 0.001 vs. other groups) and decreased glucose production (B, **P < 0.01 vs. other groups) in high fat diet fed rats. An intravenous leptin infusion at the same dose and duration did not affect the A) glucose infusion rate or B) glucose production. D) The rate of glucose uptake remained unchanged amongst all groups. Values are shown as mean + s.e.m. n = 5-6 rats per group.

Page 152: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

139

Figure 4.14 Jejunal leptin lowers plasma glucose levels and glucose production in uncontrolled diabetic rodents independent of changes in plasma insulin and glucagon levels.

(A and B) In non-clamped conditions, a jejunal leptin infusion (6.7 ng/min) decreased plasma glucose levels (* P < 0.05 vs. saline) and glucose production (* P < 0.05 vs. saline). C) Plasma glucagon levels during a jejunal saline or leptin infusion. D) Plasma insulin levels during a jejunal saline or leptin infusion. Values are shown as mean + s.e.m. n = 5-6 rats per group. STZ, streptozotocin.

Page 153: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

140

Figure 4.15 Jejunal leptin lowers plasma glucose levels and glucose production in uncontrolled diabetic rodents independent of a rise in plasma leptin levels.

(A and B) In non-clamped conditions, a jejunal leptin infusion (6.7 ng/min) decreased plasma glucose levels (* P < 0.05 vs. saline) and glucose production (* P < 0.05 vs. saline). An intravenous leptin infusion at the same dose and duration did not affect the A) plasma glucose levels or B) glucose production. Values are shown as mean + s.e.m. n = 5-6 rats per group. STZ, streptozotocin.

Page 154: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

141

Figure 4.16. Schematic of duodenal-jejunal bypass (DJB) surgery and jejunal catheter placement.

Point A: Proximal to the pyloric sphincter, a cut is made and the duodenal stump is closed off. Point B: The jejunum is reconnected to the stomach and a jejunal catheter was inserted into the lumen. Point C: The distal end of duodenal stump connected to the distal end of the jejunum/proximal ileum.

Page 155: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

142

Figure 4.17 Jejunal leptin action mediates the rapid anti-diabetic effect of DJB surgery.

A) Fed plasma glucose levels obtained from STZ-induced diabetic rats 2 days after sham or DJB surgery (* P < 0.05 vs. SHAM). DJB exerts a rapid glucose lowering effects 2 days after the DJB surgery. (B and C) Fed plasma insulin B) and glucagon C) levels before STZ injection, after STZ injection (before surgery) and 2d after surgery (C, ** P < 0.01 vs. other groups). (D, top panel) Plasma glucose levels during refeeding in STZ-diabetic rats with DJB vs. SHAM surgery and infused with intrajejunal saline. (D, bottom panel) Plasma glucose levels during refeeding in STZ-diabetic rats with DJB surgery and infused with intrajejunal saline or the SLR (φ STZ-DJB-jejunal saline vs. STZ-SHAM-jejunal saline, * STZ-DJB-jejunal saline vs. STZ-DJB-jejunal SLR, *P < 0.05, **P < 0.01, *** P < 0.001, ****P < 0.0001 (same for both symbols)). Blockade of leptin signaling through an SLR infusion results in dysregulated glucose homeostasis during refeeding. E) Accumulated food intake during the refeeding protocol. Values are shown as mean ± s.e.m. n = 5–6 rats per group. SLR, soluble leptin receptor, STZ, streptozotocin.

Page 156: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

143

Table 4.1 Plasma insulin and glucose concentrations of groups receiving intrajejunal infusions during the basal and clamp conditions

Data are means ± SEM (basal: t = 60-90 min, clamp: 180-200 min).

Page 157: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

144

Chapter 5Summary and Conclusions

5.1 Summary of Studies in this Thesis

Previous studies in our laboratory have demonstrated the existence of duodenal lipid !

PKC-δ ! CCK ! CCK-1 receptor signaling pathway that triggers a gut-brain-liver neuronal

axis to lower glucose production, which is abolished in rodents fed a high fat diet for three days.

This demonstrates that these rodents acquire duodenal CCK resistance upon short term high fat

feeding. To begin locating the potential site(s) of resistance, the first study of this thesis

delineated the downstream signaling pathway of the CCK1 receptor. In pancreatic acinar cells

the CCK1 receptor has been demonstrated to signal through PKA but it is unknown whether the

duodenal CCK1 receptor shares this same signaling pathway. In this regard, we utilized the

pancreatic (basal insulin) euglycemic clamp technique to address whether direct activation of

duodenal PKA signaling lowers glucose production. We demonstrated that direct duodenal PKA

activation ignites vagal afferent firing to activate NR1 containing NMDA receptors within the

DVC to lower glucose production and lies downstream of the CCK1 receptor. Interestingly,

direct activation of duodenal PKA in rodents fed a high fat diet bypassed CCK resistance and

lowered glucose production. This study provides evidence that CCK resistance arises, in part,

from the inability of the CCK1 receptor to activate the downstream signaling molecule PKA.

Similar to the duodenum, nutrient infusion into the jejunum (both lipids and glucose)

triggers a gut-brain-liver axis to lower glucose production, and contributes to the glucose

lowering effect of DJB surgery. It remains unknown whether gastrointestinal peptides can also

trigger this same neuronal network and also play a similar role in DJB surgery. Although most

studies focus on adipocyte-derived leptin and its glucoregulatory effects within the CNS, leptin

Page 158: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

145

is produced in gastric chief cells of the stomach and regulates various intestinal functions. Given

these findings, the focus of study 2 was to address whether leptin in the jejunum regulates

glucose production and contributes to the glucose lowering effect of DJB surgery. In this regard,

through the use of the pancreatic (basal insulin) clamp technique, we first demonstrated that a

jejunal leptin administration lowered glucose production through a long form leptin receptor-

PI3K dependent and STAT3-independent manner, which required a neuronal network. We

further demonstrated that jejunal leptin action remains intact in both STZ induced uncontrolled

diabetic rodents as well as in high fat fed rats. Lastly, we demonstrated that gastric derived

leptin may contribute to the glucose lowering effect of DJB surgery as blockade of leptin

signaling during refeeding in diabetic rodents who received DJB surgery resulted in a

dysregulation in glucose homeostasis. Thus, this study demonstrates a glucoregulatory role of

leptin within the intestine and suggests that enhancing leptin-PI3K signaling in the jejunum may

lower plasma glucose concentrations in diabetes.

5.2 General Summary

This doctoral thesis demonstrates that independent activation of the duodenal CCK-

PKA and jejunal leptin-PI3K signaling axis lowers glucose production in normal, high-fat fed

and diabetic rodents via a gut-brain-liver neuronal axis (Figure 5.1).

5.3 General Conclusion

Through the two studies in this thesis, we have demonstrated that independent local

duodenal CCK and jejunal leptin signaling, in contrast to peripheral effects of intestinal

hormonal signaling discussed in the general introduction, regulates glucose production through a

neuronal network. This opens a new area of research whereby targeting local hormonal

signaling, in addition to peripheral hormonal affects, could trigger the nervous system to

regulate glucose homeostasis.

Page 159: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

146

Figure 5.1 Summary of duodenal and jejunal hormonal signaling that triggers a neuronal network to lower glucose production

In Study 1 we demonstrated that the duodenal CCK1 receptor signals through PKA to trigger vagal afferent firing and activated NR1 containing NMDA receptors within the DVC to lower glucose production. Importantly, direct activation of duodenal PKA bypassed CCK-resistance in short-term high fat fed rodents. Moreover, Study 2 demonstrated that jejunal leptin triggers a leptin receptor ! PI3K signaling axis to lower glucose production, which remained intact in both high fat fed and STZ rodents. Further, jejunal leptin action mediates the early anti-diabetic effects of DJB surgery.

Page 160: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

147

Chapter 6General Discussion

6.1 Do nutrient sensing mechanisms interact with both CCK and leptin?

Following lipid entry into the duodenum and uptake into intestinal cells, LCFA are

metabolized into LCFA-CoA by ACS to activate PKC-δ to cause CCK secretion to subsequently

activate a CCK1-receptor ! PKA signaling cascade to lower glucose production508. Intralipid is

an emulsion containing different fatty acids with varying degrees of saturation. Thus, it remains

in question whether individual fatty acids within this emulsion cause CCK secretion to lower

glucose production. Interestingly, it has been demonstrated that individual fatty acids may

differentially regulate gastrointestinal processes. For instance, intestinal infusions of individual

monounsaturated versus polyunsaturated fatty acids differ in their effectiveness to reduce food

intake509 possibly through differences in their ability to slow gastric emptying510 and release of

gut peptides511. Moreover, the monounsaturated fatty acid, oleic acid, has been demonstrated to

cause secretion of CCK in the STC-1 cell line176. Thus, it remains to be addressed whether

individual fatty acids affect glucose production and cause CCK secretion and PKA signaling or

whether different signaling mechanisms are involved.

In addition to lipids, both glucose and proteins have been demonstrated to cause the

release of CCK. In regards to proteins, individual amino acids such as phenylalanine172 and

tryptophan173 can stimulate CCK release. Phenylalanine has been shown to reduce food intake in

association with an increase in circulating CCK levels in human patients512. The mechanism

upstream of CCK release by amino acids may involve a calcium sensing receptor (CaSR) that

was originally found in parathyroid cells. This receptor has recently been found in CCK

secreting cells and has been shown to stimulate CCK release in conjunction with an increase in

Page 161: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

148

intracellular calcium levels513. Thus, whether amino acids trigger CCK release to regulate

glucose production remains to be uncovered and may involve the CaSR receptor. In addition,

glucose uptake into cells via SGLT-1 has been shown to be required for jejunal glucose sensing

to regulate glucose production402. Although it remains unknown whether glucose administration

into the duodenum lowers glucose production, SGLT-1 is expressed in the proximal intestine514.

This suggests that if the duodenum senses an increase in glucose entry into the intestine to lower

glucose production, uptake into intestinal cells and subsequent release of CCK may play a role.

In regards to gastric leptin secretion, it has been demonstrated that the presence of

nutrients in the stomach causes the release of leptin which makes it way into the

duodenum127,143. However, these studies conducted refeeding experiments with rat chow diet

that encompasses carbohydrates, lipids and proteins. Thus, this suggests that all forms of

nutrients can cause leptin release from the stomach. However, what remains unknown is

whether any of these nutrients are more potent stimulators of leptin release, as certain

carbohydrates are more potent than others515. Given that study 2 demonstrates a beneficial

glucoregulatory effect of leptin after bypass surgery, it would be of interest to determine which

nutrient causes the greatest release of leptin from the gastric chief cells in the stomach.

Moreover, once gastric leptin is secreted and makes its way to the jejunum to activate

Leprb receptors, nutrients will also be found within the small intestine. As discussed in section

1.4.2 of the general introduction, intestinal nutrients (glucose and lipids) have been shown to

regulate glucose levels in both normal and uncontrolled diabetic conditions402. Whether glucose

and lipid sensing mechanisms converge at the level of the intestine remains unknown. However,

within the hypothalamus, convergence of glucose and lipid sensing occurs via an adenosine

monophosphate activated protein (AMPK) – malonyl-CoA – CPT-1 pathway516. More

specifically, hypothalamic glucose is converted to acetyl-CoA, which is subsequently converted

to malonyl-CoA via acetyl-CoA carboxylase (ACC). AMPK is a negative regulator of ACC,

Page 162: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

149

which when inhibited, allows for inhibition of CPT-1 by malonyl-CoA, which disrupts the

transfer of LCFA-CoA into the mitochondria to undergo β-oxidation. These processes together

sense hypothalamic glucose and lipids to regulate glucose production516. Interestingly, leptin has

been shown to inhibit hypothalamic AMPK activity to decrease feeding517 and AMPK518 and

CPT-1519 are expressed in the intestine. This raises the possibility that lipid sensing mechanisms,

and possible convergence with glucose sensing mechanisms, in the intestine are required for

leptin action to regulate glucose homeostasis.

6.2 What other intestinal hormones share similar signaling mechanisms as CCK

and leptin?

6.2.1 PKA

Study 1 demonstrates that a duodenal CCK! CCK1 receptor ! PKA signaling cascade

exists to regulate glucose production. In addition to the CCK1 receptor, PKA is also activated

by many different GPCRs. Specifically, the GLP-1R is known to signal through a PKA

mediated pathway in the β cell308 as well as the brain305. It is traditionally believed that GLP-1 is

secreted from L cells within the ileum to exert its effects. However, as discussed in section

1.3.3.1 of the Introduction, it is now currently debated whether L cells are also found in the

proximal small intestine271. Given that GLP-1R are expressed on vagal afferents270, perhaps

GLP-1 within the duodenum also activates PKA to regulate glucose homeostasis. In line with

this finding, inhibition of DDP-IV, the enzyme that rapidly degrades GLP-1, within the small

intestine through administration of an oral DDP-IV inhibitor, activates vagal afferents and

improves glucose tolerance285. Thus, it remains possible that GLP-1 mediated activation of PKA

in the small intestine may contribute to the regulation of glucose homeostasis.

In addition to GLP-1, other intestinal hormone receptors are GPCRs that signal through

PKA. GIP has been shown to activate PKA to regulate GSIS263. However, the GIPR is

Page 163: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

150

expressed in the intestine261 but is not found to be expressed in vagal afferents270 suggesting that

GIP may not activate PKA within vagal afferents. In contrast, the GLP-2 receptor also activates

PKA to regulate GSIS332 and has been found to be expressed in vagal afferents and increase

activity within the NTS upon activation520. Given that L cells may exist in the proximal

intestine, GLP-2 may signal through duodenal PKA, but whether GLP-2 triggers a neuronal

network to regulate glucose production remains to be investigated.

6.2.2 PI3K

Study 2 demonstrated that activation of PI3K is required for jejunal leptin to lower

glucose production. Interestingly, in addition to activating PKA and PLC, CCK has also been

shown to activate PI3K in pancreatic acinar cells521. Thus it may be possible that CCK signaling

in the duodenum shares common signaling with jejunal leptin by activating PI3K to lower

glucose production. Although a duodenal leptin administration failed to activate PI3K and

subsequently did not lower glucose production, the leptin receptor is a tyrosine kinase associated

receptor that requires autophosphorylation of tyrosine residues to cause activation of

downstream molecules. This is different from GPCR signaling which is mediated by G proteins.

Thus, it remains possible that the CCK1 receptor could also signal via PI3K to regulate glucose

production. Moreover, the GLP-2R has been shown to signal through PI3K in the brain347.

Whether this signaling occurs in the intestine remains unknown, but given that activation of the

GLP-2R on vagal afferents signals to the NTS as described above, GLP-2 could signal through

PI3K in addition to PKA to trigger a neuronal network like CCK and leptin. Similar to GLP-2,

the GLP-1R has also been found to signal via PI3K in the brain to regulate food intake304. Given

that GLP-1R signaling in the upper intestine may regulate glucose homeostasis, it remains to be

addressed whether GLP-1R signaling via PI3K triggers a similar axis as CCK and leptin.

Page 164: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

151

6.3 What is the cellular location of CCK-PKA and leptin-PI3K signaling in the

intestine?

The current set of studies in this thesis set out to dissect intestinal hormone signaling

mechanisms and the regulation of glucose production. Study 1 demonstrated that PKA lies

downstream of CCK to trigger a gut-brain-liver axis to lower glucose production where study 2

demonstrated that jejunal leptin administration activates PI3K to trigger a neuronal network (and

likely a gut-brain-liver axis previously demonstrated for jejunal nutrient sensing) to lower

glucose production. However, the current set of data does not address the exact location of these

hormonal signaling pathways. What is known from these studies is that these signaling

pathways occur in the mucosa and/or smooth muscle layer of the small intestine as the samples

measured for activation of downstream signaling molecules were conducted in these layers.

Given the complexity of the small intestinal tract in terms of cell types, it remains to be

addressed whether these signaling cascades take place in intestinal cells such as enteroendocrine

cells, epithelial cells or exclusively in vagal afferents that innervate both the smooth muscle and

mucosal layer of the small intestine.

In regards to PKA signaling, study 1 measured PKA activity from the whole tissue

encompassing both the mucosal and smooth muscle layer. However, the findings that activation

of PKA lowered glucose production in the presence of the CCK1 receptor inhibitor, MK-329,

demonstrates that PKA lies downstream of the CCK1 receptor and not upstream. It has been

demonstrated that the CCK1 receptor is expressed on vagal afferents innervating the small

intestine522. Thus, given our findings as well as the findings of CCK1 receptor expression, it

remains likely that PKA signaling occurs in vagal afferent neurons. To better confirm this

hypothesis, anterograde labeling could be used, a technique that involves injection of an

antibody into the nodose ganglion that travels down the vagus nerve to the small intestine, in

Page 165: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

152

order to visualize the innervation through immunohistochemistry155. Through co-staining for

PKA, whether PKA is found within vagal afferent neuronal cells could be determined.

Long form leptin receptor expression is more complex in the small intestine as it has

been found to be expressed in many different cell types including epithelial cells480, CCK and

GLP-1 secreting cells143,160 and vagal afferents innervating the small intestine149–151. Given this

complexity of expression, the intestinal effects of leptin could be mediated by any one of these

various cell types. In study 2, we measured long form leptin receptor expression and PI3K

activity separately in the mucosal and smooth muscle layer and detected leptin receptor

expression and an increase in PI3K activity in both layers. Unfortunately, this does not narrow

down the cell type responsible for mediating leptin-induced activation of PI3K and subsequent

regulation of glucose production. However, two recent studies153,154 begin to narrow down the

cell type that may not be responsible for jejunal leptin’s effects. In brief, these groups generated

epithelial cell specific long form leptin receptor knock out mice through the crossing of villin-

Cre mice with Leprflox/flox mice. Interestingly, these specific knock out mice were no different

from the control mice in terms of food intake and body weight regulation. Given these findings,

it may be that epithelial cell long form leptin receptor activation is not responsible for mediating

the glucoregulatory effect of jejunal leptin. In contrast to these findings, a recent paper

demonstrated that deletion of the long form leptin receptor in vagal afferent neurons leads to

hyperphagia and obesity. These findings demonstrate that vagal afferent long form leptin

receptors, rather than receptors in epithelial cells, play a role in mediating leptin’s effects152. In

order to better address the hypothesis that long form leptin receptors specifically in vagal

afferent neurons regulate glucose homeostasis as a follow up to Study 2 in the current thesis, the

pancreatic clamp technique could be performed in both knock out mice with a jejunal leptin

infusion, which would be expected to lower glucose production in epithelial cell long form

leptin receptor knock out if leptin receptor and subsequent PI3K activation within epithelial

Page 166: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

153

cells is not required for jejunal leptin’s glucoregulatory effect and would be abolished in mice

with a deletion of the long form leptin receptor in vagal afferents. In fact, the latter technique

could be utilized for PKA signaling (using CCK1 receptorflox/flox mice) to confirm PKA

activation within vagal afferent neuronal cells is required for CCK to lower glucose production.

Furthermore, as suggested for PKA, the visualization of leptin receptors in vagal afferents could

be conducted through the use of anterograde tracing and immunohistochemistry.

6.4 What is the relevance of CCK and leptin signaling in disease models?

A common theme throughout this thesis is to begin to dissect local intestinal hormonal

signaling mechanisms in the regulation of glucose production in both normal and diabetic/obese

settings in hopes to unveil therapeutic targets to lower blood glucose concentrations in diabetes

and obesity. Indeed, our previous studies suggest that even short term exposure of the duodenum

to a diet high in fat content causes early onset insulin resistance and disrupts the ability of

duodenal lipids to activate the CCK1 receptor to lower glucose production. In study 1, we

uncovered a possible site of duodenal CCK resistance, which is the inability of the CCK1

receptor to activate PKA. As such, one possible therapeutic target to overcome this resistance

after high fat feeding is to directly activate PKA to lower glucose levels.

In fact, the therapeutic potential of targeting the gastrointestinal tract, and subsequently

PKA signaling to regulate glucose homeostasis in diabetes and obesity has recently been

established for metformin and resveratrol (metformin is the most widely prescribed type 2

diabetic drug, and resveratrol is a polyphenolic compound well known for its insulin sensitizing

effects). In regards to metformin, in the same three day high fat diet model used in the current

thesis, an acute metformin infusion for 50 minutes during the basal insulin euglycemic

pancreatic clamp lowered glucose production via a neuronal network523. This was independent

of its direct effect on the liver60 as a metformin infusion at the same dose and duration into the

Page 167: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

154

portal vein failed to lower glucose production in the 50 minute time frame. This preabsorptive

effect of metformin required GLP-1R and subsequent PKA activation as co-infusion of

metformin with exendin-9 (GLP-1R antagonist) or Rp-CAMPS negated the metformin induced

suppression of glucose production (Figure 5.1). Similarly, in another study, resveratrol was

infused for 50 minutes during a hyperinsulinemic euglycemic pancreatic clamp in the same

three day high fat fed rodent model and improved hepatic insulin sensitivity and lowered

glucose production524. This effect was also dependent on PKA activation, as seen with

metformin (Figure 5.1). Thus, these studies demonstrate that anti-diabetic compounds such as

metformin and resveratrol utilize a GLP-1R-PKA signaling cascade to lower glucose levels in

an early onset insulin resistant, duodenal CCK resistant, high fat diet rodent model. More

importantly, metformin and resveratrol were still effective to regulate glycemia in 28 day high

fat diet induced obese and insulin resistant rodents, as well as in

nicotinamide/streptozotocin/high fat diet fed induced type 2 diabetic rodents suggesting that

PKA signaling remains intact in more chronic disease models. This strengthens the idea that

PKA signaling could be targeted in diabetes and obesity to lower glucose levels.

Another potential therapeutic strategy to lower blood glucose concentrations is the use of

gastric bypass surgery, which is commonly performed in obese patients and has been shown to

have beneficial effects on glucose homeostasis. Surgical procedures such as RYGB and DJB

surgery bypass the proximal duodenum and connect the jejunum to the stomach, where RYGB

also alters the stomach size. Thus, it is likely that these surgical procedures bypass the

duodenum that is unresponsive to lipids (and possibly other nutrients) and directs nutrient flow

into the jejunum in which hormonal signaling likely remains intact. This is suggested in study 2,

as a direct leptin administration into the jejunum activated a leptin receptor-PI3K signaling

cascade to lower glucose production in high fat fed or uncontrolled diabetic rodents via a

neuronal network, and contributed to the early anti-diabetic effect of bariatric surgery. In line

Page 168: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

155

with these findings, a recent study reported increased intestinal PI3K activation after RYGB

surgery525. Whether nutrient sensing and/or other hormones lie upstream or downstream of

jejunal leptin signaling to regulate glucose levels after DJB surgery remains to be addressed.

However, study 2 suggests that alternate mechanisms are required for the rapid lowering of

glycemia after DJB surgery given the findings that disrupting jejunal leptin signaling in STZ-

DJB rats during refeeding did not disrupt glucose control to the extent of that seen in STZ-sham

rats. Given that leptin has been demonstrated to cause GLP-1 secretion160, and the finding that

DJB surgery in STZ rodents elevated plasma GLP-1 levels 2 days after surgery402, GLP-1 may

play a role in mediating the rapid glucose lowering effect (Figure 5.1). However, in the

autoimmune diabetes-prone Biobreeding rat, DJB surgery rapidly lowered glucose levels

independent of a rise in GLP-1 levels402 and thus the functional relevance of GLP-1 in

mediating the rapid anti-diabetic effect of DJB surgery remains to be clarified. Dissecting

whether jejunal leptin signaling converges with nutrient sensing mechanisms402 may shed light

on additional mechanisms required (Figure 5.1). In fact, a recent study suggests that after

RYGB surgery, the number of CCK secreting I cells increases in the roux and common limbs526.

Although study 1 demonstrated that PKA activation in the jejunum did not regulate glucose

production, CCK signaling in the jejunum may still be relevant as in addition to PKA, duodenal

CCK also signals through PLC. It is worth investigating whether nutrient sensing in the

jejunum402 causes CCK release to work in concert with leptin to regulate glucose levels after

DJB surgery, as leptin has been previously shown to enhance the ability of CCK to decrease

food intake156. In summary, our results, in combination with other group’s findings, suggest that

activation of jejunal leptin-PI3K signaling (and possibly other hormonal pathways) may mimic

the anti-diabetic effect of bariatric surgery.

Page 169: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

156

Chapter 7Limitations of the Studies

1. One of the major limitations to the current studies is that we do not demonstrate the cell

type where CCK and leptin activate downstream molecules PKA and PI3K, respectively.

As mentioned in the general discussion, it is likely that PKA signaling occurs in vagal

afferent neuronal cells where the CCK1 receptor is expressed522, which remains to be

confirmed. Furthermore, we currently cannot rule out the possibility that leptin acts on

leptin receptors on epithelial480, enteroendocrine143 or neuronal cells149–151 as the long

form leptin receptor has been found to be expressed on each cell type. The current study

only suggests that PI3K activation occurs within both the mucosal and smooth muscle

layer. Thus, as discussed above, it would be of interest to utilize either visualization

techniques or epithelial/vagal specific knock out models to address the exact location of

PKA and PI3K signaling triggered by CCK and leptin, respectively.

2. In study 2, the use of whole body long form leptin receptor knock out models was used

to address the involvement of the receptor in mediating the effects of jejunal leptin on

glucose production suppression. In addition to these models, we also utilized co-infusion

of leptin with the SLR, which binds leptin and prevents it from binding to its receptor.

We are currently lacking a molecular intestinal specific knock down approach for the

long form leptin receptor. Due to the time constraints of this thesis, the production of an

adenovirus expressing shRNA to the long form leptin receptor was not conducted.

However, this could be done as a future experiment as we have previously developed

adenoviruses expressing shRNA to MEK1527.

Page 170: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

157

3. In both studies, an intestinal infusion of various compounds was given for 50 minutes.

Given this long duration of infusion, it is possible that although we aim to target specific

regions of the small intestine (either the duodenum or jejunum) these compounds may

leak into other regions during the clamp studies. In study 1, we did address this in

regards to a Sp-CAMPs (PKA activator) infusion. Sp-CAMPs was given directly into the

jejunum during the pancreatic clamp, which had no effect on the glucose kinetics

suggesting that PKA activation was specific to the duodenum to lower glucose

production during a duodenal Sp-CAMPS infusion (and not due to a leak into the

jejunum). We did not examine whether CCK-8 administration into the jejunum triggers a

neuronal network to lower glucose production, which could be addressed by giving a

direct jejunal CCK-8 infusion during the pancreatic clamp studies. In regards to leptin,

we first gave a duodenal leptin infusion and did not see any changes in the glucose

kinetics even though the receptor is expressed in the duodenum. We then demonstrated

that a direct jejunal leptin infusion lowered glucose production, confirming that a

duodenal leptin administration did not leak into the jejunum during the clamp studies.

Whether a jejunal leptin infusion travels to the ileum to regulate glucose production

remains unknown. This region of the small intestine has yet to be studied in regards to

triggering a gut-brain-liver axis although the long form leptin receptor is said to be

expressed in L cells in the ileum160, suggesting that such an axis could exist for leptin in

this region. This could also be addressed by a direct infusion of leptin into the ileum

during the pancreatic clamp.

4. The pancreatic clamp technique is used to address whether activation of intestinal

hormonal signaling regulates glucose production in the absence of changes in circulating

glucoregulatory hormones, which does not directly address the physiological relevance

of such signaling. The physiological relevance of such activation can be addressed

Page 171: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

158

through the use of a refeeding non-clamp protocol and an intravenous glucose tolerance

test where circulating glucoregulatory hormones are allowed to change at will. For the

fasting and refeeding protocol, PKA activation or leptin receptor signaling could be

inhibited to see whether this results in a dysregulation in glycemia. In addition, an

intravenous glucose tolerance test could be conducted with infusion of Sp-CAMPS into

the duodenum or leptin into the jejunum, to address whether activation of either

signaling pathway improves glucose tolerance and whether inhibiting these pathways

disrupts the improvement. Together these techniques would demonstrate that CCK-PKA

and leptin-PI3K signaling regulates glucose homeostasis in a more physiological setting.

Page 172: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

159

Chapter 8Future Directions

1. It has previously been demonstrated that duodenal lipid metabolism is required for CCK

release and subsequent activation of a gut-brain-liver axis to lower glucose production.

Study 1 extends these findings by demonstrating that PKA activation is required for the

glucose production lowering effect of CCK. Given that other nutrients also lead to CCK

release as discussed in 1.3.2.1, these nutrients may trigger CCK release to activate a gut-

brain-liver axis through activation of PKA. If so, this would suggest that CCK acts as a

converging point for duodenal nutrients to regulate glucose homeostasis. Furthermore, it

would also be of interest to then establish whether diets high in sugar or protein also

cause duodenal CCK resistance, as this would demonstrate that a balanced diet is

integral to maintain the regulation of glucose homeostasis by intestinal nutrient sensing

mechanisms. It would also be of interest to determine whether leptin requires lipid-

sensing mechanisms to regulate glucose homeostasis, which has been suggested for

leptin in regards to the regulation of food intake at the level of the hypothalamus.

2. Another future direction, and major limitation to the current studies, is that the exact

localization of peptide hormone signaling is at best narrowed down to the smooth muscle

and/or mucosal layer of the small intestine. Given that both of the leptin receptor149–151

and CCK1 receptor528 are expressed on vagal afferents innervating the small intestine, as

described previously, our current hypothesis is that upon binding of these hormones to

their receptors, downstream signaling events are activated to increase vagal afferent

firing. In order to better address the exact location of signaling, we could first use

anterograde tracing of vagal afferent neurons combined with immunohistochemistry to

Page 173: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

160

co-localize the molecules of interest with specific cell types. To assess the requirement

of various cell types the following could be conducted: i) create Nav1.8-Cre Leprflox/flox

or CCK1 receptorflox/flox mice to knock out the leptin receptor or CCK1 receptor in vagal

afferent neurons or ii) generate an adenovirus expressing shRNA to either the CCK1

receptor or leptin receptor which could be injected into the nodose ganglion. Upon

confirmation of knocking down either receptor from neuronal cells, infusion of CCK-

8/Sp-CAMPS or leptin into the intestine during the pancreatic clamp could be conducted

to address the requirement of CCK1 receptor or leptin receptor activation in vagal

afferent neurons. This would demonstrate that both PKA and PI3K are localized within

vagal afferents, and address which cell type CCK-1 and leptin receptor activation is

required to exert their glucoregulatory effect.

3. Although the studies in this thesis dissect hormonal signaling mechanisms in two

different regions of the small intestine in regards to the control of glucose homeostasis, it

remains to be addressed whether these hormones could work together in order to control

glucose homeostasis as has been demonstrated for the regulation of food intake. Given

that duodenal leptin administration failed to lower glucose production, it is likely that

leptin and CCK do not work together in this region of the small intestine. In regards to

the jejunum, CCK secreting I cells have been located to this region of the small intestine.

Thus, it may be that leptin and CCK work together in the jejunum to lower glucose

production. In order to address this question, it would first have to be determined

whether a CCK-8 administration into the jejunum lowers glucose production during the

pancreatic clamp. Given that in study 1, the administration of the PKA activator Sp-

CAMPS into the jejunum failed to lower glucose production, if CCK does indeed have

an effect in the jejunum, this would be a PKA-independent effect. Rather, jejunal CCK

could activate a CCK1 ! PLC signaling pathway to regulate glucose production as a co-

Page 174: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

161

infusion of CCK-8 with a PLC inhibitor into the duodenum abolished the suppression in

glucose production. After demonstrating a gluco-regulatory effect of CCK in the

jejunum, a co-infusion of CCK with the SLR or leptin with MK-329 could address

whether these hormones work together to regulate glucose production. It would also be

interesting to address whether co-administration of leptin and CCK have synergistic or

additive effects at the level of the jejunum by direct co-infusion of both hormones during

the pancreatic clamp.

4. The current studies in this thesis begin to look at the downstream signaling molecules for

both the CCK1 receptor and long form leptin receptor. There are other common

downstream signaling molecules activated by these receptors, which also may play a role

in mediating their effects on glucose production regulation. In regards to CCK1 receptor

signaling, study 1 further indicates that a PLC dependent pathway may be activated

downstream of the receptor as blockade of PLC signaling disrupted the ability of a CCK-

8 administration to lower glucose production. Interestingly, CCK also activates PI3K in

pancreatic acinar cells521. Thus, it may be possible that CCK signaling in the duodenum

shares common signaling with jejunal leptin by activating PI3K to lower glucose

production. Co-infusion with the PI3K inhibitors used in study 2 with CCK-8 during the

pancreatic clamp could address the possibility. Furthermore, whether intestinal leptin

activates PLC to cause activation of PI3K remains unknown, as demonstrated in the

brain529. This could also be addressed by co-infusion of leptin with the PLC inhibitor

used in study 1 during the pancreatic clamp.

5. Both studies in this thesis use an early onset model of insulin resistance induced through

short-term high fat feeding to address whether both PKA activation in the duodenum and

a jejunal leptin administration were still able to lower glucose production. This model

has important implications as addressing whether intestinal CCK and leptin action

Page 175: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

162

remains intact may unveil the intestine as a site of hormone action which can be targeted

to lower glucose production before hyperglycemia occurs. Nonetheless, it still remains

important to address whether these signaling mechanisms also remain intact after longer

term high fat feeding or in genetically obese models. In order to address this question, a

more chronic high fat diet/STZ rodent model of type 2 diabetes530 could be used

whereby intestinal infusions of CCK-8/Sp-CAMPS and leptin could be performed and

plasma glucose levels could be monitored to see whether a glucose lowering effect

remains. For study 1 and 2, we could also utilize diabetic db/db mice to address whether

direct activation of PKA and/or PI3K lowers glucose levels. In addition, in study 2 the

effect of jejunal leptin was tested in the STZ induced uncontrolled diabetic rodent model.

Leptin’s effects remained intact in this model whereby a jejunal leptin administration

lowered plasma glucose levels and glucose production in hyperglycemic rats. This

occurred in the absence of a suppression of glucagon levels seen previously with a CNS

leptin administration. Thus, it remains to be addressed whether a jejunal leptin infusion

would similarly lower glucose levels and production in a more chronic STZ model

through suppression of hyperglucagonemia.

6. In study 2, a duodenal leptin administration failed to lower glucose production. Upon

looking at downstream signaling molecules, we found that duodenal leptin activated

STAT3, but failed to activate PI3K. Given our findings that jejunal leptin activated PI3K

to lower glucose production, we believe that the failure of duodenal leptin to lower

glucose production is due to its inability to activate PI3K. In this regard, it would be of

interest to determine why duodenal leptin fails to activate PI3K. Upon binding of the

leptin to its receptor, there is subsequent phosphorylation of three key tyrosine residues

that are involved in activating different downstream signaling pathways. STAT3 is

activated by Tyr1138 recruitment and subsequent phosphorylation by Jak2 where PI3K is

Page 176: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

163

activated by Jak2 phosphorylation of IRS158. Thus, examining whether IRS is expressed

in the duodenum, or if IRS fails to be phosphorylated by duodenal leptin administration,

may address why duodenal leptin fails to activate PI3K and regulate glucose production.

Page 177: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

164

Chapter 9References

1. Ng, Marie, Tom Fleming, Margaret Robinson, Blake Thomson, Nicholas Graetz, Christopher Margono, E. C. M. et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766–781 (2014).

2. Kelsey, M. M., Zaepfel, A., Bjornstad, P. & Nadeau, K. J. Age-related consequences of childhood obesity. Gerontology 60, 222–8 (2014).

3. World Health Organization. WHO | Obesity and overweight. (2013). at <http://www.who.int/mediacentre/factsheets/fs311/en/>

4. Public Health Agency of Canada. Obesity in Canada.

5. Cummings, D. E. & Overduin, J. Gastrointestinal regulation of food intake. J.Clin.Invest 117, 13–23 (2007).

6. Blouet, C. & Schwartz, G. J. Duodenal lipid sensing activates vagal afferents to regulate non-shivering brown fat thermogenesis in rats. PLoS One 7, e51898 (2012).

7. Koch, L. et al. Central insulin action regulates peripheral glucose and fat metabolism in mice. J. Clin. Invest. 118, 2132–2147 (2008).

8. Buettner, C. et al. Leptin controls adipose tissue lipogenesis via central, STAT3-independent mechanisms. Nat. Med. 14, 667–75 (2008).

9. Morton, G. J., Cummings, D. E., Baskin, D. G., Barsh, G. S. & Schwartz, M. W. Central nervous system control of food intake and body weight. Nature 443, 289–295 (2006).

10. Dodd, G. T. et al. Leptin and Insulin Act on POMC Neurons to Promote the Browning of White Fat. Cell 160, 88–104 (2015).

11. Boyd, K. & O’donovan, D. High-fat diet effects on gut motility, hormone, and appetite responses to duodenal lipid in healthy men. Am. J. Physiol. Gastrointest. Liver Physiol. 5000, 188–196 (2003).

12. Covasa, M. & Ritter, R. C. Reduced sensitivity to the satiation effect of intestinal oleate in rats adapted to high-fat diet. Am.J.Physiol 277, R279–R285 (1999).

13. Covasa, M, Marcuson, J, Ritter, R. Diminished satiation in rats exposed to elevated levels of endogenous or exogenous cholecystokinin. Am. J. Physiol. Regul. Integr. Comp. Physiol. 6520, 331–337 (2001).

Page 178: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

165

14. Covasa, M. & Ritter, R. C. Rats maintained on high-fat diets exhibit reduced satiety in response to CCK and bombesin. Peptides 19, 1407–1415 (1998).

15. Covasa, M., Grahn, J. & Ritter, R. C. High fat maintenance diet attenuates hindbrain neuronal response to CCK. Regul. Pept. 86, 83–8 (2000).

16. Savastano, D. & Covasa, M. Adaptation to a high-fat diet leads to hyperphagia and diminished sensitivity to cholecystokinin in rats. J. Nutr. 135, 1953–1959 (2005).

17. Carvalheira, J. B. C. et al. Selective impairment of insulin signalling in the hypothalamus of obese Zucker rats. Diabetologia 46, 1629–40 (2003).

18. De Souza, C. T. et al. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 146, 4192–9 (2005).

19. Posey, K. a et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. Am. J. Physiol. Endocrinol. Metab. 296, E1003–12 (2009).

20. Begg, D. P. et al. Reversal of diet-induced obesity increases insulin transport into cerebrospinal fluid and restores sensitivity to the anorexic action of central insulin in male rats. Endocrinology 154, 1047–54 (2013).

21. Filippi, B. M. et al. Insulin signals through the dorsal vagal complex to regulate energy balance. Diabetes 63, 892–9 (2014).

22. Levin, B. & Dunn-Meynell, A. Reduced central leptin sensitivity in rats with diet-induced obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 1095, 941–948 (2002).

23. Ozcan, L. et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 9, 35–51 (2009).

24. Widdowson, P. S., Upton, R., Buckingham, R., Arch, J. & Williams, G. Inhibition of Food Response to Intracerebroventricular Injection of Leptin Is Attenuated in Rats With Diet-Induced Obesity. Diabetes 46, 1782–1785 (1997).

25. Howard, J. K. et al. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat. Med. 10, 734–8 (2004).

26. Mori, H. et al. Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nat. Med. 10, 739–43 (2004).

27. Halaas, J. L. et al. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc.Natl.Acad.Sci.U.S.A 94, 8878–8883 (1997).

28. Zhang, X. et al. Hypothalamic IKKB-NK-KB and ER Stress Link Overnutrition to Energy Balance and Obesity. Cell 135, 61–73 (2008).

Page 179: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

166

29. Montague, C. T. et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387, 903–8 (1997).

30. Yeo, G. S. et al. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat. Genet. 20, 111–2 (1998).

31. Panaro, B. L. et al. The Melanocortin-4 Receptor Is Expressed in Enteroendocrine L Cells and Regulates the Release of Peptide YY and Glucagon-Like Peptide 1 In Vivo. Cell Metab. 1–12 (2014).

32. Berglund, E. D. et al. Melanocortin 4 receptors in autonomic neurons regulate thermogenesis and glycemia. Nat. Neurosci. 17, 911–3 (2014).

33. Fall, T. & Ingelsson, E. Genome-wide association studies of obesity and metabolic syndrome. Mol. Cell. Endocrinol. 382, 740–57 (2014).

34. Frayling, T. M. et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316, 889–94 (2007).

35. Smemo, S. et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507, 371–5 (2014).

36. Hakala, P., Rissanen, A., Koskenvuo, M., Kaprio, J. & Rönnemaa, T. Environmental factors in the development of obesity in identical twins. Int. J. Obes. Relat. Metab. Disord. 23, 746–53 (1999).

37. Luke, A. & Cooper, R. S. Physical activity does not influence obesity risk: time to clarify the public health message. Int. J. Epidemiol. 42, 1831–6 (2013).

38. Jakicic, J. M. Physical activity and weight loss. Nestle Nutr. Inst. Workshop Ser. 73, 21–36 (2012).

39. Hansen, D. & Toubro, S. The effect of sibutramine on energy expenditure and appetite during chronic treatment without dietary restriction. Int. J. Obes. 23, 1016–1024 (1999).

40. Anthes, E. Marginal gains. Nature 508, 8–10 (2014).

41. Rodgers, R. J., Tschöp, M. H. & Wilding, J. P. H. Anti-obesity drugs: past, present and future. Dis. Model. Mech. 5, 621–6 (2012).

42. Thivel, D. et al. Surgical weight loss: impact on energy expenditure. Obes. Surg. 23, 255–66 (2013).

43. Madura, J. a & Dibaise, J. K. Quick fix or long-term cure? Pros and cons of bariatric surgery. F1000 Med. Rep. 4, 19 (2012).

44. Rubino, F., Moo, T.-A., Rosen, D. J., Dakin, G. F. & Pomp, A. Diabetes surgery: a new approach to an old disease. Diabetes Care 32 Suppl 2, S368–72 (2009).

Page 180: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

167

45. Canadian Diabetes Assocation. Obesity. - (2013). at <http://www.diabetes.ca/research/obesity/>

46. IDF Diabetes Atlas: Sixth edition. (2013).

47. Patterson, C. et al. Diabetes in the young - a global view and worldwide estimates of numbers of children with type 1 diabetes. Diabetes Res. Clin. Pract. 103, 161–75 (2014).

48. D’Adamo, E. & Caprio, S. Type 2 diabetes in youth: epidemiology and pathophysiology. Diabetes Care 34 Suppl 2, S161–5 (2011).

49. Canadian Diabetes Association. The prevelance and cost of diabetes. - (2013). at <http://www.diabetes.ca/diabetes-and-you/what/prevalence/>

50. Lecavalier, L., Bolli, G., Cryer, P. & Gerich, J. Contributions of gluconeogenesis and glycogenolysis during glucose counterregulation in normal humans. Am. J. Physiol. 256, E844–51 (1989).

51. DeFronzo, R. A. Pathogenesis of type 2 diabetes mellitus. Med. Clin. North Am. 88, 787–835, ix (2004).

52. Rayner, C. K., Samsom, M., Jones, K. L. & Horowitz, M. Relationships of upper gastrointestinal motor and sensory function with glycemic control. Diabetes Care 24, 371–81 (2001).

53. Horowitz, M., Edelbroek, M. A., Wishart, J. M. & Straathof, J. W. Relationship between oral glucose tolerance and gastric emptying in normal healthy subjects. Diabetologia 36, 857–62 (1993).

54. Del Prato, S., Marchetti, P. & Bonadonna, R. C. Phasic Insulin Release and Metabolic Regulation in Type 2 Diabetes. Diabetes 51, S109–S116 (2002).

55. Bluestone, J. A., Herold, K. & Eisenbarth, G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature 464, 1293–300 (2010).

56. Lewis, G. F., Carpentier, A., Adeli, K. & Giacca, A. Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr. Rev. 23, 201–29 (2002).

57. Lin, Y. & Sun, Z. Current views on type 2 diabetes. J. Endocrinol. 204, 1–11 (2010).

58. Raskin, P. & Unger, R. H. Hyperglucagonemia and its suppression. Importance in the metabolic control of diabetes. N. Engl. J. Med. 299, 433–6 (1978).

59. Taylor, S. I. Deconstructing type 2 diabetes. Cell 97, 9–12 (1999).

Page 181: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

168

60. Pawlyk, a. C., Giacomini, K. M., McKeon, C., Shuldiner, a. R. & Florez, J. C. Metformin Pharmacogenomics: Current Status and Future Directions. Diabetes 63, 2590–2599 (2014).

61. Drucker, D. J. et al. Incretin-based therapies for the treatment of type 2 diabetes: evaluation of the risks and benefits. Diabetes Care 33, 428–33 (2010).

62. Mingrone, G. et al. Bariatric surgery versus conventional medical therapy for type 2 diabetes. N.Engl.J.Med. 366, 1577–1585 (2012).

63. Schauer, P. R. et al. Bariatric surgery versus intensive medical therapy in obese patients with diabetes. N. Engl. J. Med. 366, 1567–76 (2012).

64. Jiao, J. et al. Restoration of euglycemia after duodenal bypass surgery is reliant on central and peripheral inputs in Zucker fa/fa rats. Diabetes 62, 1074–83 (2013).

65. Sala, P. C., Torrinhas, R. S., Giannella-Neto, D. & Waitzberg, D. L. Relationship between gut hormones and glucose homeostasis after bariatric surgery. Diabetol. Metab. Syndr. 6, 87 (2014).

66. Berthoud, H. R. & Powley, T. L. Vagal afferent innervation of the rat fundic stomach: morphological characterization of the gastric tension receptor. J. Comp. Neurol. 319, 261–76 (1992).

67. Phillips, R. J. & Powley, T. L. Tension and stretch receptors in gastrointestinal smooth muscle: re-evaluating vagal mechanoreceptor electrophysiology. Brain Res. Brain Res. Rev. 34, 1–26 (2000).

68. Ritter, R. C. Gastrointestinal mechanisms of satiation for food. Physiol. Behav. 81, 249–73 (2004).

69. Duca, F. a & Lam, T. K. T. Gut microbiota, nutrient sensing and energy balance. Diabetes. Obes. Metab. 16 Suppl 1, 68–76 (2014).

70. Schwartz, M. W. et al. Cooperation between brain and islet in glucose homeostasis and diabetes. Nature 503, 59–66 (2013).

71. Raybould, H. E. Capsaicin-sensitive vagal afferents and CCK in inhibition of gastric motor function induced by intestinal nutrients. Peptides 12, 1279–83 (1991).

72. Müller, T. D. & Tschöp, M. H. Ghrelin - a key pleiotropic hormone-regulating systemic energy metabolism. Endocr. Dev. 25, 91–100 (2013).

73. Kojima, M. et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402, 656–60 (1999).

74. Yabuki, A. et al. Characterization and species differences in gastric ghrelin cells from mice, rats and hamsters. J. Anat. 205, 239–46 (2004).

Page 182: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

169

75. Date, Y. et al. Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology 141, 4255–61 (2000).

76. Zhu, X., Cao, Y., Voogd, K., Voodg, K. & Steiner, D. F. On the processing of proghrelin to ghrelin. J. Biol. Chem. 281, 38867–70 (2006).

77. Pemberton, C. J. & Richards, A. M. Biochemistry of ghrelin precursor peptides. Vitam. Horm. 77, 13–30 (2008).

78. Yang, J., Brown, M. S., Liang, G., Grishin, N. V & Goldstein, J. L. Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell 132, 387–96 (2008).

79. Murakami, N. et al. Role for central ghrelin in food intake and secretion profile of stomach ghrelin in rats. J. Endocrinol. 174, 283–8 (2002).

80. Bednarek, M. A. et al. Structure-function studies on the new growth hormone-releasing peptide, ghrelin: minimal sequence of ghrelin necessary for activation of growth hormone secretagogue receptor 1a. J. Med. Chem. 43, 4370–6 (2000).

81. Heppner, K. M. et al. Both Acyl and Des-Acyl Ghrelin Regulate Adiposity and Glucose Metabolism via Central Nervous System Ghrelin Receptors. Diabetes 63, 122–31 (2014).

82. Monteleone, P., Bencivenga, R., Longobardi, N., Serritella, C. & Maj, M. Differential responses of circulating ghrelin to high-fat or high-carbohydrate meal in healthy women. J. Clin. Endocrinol. Metab. 88, 5510–4 (2003).

83. Overduin, J., Frayo, R. S., Grill, H. J., Kaplan, J. M. & Cummings, D. E. Role of the duodenum and macronutrient type in ghrelin regulation. Endocrinology 146, 845–50 (2005).

84. Drazen, D. L., Vahl, T. P., D’Alessio, D. A., Seeley, R. J. & Woods, S. C. Effects of a fixed meal pattern on ghrelin secretion: evidence for a learned response independent of nutrient status. Endocrinology 147, 23–30 (2006).

85. Cummings, D. E. et al. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50, 1714–9 (2001).

86. Tschöp, M., Smiley, D. L. & Heiman, M. L. Ghrelin induces adiposity in rodents. Nature 407, 908–13 (2000).

87. Wren, A. M. et al. Ghrelin enhances appetite and increases food intake in humans. J. Clin. Endocrinol. Metab. 86, 5992 (2001).

88. Wren, A. M. et al. Ghrelin causes hyperphagia and obesity in rats. Diabetes 50, 2540–7 (2001).

Page 183: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

170

89. Asakawa, A. et al. Antagonism of ghrelin receptor reduces food intake and body weight gain in mice. Gut 52, 947–52 (2003).

90. Nakazato, M. et al. A role for ghrelin in the central regulation of feeding. Nature 409, 194–8 (2001).

91. Cowley, M. A. et al. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37, 649–61 (2003).

92. Currie, P. J., Mirza, A., Fuld, R., Park, D. & Vasselli, J. R. Ghrelin is an orexigenic and metabolic signaling peptide in the arcuate and paraventricular nuclei. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289, R353–R358 (2005).

93. Lawrence, C. B., Snape, A. C., Baudoin, F. M.-H. & Luckman, S. M. Acute central ghrelin and GH secretagogues induce feeding and activate brain appetite centers. Endocrinology 143, 155–62 (2002).

94. Takayama, K. et al. Expression of c-Fos protein in the brain after intravenous injection of ghrelin in rats. Neurosci. Lett. 417, 292–6 (2007).

95. Chen, W. & Enriori, P. J. Ghrelin  : a journey from GH secretagogue to regulator of metabolism. 4, 14–27 (2015).

96. Date, Y. et al. The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology 123, 1120–8 (2002).

97. Le Roux, C. W. et al. Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy. J. Clin. Endocrinol. Metab. 90, 4521–4 (2005).

98. Burdyga, G., Varro, A., Dimaline, R., Thompson, D. G. & Dockray, G. J. Ghrelin receptors in rat and human nodose ganglia: putative role in regulating CB-1 and MCH receptor abundance. Am. J. Physiol. Gastrointest. Liver Physiol. 290, G1289–97 (2006).

99. De Lartigue, G., Dimaline, R., Varro, A. & Dockray, G. J. Cocaine- and amphetamine-regulated transcript: stimulation of expression in rat vagal afferent neurons by cholecystokinin and suppression by ghrelin. J. Neurosci. 27, 2876–82 (2007).

100. Reimer, M. K., Pacini, G. & Ahrén, B. Dose-dependent inhibition by ghrelin of insulin secretion in the mouse. Endocrinology 144, 916–21 (2003).

101. Dezaki, K., Kakei, M. & Yada, T. Ghrelin uses Galphai2 and activates voltage-dependent K+ channels to attenuate glucose-induced Ca2+ signaling and insulin release in islet beta-cells: novel signal transduction of ghrelin. Diabetes 56, 2319–27 (2007).

102. Dezaki, K. et al. Blockade of pancreatic islet-derived ghrelin enhances insulin secretion to prevent high-fat diet-induced glucose intolerance. Diabetes 55, 3486–93 (2006).

Page 184: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

171

103. Gnanapavan, S. et al. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J. Clin. Endocrinol. Metab. 87, 2988 (2002).

104. Volante, M. et al. Expression of ghrelin and of the GH secretagogue receptor by pancreatic islet cells and related endocrine tumors. J. Clin. Endocrinol. Metab. 87, 1300–8 (2002).

105. Date, Y. et al. Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes 51, 124–9 (2002).

106. Wierup, N., Svensson, H., Mulder, H. & Sundler, F. The ghrelin cell: a novel developmentally regulated islet cell in the human pancreas. Regul. Pept. 107, 63–9 (2002).

107. Wierup, N., Yang, S., McEvilly, R. J., Mulder, H. & Sundler, F. Ghrelin is expressed in a novel endocrine cell type in developing rat islets and inhibits insulin secretion from INS-1 (832/13) cells. J. Histochem. Cytochem. 52, 301–10 (2004).

108. Tong, J. et al. Ghrelin suppresses glucose-stimulated insulin secretion and deteriorates glucose tolerance in healthy humans. Diabetes 59, 2145–51 (2010).

109. Stark, R. et al. Acyl ghrelin acts in the brain to control liver function and peripheral glucose homeostasis in male mice. Endocrinology en20141733 (2014).

110. Salehi, A., Dornonville de la Cour, C., Håkanson, R. & Lundquist, I. Effects of ghrelin on insulin and glucagon secretion: a study of isolated pancreatic islets and intact mice. Regul. Pept. 118, 143–50 (2004).

111. Chuang, J.-C. et al. Ghrelin directly stimulates glucagon secretion from pancreatic alpha-cells. Mol. Endocrinol. 25, 1600–11 (2011).

112. Fang, P. et al. Endogenous peptides as risk markers to assess the development of insulin resistance. Peptides 51, 9–14 (2014).

113. Vestergaard, E. T. et al. Acute peripheral metabolic effects of intraarterial ghrelin infusion in healthy young men. J. Clin. Endocrinol. Metab. 96, 468–77 (2011).

114. Vestergaard, E. T. et al. Ghrelin infusion in humans induces acute insulin resistance and lipolysis independent of growth hormone signaling. Diabetes 57, 3205–10 (2008).

115. Heijboer, A. C. et al. Ghrelin differentially affects hepatic and peripheral insulin sensitivity in mice. Diabetologia 49, 732–8 (2006).

116. Broglio, F. et al. Ghrelin and the endocrine pancreas. Endocrine 22, 19–24 (2003).

117. Ikezaki, A. et al. Fasting plasma ghrelin levels are negatively correlated with insulin resistance and PAI-1, but not with leptin, in obese children and adolescents. Diabetes 51, 3408–11 (2002).

Page 185: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

172

118. Katsuki, A. et al. Circulating levels of active ghrelin is associated with abdominal adiposity, hyperinsulinemia and insulin resistance in patients with type 2 diabetes mellitus. Eur. J. Endocrinol. 151, 573–7 (2004).

119. Pöykkö, S. M. et al. Low plasma ghrelin is associated with insulin resistance, hypertension, and the prevalence of type 2 diabetes. Diabetes 52, 2546–53 (2003).

120. Vestergaard, E. T. et al. Ghrelin- and GH-induced insulin resistance: no association with retinol-binding protein-4. Endocr. Connect. 2, 96–103 (2013).

121. Amini, P. et al. Serum acylated ghrelin is negatively correlated with the insulin resistance in the CODING study. PLoS One 7, e45657 (2012).

122. Levin, F. et al. Ghrelin stimulates gastric emptying and hunger in normal-weight humans. J. Clin. Endocrinol. Metab. 91, 3296–302 (2006).

123. Falkén, Y. et al. Actions of prolonged ghrelin infusion on gastrointestinal transit and glucose homeostasis in humans. Neurogastroenterol. Motil. 22, e192–200 (2010).

124. Fukuda, H. et al. Ghrelin enhances gastric motility through direct stimulation of intrinsic neural pathways and capsaicin-sensitive afferent neurones in rats. Scand. J. Gastroenterol. 39, 1209–14 (2004).

125. Zhang, Y. et al. Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–432 (1994).

126. Considine, R. V et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N.Engl.J.Med. 334, 292–295 (1996).

127. Bado, a et al. The stomach is a source of leptin. Nature 394, 790–3 (1998).

128. Masuzaki, H. et al. Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nat. Med. 3, 1029–33 (1997).

129. Wang, J., Liu, R., Hawkins, M., Barzilai, N. & Rossetti, L. A nutrient-sensing pathway regulates leptin gene expression in muscle and fat. Nature 393, 684–8 (1998).

130. Casabiell, X. et al. Presence of leptin in colostrum and/or breast milk from lactating mothers: a potential role in the regulation of neonatal food intake. J. Clin. Endocrinol. Metab. 82, 4270–3 (1997).

131. Nakashima, K., Narazaki, M. & Taga, T. Leptin receptor (OB-R) oligomerizes with itself but not with its closely related cytokine signal transducer gp130. FEBS Lett. 403, 79–82 (1997).

132. Schulz, L. C. & Widmaier, E. P. Leptin Receptors. 11–31 (Springer US, 2007).

Page 186: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

173

133. Chua, S. C. et al. Fine structure of the murine leptin receptor gene: splice site suppression is required to form two alternatively spliced transcripts. Genomics 45, 264–70 (1997).

134. Maamra, M. et al. Generation of human soluble leptin receptor by proteolytic cleavage of membrane-anchored receptors. Endocrinology 142, 4389–93 (2001).

135. Ge, H., Huang, L., Pourbahrami, T. & Li, C. Generation of soluble leptin receptor by ectodomain shedding of membrane-spanning receptors in vitro and in vivo. J. Biol. Chem. 277, 45898–903 (2002).

136. Banks, A. S., Davis, S. M., Bates, S. H. & Myers Jr., M. G. Activation of downstream signals by the long form of the leptin receptor. J.Biol.Chem. 275, 14563–14572 (2000).

137. Niswender, K. D. et al. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature 413, 794–795 (2001).

138. Li, Z., Ceccarini, G., Eisenstein, M., Tan, K. & Friedman, J. M. Phenotypic effects of an induced mutation of the ObRa isoform of the leptin receptor. Mol. Metab. 2, 364–75 (2013).

139. Cinti, S. et al. Secretory granules of endocrine and chief cells of human stomach mucosa contain leptin. Int. J. Obes. Relat. Metab. Disord. 24, 789–93 (2000).

140. Cammisotto, P. G. et al. Endocrine and exocrine secretion of leptin by the gastric mucosa. J. Histochem. Cytochem. 53, 851–60 (2005).

141. Oliver, P., Picó, C., De Matteis, R., Cinti, S. & Palou, A. Perinatal expression of leptin in rat stomach. Dev. Dyn. 223, 148–54 (2002).

142. Sobhani, I. et al. Leptin secretion and leptin receptor in the human stomach. Gut 47, 178–83 (2000).

143. Guilmeau, S., Buyse, M., Tsocas, A. & Bado, A. Duodenal Leptin Stimulates Cholecystokinin Secretion. Diabetes 52, (2003).

144. Cammisotto, P. & Bendayan, M. A review on gastric leptin: the exocrine secretion of a gastric hormone. Anat. Cell Biol. 45, 1–16 (2012).

145. Zhang, J. & Scarpace, P. J. The soluble leptin receptor neutralizes leptin-mediated STAT3 signalling and anorexic responses in vivo. Br. J. Pharmacol. 158, 475–82 (2009).

146. Yang, G., Ge, H., Boucher, A., Yu, X. & Li, C. Modulation of direct leptin signaling by soluble leptin receptor. Mol. Endocrinol. 18, 1354–62 (2004).

147. Kastin, A. J., Pan, W., Maness, L. M., Koletsky, R. J. & Ernsberger, P. Decreased transport of leptin across the blood-brain barrier in rats lacking the short form of the leptin receptor. Peptides 20, 1449–53 (1999).

Page 187: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

174

148. Mix, H. et al. Expression of leptin and leptin receptor isoforms in the human stomach. Gut 47, 481–6 (2000).

149. Buyse, M. et al. Expression and regulation of leptin receptor proteins in afferent and efferent neurons of the vagus nerve. Eur.J.Neurosci. 14, 64–72 (2001).

150. Barrenetxe, J. et al. Distribution of the long leptin receptor isoform in brush border, basolateral membrane, and cytoplasm of enterocytes. Gut 50, 797–802 (2002).

151. Morton, N. M., Emilsson, V., Liu, Y. L. & Cawthorne, M. a. Leptin action in intestinal cells. J. Biol. Chem. 273, 26194–201 (1998).

152. De Lartigue, G., Ronveaux, C. C. & Raybould, H. E. Deletion of leptin signaling in vagal afferent neurons results in hyperphagia and obesity. Mol. Metab. 3, 595–607 (2014).

153. Rajala, M. W. et al. Leptin acts independently of food intake to modulate gut microbial composition in male mice. Endocrinology 748–757 (2014).

154. Tavernier, A. et al. Intestinal deletion of leptin signaling alters activity of nutrient transporters and delayed the onset of obesity in mice. FASEB J. 28, 4100–4110 (2014).

155. Patterson, L. M., Zheng, H. & Berthoud, H. Vagal Afferents Innervating the Gastrointestinal Tract and CCKA-. 20, 10–20 (2002).

156. De Lartigue, G. et al. EGR1 Is a target for cooperative interactions between cholecystokinin and leptin, and inhibition by ghrelin, in vagal afferent neurons. Endocrinology 151, 3589–99 (2010).

157. Heldsinger, A., Grabauskas, G., Song, I. & Owyang, C. Synergistic interaction between leptin and cholecystokinin in the rat nodose ganglia is mediated by PI3K and STAT3 signaling pathways: implications for leptin as a regulator of short term satiety. J. Biol. Chem. 286, 11707–15 (2011).

158. Morton, G. J. & Schwartz, M. W. Leptin and the Central Nervous System Control of Glucose Metabolism. Physiol. Rev. 91, 389–411 (2011).

159. Cakir, B., Kasimay, O., Devseren, E. & Yeğen, B. C. Leptin inhibits gastric emptying in rats: role of CCK receptors and vagal afferent fibers. Physiol. Res. 56, 315–22 (2007).

160. Anini, Y. & Brubaker, P. L. Role of leptin in the regulation of glucagon-like peptide-1 secretion. Diabetes 52, 252–259 (2003).

161. Ducroc, R. et al. Luminal leptin induces rapid inhibition of active intestinal absorption of glucose mediated by sodium-glucose cotransporter 1. Diabetes 54, 348–354 (2005).

162. Ivy, A. & Oldberg, E. A hormone mechanism for gall-bladder contraction and evacuation. Am. J. Physiol. 86, 599–613 (1928).

Page 188: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

175

163. Moran, T. H. & Kinzig, K. P. Gastrointestinal satiety signals II. Cholecystokinin. Am.J.Physiol Gastrointest.Liver Physiol 286, G183–G188 (2004).

164. Rehfeld, J. F., Friis-Hansen, L., Goetze, J. P. & Hansen, T. V. The biology of cholecystokinin and gastrin peptides. Curr.Top.Med.Chem. 7, 1154–1165 (2007).

165. Little, T. J. et al. The release of GLP-1 and ghrelin, but not GIP and CCK, by glucose is dependent upon the length of small intestine exposed. Am.J.Physiol Endocrinol.Metab 291, E647–E655 (2006).

166. Baum, F. et al. Role of endogenously released cholecystokinin in determining postprandial insulin levels in man: effects of loxiglumide, a specific cholecystokinin receptor antagonist. Digestion 53, 189–99 (1992).

167. Matzinger, D. et al. Inhibition of food intake in response to intestinal lipid is mediated by cholecystokinin in humans. Am.J.Physiol 277, R1718–R1724 (1999).

168. Pilichiewicz, A. N. et al. Load-dependent effects of duodenal glucose on glycemia, gastrointestinal hormones, antropyloroduodenal motility, and energy intake in healthy men. Am. J. Physiol. Endocrinol. Metab. 293, E743–53 (2007).

169. Feltrin, K. L. et al. Effects of intraduodenal fatty acids on appetite, antropyloroduodenal motility, and plasma CCK and GLP-1 in humans vary with their chain length. Am. J. Physiol. Regul. Integr. Comp. Physiol. 287, R524–33 (2004).

170. Feltrin, K. L. et al. Comparative effects of intraduodenal infusions of lauric and oleic acids on antropyloroduodenal motility, plasma cholecystokinin and peptide YY, appetite, and energy intake in healthy men. Am. J. Clin. Nutr. 87, 1181–7 (2008).

171. Little, T. J. et al. Dose-related effects of lauric acid on antropyloroduodenal motility, gastrointestinal hormone release, appetite, and energy intake in healthy men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289, R1090–8 (2005).

172. Yox, D. P., Brenner, L. & Ritter, R. C. CCK-receptor antagonists attenuate suppression of sham feeding by intestinal nutrients. Am. J. Physiol. 262, R554–61 (1992).

173. Nawrot-Porabka, K. et al. Involvement of vagal nerves in the pancreatostimulatory effects of luminal melatonin, or its precursor L-tryptophan. Study in the rats. J. Physiol. Pharmacol. 58 Suppl 6, 81–95 (2007).

174. Nassir, F., Wilson, B., Han, X., Gross, R. W. & Abumrad, N. A. CD36 is important for fatty acid and cholesterol uptake by the proximal but not distal intestine. J.Biol.Chem. 282, 19493–19501 (2007).

175. Liou, A. P. et al. The G-protein-coupled receptor GPR40 directly mediates long-chain fatty acid-induced secretion of cholecystokinin. Gastroenterology 140, 903–912 (2011).

Page 189: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

176

176. Chang, C. H., Chey, W. Y. & Chang, T. M. Cellular mechanism of sodium oleate-stimulated secretion of cholecystokinin and secretin. Am.J.Physiol Gastrointest.Liver Physiol 279, G295–G303 (2000).

177. Uchida, T. et al. Protein kinase Cdelta plays a non-redundant role in insulin secretion in pancreatic beta cells. J.Biol.Chem. 282, 2707–2716 (2007).

178. Nemoz-Gaillard, E. et al. Expression of SNARE proteins in enteroendocrine cell lines and functional role of tetanus toxin-sensitive proteins in cholecystokinin release. FEBS Lett. 425, 66–70 (1998).

179. Chandra, R. et al. Immunoglobulin-like domain containing receptor 1 mediates fat-stimulated cholecystokinin secretion. J. Clin. Invest. 123, 3343–52 (2013).

180. Liddle, R. A., Morita, E. T., Conrad, C. K. & Williams, J. A. Regulation of gastric emptying in humans by cholecystokinin. J. Clin. Invest. 77, 992–6 (1986).

181. Liddle, R. A. Integrated actions of cholecystokinin on the gastrointestinal tract: use of the cholecystokinin bioassay. Gastroenterol. Clin. North Am. 18, 735–56 (1989).

182. Fraser, R., Fone, D., Horowitz, M. & Dent, J. Cholecystokinin octapeptide stimulates phasic and tonic pyloric motility in healthy humans. Gut 34, 33–7 (1993).

183. Muurahainen, N., Kissileff, H. R., Derogatis, A. J. & Pi-Sunyer, F. X. Effects of cholecystokinin-octapeptide (CCK-8) on food intake and gastric emptying in man. Physiol. Behav. 44, 645–9 (1988).

184. Chandra, R. & Liddle, R. A. Cholecystokinin. Curr.Opin.Endocrinol.Diabetes Obes. 14, 63–67 (2007).

185. Cox, J. E. Effect of pyloric cuffs on cholecystokinin satiety. Physiol Behav. 60, 1023–1026 (1996).

186. Gibbs, J., Young, R. C. & Smith, G. P. Cholecystokinin elicits satiety in rats with open gastric fistulas. Nature 245, 323–5 (1973).

187. Kissileff, H. R., Pi-Sunyer, F. X., Thornton, J. & Smith, G. P. C-terminal octapeptide of cholecystokinin decreases food intake in man. Am.J.Clin.Nutr. 34, 154–160 (1981).

188. Kraly, F. S., Carty, W. J., Resnick, S. & Smith, G. P. Effect of cholecystokinin on meal size and intermeal interval in the sham-feeding rat. J.Comp Physiol Psychol. 92, 697–707 (1978).

189. Moran, T. H., Ameglio, P. J., Schwartz, G. J. & McHugh, P. R. Blockade of type A, not type B, CCK receptors attenuates satiety actions of exogenous and endogenous CCK. Am.J.Physiol 262, R46–R50 (1992).

Page 190: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

177

190. Weatherford, S. C., Chiruzzo, F. Y. & Laughton, W. B. Satiety induced by endogenous and exogenous cholecystokinin is mediated by CCK-A receptors in mice. Am.J.Physiol 262, R574–R578 (1992).

191. Woltman, T., Castellanos, D. & Reidelberger, R. Role of cholecystokinin in the anorexia produced by duodenal delivery of oleic acid in rats. Am.J.Physiol 269, R1420–R1433 (1995).

192. Gutzwiller, J. P., Degen, L., Matzinger, D., Prestin, S. & Beglinger, C. Interaction between GLP-1 and CCK-33 in inhibiting food intake and appetite in men. Am.J.Physiol Regul.Integr.Comp Physiol 287, R562–R567 (2004).

193. Burton-Freeman, B., Davis, P. A. & Schneeman, B. O. Plasma cholecystokinin is associated with subjective measures of satiety in women. Am.J.Clin.Nutr. 76, 659–667 (2002).

194. Smith, G. P., Jerome, C., Cushin, B. J., Eterno, R. & Simansky, K. J. Abdominal vagotomy blocks the satiety effect of cholecystokinin in the rat. Science 213, 1036–7 (1981).

195. Monnikes, H. et al. Pathways of Fos expression in locus ceruleus, dorsal vagal complex, and PVN in response to intestinal lipid. Am.J.Physiol 273, R2059–R2071 (1997).

196. Greenberg, D., Smith, G. P. & Gibbs, J. Intraduodenal infusions of fats elicit satiety in sham-feeding rats. Am.J.Physiol 259, R110–R118 (1990).

197. Matzinger, D. et al. The role of long chain fatty acids in regulating food intake and cholecystokinin release in humans. Gut 46, 688–693 (2000).

198. Lieverse, R. J., Jansen, J. B., Masclee, A. A., Rovati, L. C. & Lamers, C. B. Effect of a low dose of intraduodenal fat on satiety in humans: studies using the type A cholecystokinin receptor antagonist loxiglumide. Gut 35, 501–505 (1994).

199. Schwartz, G. J., Whitney, A., Skoglund, C., Castonguay, T. W. & Moran, T. H. Decreased responsiveness to dietary fat in Otsuka Long-Evans Tokushima fatty rats lacking CCK-A receptors. Am.J.Physiol 277, R1144–R1151 (1999).

200. Webster, W. & Beyak, M. The long chain fatty acid oleate activates mouse intestinal afferent nerves in vitro. Can. J. Physiol. Pharmacol. 379, 375–379 (2013).

201. Brown, T. A., Washington, M. C., Metcalf, S. A. & Sayegh, A. I. The feeding responses evoked by cholecystokinin are mediated by vagus and splanchnic nerves. Peptides 32, 1581–1586 (2011).

202. Wright, J. et al. Reduction of food intake by cholecystokinin requires activation of hindbrain NMDA-type glutamate receptors. Am.J.Physiol Regul.Integr.Comp Physiol 301, R448–R455 (2011).

Page 191: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

178

203. Campos, C. A., Wright, J. S., Czaja, K. & Ritter, R. C. CCK-induced reduction of food intake and hindbrain MAPK signaling are mediated by NMDA receptor activation. Endocrinology 153, 2633–46 (2012).

204. Lateef, D. M., Washington, M. C. & Sayegh, A. I. The short term satiety peptide cholecystokinin reduces meal size and prolongs intermeal interval. Peptides 32, 1289–95 (2011).

205. Washington, M. C., Coggeshall, J. & Sayegh, A. I. Cholecystokinin-33 inhibits meal size and prolongs the subsequent intermeal interval. Peptides 32, 971–7 (2011).

206. Blevins, J. E., Stanley, B. G. & Reidelberger, R. D. Brain regions where cholecystokinin suppresses feeding in rats. Brain Res. 860, 1–10 (2000).

207. Zhu, G., Yan, J., Smith, W. W., Moran, T. H. & Bi, S. Roles of dorsomedial hypothalamic cholecystokinin signaling in the controls of meal patterns and glucose homeostasis. Physiol. Behav. 105, 234–41 (2012).

208. Edwards, G. L., Ladenheim, E. E. & Ritter, R. C. Dorsomedial hindbrain participation in cholecystokinin-induced satiety. Am.J.Physiol 251, R971–R977 (1986).

209. Mccown, M., Tyler, J. & Cox, J. E. Inhibition superior of sucrose intake by continuous celiac , and intravenous CCK-8 infusions. Am J Physiol Regul Integr Comp Physiol 270, R319–R325 (1996).

210. Kimura, I., Nakashima, N., Komori, T., Kameda, Y. & Kimura, M. Dependence of cholecystokinin-8-stimulated insulin release on high glucose levels is evidenced by pseudo-alpha-D-glucose in rat pancreas and islets. Jpn. J. Pharmacol. 64, 103–7 (1994).

211. Malm, D., Giaever, A., Vonen, B. & Florholmen, J. Cholecystokinin and somatostatin modulate the glucose-induced insulin secretion by different mechanisms in pancreatic islets. A study on phospholipase C activity and calcium requirement. Scand. J. Clin. Lab. Invest. 53, 671–6 (1993).

212. Otsuki, M. et al. Action of cholecystokinin analogues on exocrine and endocrine rat pancreas. Am. J. Physiol. 250, G405–11 (1986).

213. Sakamoto, C. et al. Glucose-dependent insulinotropic action of cholecystokinin and caerulein in the isolated perfused rat pancreas. Endocrinology 110, 398–402 (1982).

214. Schmid, R., Schusdziarra, V., Schulte-Frohlinde, E., Maier, V. & Classen, M. Effect of CCK on insulin, glucagon, and pancreatic polypeptide levels in humans. Pancreas 4, 653–61 (1989).

215. Shikado, F., Miyasaka, K., Funakoshi, A. & Kitani, K. Necessity of hyperglycemia for effects of endogenous cholecystokinin on insulin and pancreatic exocrine secretion in conscious rats. Jpn. J. Physiol. 40, 383–91 (1990).

Page 192: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

179

216. Verspohl, E. J., Ammon, H. P., Williams, J. A. & Goldfine, I. D. Evidence that cholecystokinin interacts with specific receptors and regulates insulin release in isolated rat islets of Langerhans. Diabetes 35, 38–43 (1986).

217. Verspohl, E. J., Breuning, I., Ammon, H. P. & Mark, M. Significance of Ca2+, Rb+ fluxes, of cAMP and cGMP for the CCK8-modulated insulin release. Regul. Pept. 17, 229–41 (1987).

218. Zawalich, W., Takuwa, N., Takuwa, Y., Diaz, V. A. & Rasmussen, H. Interactions of cholecystokinin and glucose in rat pancreatic islets. Diabetes 36, 426–33 (1987).

219. Zawalich, W. S., Cote, S. B. & Diaz, V. A. Influence of cholecystokinin on insulin output from isolated perifused pancreatic islets. Endocrinology 119, 616–21 (1986).

220. Jensen, S. L. et al. Secretory effects of cholecystokinins on the isolated perfused porcine pancreas. Acta Physiol. Scand. 111, 225–31 (1981).

221. Jensen, S. L., Holst, J. J., Nielsen, O. V & Rehfeld, J. F. Effect of sulfation of CCK-8 on its stimulation of the endocrine and exocrine secretion from the isolated perfused porcine pancreas. Digestion 22, 305–9 (1981).

222. Reimers, J. et al. Lack of insulinotropic effect of endogenous and exogenous cholecystokinin in man. Diabetologia 31, 271–80 (1988).

223. Rushakoff, R. J., Goldfine, I. D., Carter, J. D. & Liddle, R. A. Physiological concentrations of cholecystokinin stimulate amino acid-induced insulin release in humans. J. Clin. Endocrinol. Metab. 65, 395–401 (1987).

224. Liddle, R. A. et al. Physiological role for cholecystokinin in reducing postprandial hyperglycemia in humans. J. Clin. Invest. 81, 1675–81 (1988).

225. Balkan, B., Steffens, A. B., Strubbe, J. H. & Bruggink, J. E. Biphasic insulin secretion after intravenous but not after intraportal CCK-8 infusion in rats. Diabetes 39, 702–6 (1990).

226. Lo, C.-M. et al. Impaired insulin secretion and enhanced insulin sensitivity in cholecystokinin-deficient mice. Diabetes 60, 2000–7 (2011).

227. Karlsson, S. & Ahrén, B. CCKA receptor antagonism inhibits mechanisms underlying CCK-8-stimulated insulin release in isolated rat islets. Eur. J. Pharmacol. 202, 253–7 (1991).

228. Sakamoto, C., Goldfine, I. D., Roach, E. & Williams, J. A. Localization of Saturable CCK Binding Sites in Rat Pancreatic Islets by Light and Electron Microscope Autoradiography. Diabetes 34, 390–394 (1985).

229. Verspohl, E. J. & Herrmann, K. Involvement of G proteins in the effect of carbachol and cholecystokinin in rat pancreatic islets. Am. J. Physiol. 271, E65–72 (1996).

Page 193: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

180

230. Karlsson, S. & Ahrén, B. Cholecystokinin-stimulated insulin secretion and protein kinase C in rat pancreatic islets. Acta Physiol. Scand. 142, 397–403 (1991).

231. Simonsson, E., Karlsson, S. & Ahrén, B. Involvement of phospholipase A2 and arachidonic acid in cholecystokinin-8-induced insulin secretion in rat islets. Regul. Pept. 65, 101–7 (1996).

232. Simonsson, E., Karlsson, S. & Ahrén, B. Ca2+-independent phospholipase A2 contributes to the insulinotropic action of cholecystokinin-8 in rat islets: dissociation from the mechanism of carbachol. Diabetes 47, 1436–43 (1998).

233. Borovicka, J. et al. Role of cholecystokinin as a regulator of solid and liquid gastric emptying in humans. Am. J. Physiol. 271, G448–53 (1996).

234. Fried, M. et al. Physiological role of cholecystokinin on postprandial insulin secretion and gastric meal emptying in man. Studies with the cholecystokinin receptor antagonist loxiglumide. Diabetologia 34, 721–6 (1991).

235. Fried, M. et al. Role of cholecystokinin in the regulation of gastric emptying and pancreatic enzyme secretion in humans. Studies with the cholecystokinin-receptor antagonist loxiglumide. Gastroenterology 101, 503–11 (1991).

236. Hidalgo, L. et al. Effect of cholecystokinin-A receptor blockade on postprandial insulinaemia and gastric emptying in humans. Neurogastroenterol. Motil. 14, 519–25 (2002).

237. Moran, T. H., Kornbluh, R., Moore, K. & Schwartz, G. J. Cholecystokinin inhibits gastric emptying and contracts the pyloric sphincter in rats by interacting with low affinity CCK receptor sites. Regul. Pept. 52, 165–72 (1994).

238. Schwartz, G. J., Berkow, G., McHugh, P. R. & Moran, T. H. Gastric branch vagotomy blocks nutrient and cholecystokinin-induced suppression of gastric emptying. Am. J. Physiol. 264, R630–7 (1993).

239. Rayner, C. K., Park, H. S., Doran, S. M., Chapman, I. M. & Horowitz, M. Effects of cholecystokinin on appetite and pyloric motility during physiological hyperglycemia. Am. J. Physiol. Gastrointest. Liver Physiol. 278, G98–G104 (2000).

240. Takeda, J. et al. Sequence of an intestinal cDNA encoding human gastric inhibitory polypeptide precursor. Proc. Natl. Acad. Sci. U. S. A. 84, 7005–8 (1987).

241. Brown, J. C., Dryburgh, J. R., Ross, S. A. & Dupré, J. Identification and actions of gastric inhibitory polypeptide. Recent Prog. Horm. Res. 31, 487–532 (1975).

242. Thomas, F. B. et al. Localization of gastric inhibitory polypeptide release by intestinal glucose perfusion in man. Gastroenterology 72, 49–54 (1977).

Page 194: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

181

243. Pilichiewicz, A. N. et al. Load-dependent effects of duodenal glucose on glycemia, gastrointestinal hormones, antropyloroduodenal motility, and energy intake in healthy men. Am.J.Physiol Endocrinol.Metab 293, E743–E753 (2007).

244. Chaikomin, R. et al. Initially more rapid small intestinal glucose delivery increases plasma insulin, GIP, and GLP-1 but does not improve overall glycemia in healthy subjects. Am. J. Physiol. Endocrinol. Metab. 289, E504–7 (2005).

245. Kuo, P. et al. Transient, early release of glucagon-like peptide-1 during low rates of intraduodenal glucose delivery. Regul. Pept. 146, 1–3 (2008).

246. Lavin, J. H. et al. Interaction of insulin, glucagon-like peptide 1, gastric inhibitory polypeptide, and appetite in response to intraduodenal carbohydrate. Am. J. Clin. Nutr. 68, 591–8 (1998).

247. McCullough, A. J., Miller, L. J., Service, F. J. & Go, V. L. Effect of graded intraduodenal glucose infusions on the release and physiological action of gastric inhibitory polypeptide. J. Clin. Endocrinol. Metab. 56, 234–41 (1983).

248. Lucey, M. R. et al. Response of circulating somatostatin, insulin, gastrin and GIP, to intraduodenal infusion of nutrients in normal man. Clin. Endocrinol. (Oxf). 21, 209–17 (1984).

249. O’Dorisio, T. M., Cataland, S., Stevenson, M. & Mazzaferri, E. L. Gastric inhibitory polypeptide (GIP). Intestinal distribution and stimulation by amino acids and medium-chain triglycerides. Am. J. Dig. Dis. 21, 761–5 (1976).

250. Cho, Y. M. & Kieffer, T. J. K-cells and Glucose-Dependent Insulinotropic Polypeptide in Health and Disease. Vitam. Horm. Incretins Insul. Secret. 84, 111–150 (Elsevier Inc., 2010).

251. Parker, H. E., Habib, A. M., Rogers, G. J., Gribble, F. M. & Reimann, F. Nutrient-dependent secretion of glucose-dependent insulinotropic polypeptide from primary murine K cells. Diabetologia 52, 289–98 (2009).

252. Gorboulev, V. et al. Na(+)-D-glucose cotransporter SGLT1 is pivotal for intestinal glucose absorption and glucose-dependent incretin secretion. Diabetes 61, 187–96 (2012).

253. Asmar, M. et al. On the role of glucose-dependent insulintropic polypeptide in postprandial metabolism in humans. Am. J. Physiol. Endocrinol. Metab. 298, E614–21 (2010).

254. Edholm, T. et al. Differential incretin effects of GIP and GLP-1 on gastric emptying, appetite, and insulin-glucose homeostasis. Neurogastroenterol. Motil. 22, 1191–2000 (2010).

Page 195: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

182

255. Szecówka, J., Lins, P. E. & Efendić, S. Effects of cholecystokinin, gastric inhibitory polypeptide, and secretin on insulin and glucagon secretion in rats. Endocrinology 110, 1268–72 (1982).

256. Elahi, D. et al. The insulinotropic actions of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (7-37) in normal and diabetic subjects. Regul. Pept. 51, 63–74 (1994).

257. Saxena, R. et al. Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat. Genet. 42, 142–8 (2010).

258. Flock, G., Holland, D., Seino, Y. & Drucker, D. J. GPR119 regulates murine glucose homeostasis through incretin receptor-dependent and independent mechanisms. Endocrinology 152, 374–83 (2011).

259. Lewis, J. T., Dayanandan, B., Habener, J. F. & Kieffer, T. J. Glucose-dependent insulinotropic polypeptide confers early phase insulin release to oral glucose in rats: demonstration by a receptor antagonist. Endocrinology 141, 3710–6 (2000).

260. Moens, K. et al. Expression and functional activity of glucagon, glucagon-like peptide I, and glucose-dependent insulinotropic peptide receptors in rat pancreatic islet cells. Diabetes 45, 257–61 (1996).

261. Usdin, T. B., Mezey, E., Button, D. C., Brownstein, M. J. & Bonner, T. I. Gastric inhibitory polypeptide receptor, a member of the secretin-vasoactive intestinal peptide receptor family, is widely distributed in peripheral organs and the brain. Endocrinology 133, 2861–70 (1993).

262. Kashima, Y. et al. Critical role of cAMP-GEFII--Rim2 complex in incretin-potentiated insulin secretion. J. Biol. Chem. 276, 46046–53 (2001).

263. Miki, T. et al. Distinct effects of glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 on insulin secretion and gut motility. Diabetes 54, 1056–63 (2005).

264. Lu, M., Wheeler, M. B., Leng, X. H. & Boyd, A. E. The role of the free cytosolic calcium level in beta-cell signal transduction by gastric inhibitory polypeptide and glucagon-like peptide I(7-37). Endocrinology 132, 94–100 (1993).

265. Meier, J. J. et al. Gastric inhibitory polypeptide (GIP) dose-dependently stimulates glucagon secretion in healthy human subjects at euglycaemia. Diabetologia 46, 798–801 (2003).

266. Christensen, M., Vedtofte, L., Holst, J. J., Vilsbøll, T. & Knop, F. K. Glucose-dependent insulinotropic polypeptide: a bifunctional glucose-dependent regulator of glucagon and insulin secretion in humans. Diabetes 60, 3103–9 (2011).

267. Meier, J. J. et al. Gastric inhibitory polypeptide does not inhibit gastric emptying in humans. Am. J. Physiol. Endocrinol. Metab. 286, E621–5 (2004).

Page 196: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

183

268. Ogawa, E. et al. The effect of gastric inhibitory polypeptide on intestinal glucose absorption and intestinal motility in mice. Biochem. Biophys. Res. Commun. 404, 115–120 (2011).

269. Singh, S. K. et al. Glucose-dependent insulinotropic polypeptide (GIP) stimulates transepithelial glucose transport. Obesity (Silver Spring). 16, 2412–6 (2008).

270. Nakagawa, A. et al. Receptor gene expression of glucagon-like peptide-1, but not glucose-dependent insulinotropic polypeptide, in rat nodose ganglion cells. Auton.Neurosci. 110, 36–43 (2004).

271. Theodorakis, M. J. et al. Human duodenal enteroendocrine cells: source of both incretin peptides, GLP-1 and GIP. Am.J.Physiol Endocrinol.Metab 290, E550–E559 (2006).

272. Larsen, P. J., Tang-Christensen, M., Holst, J. J. & Orskov, C. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience 77, 257–70 (1997).

273. Eissele, R. et al. Glucagon-like peptide-1 cells in the gastrointestinal tract and pancreas of rat, pig and man. Eur. J. Clin. Invest. 22, 283–91 (1992).

274. Holst, J. J., Orskov, C., Nielsen, O. V & Schwartz, T. W. Truncated glucagon-like peptide I, an insulin-releasing hormone from the distal gut. FEBS Lett. 211, 169–74 (1987).

275. Mojsov, S., Weir, G. C. & Habener, J. F. Insulinotropin: glucagon-like peptide I (7-37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J. Clin. Invest. 79, 616–9 (1987).

276. D’Alessio, D. et al. Fasting and postprandial concentrations of GLP-1 in intestinal lymph and portal plasma: evidence for selective release of GLP-1 in the lymph system. Am. J. Physiol. Regul. Integr. Comp. Physiol. 293, R2163–9 (2007).

277. Deacon, C. F., Pridal, L., Klarskov, L., Olesen, M. & Holst, J. J. Glucagon-like peptide 1 undergoes differential tissue-specific metabolism in the anesthetized pig. Am. J. Physiol. 271, E458–64 (1996).

278. Mentlein, R., Gallwitz, B. & Schmidt, W. E. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7-36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur. J. Biochem. 214, 829–35 (1993).

279. Elliott, R. M. et al. Glucagon-like peptide-1 (7-36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J. Endocrinol. 138, 159–66 (1993).

280. Dillon, J. S. et al. Cloning and functional expression of the human glucagon-like peptide-1 (GLP-1) receptor. Endocrinology 133, 1907–10 (1993).

Page 197: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

184

281. Richards, P. et al. Identification and Characterization of GLP-1 Receptor-Expressing Cells Using a New Transgenic Mouse Model. Diabetes 63, 1224–1233 (2014).

282. Rocca, A. S. & Brubaker, P. L. Role of the vagus nerve in mediating proximal nutrient-induced glucagon-like peptide-1 secretion. Endocrinology 140, 1687–94 (1999).

283. Vahl, T. P. et al. Glucagon-like peptide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein mediate the effects of endogenous GLP-1 on glucose tolerance in rats. Endocrinology 148, 4965–73 (2007).

284. Gaisano, G. G., Park, S. J., Daly, D. M. & Beyak, M. J. Glucagon-like peptide-1 inhibits voltage-gated potassium currents in mouse nodose ganglion neurons. Neurogastroenterol. Motil. 22, 470–9, e111 (2010).

285. Waget, A. et al. Physiological and pharmacological mechanisms through which the DPP-4 inhibitor sitagliptin regulates glycemia in mice. Endocrinology 152, 3018–29 (2011).

286. Jang, H.-J. et al. Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc. Natl. Acad. Sci. U. S. A. 104, 15069–74 (2007).

287. Shirazi-Beechey, S. P., Moran, A. W., Batchelor, D. J., Daly, K. & Al-Rammahi, M. Glucose sensing and signalling; regulation of intestinal glucose transport. Proc. Nutr. Soc. 70, 185–93 (2011).

288. Tolhurst, G., Reimann, F. & Gribble, F. M. Intestinal sensing of nutrients. Handb. Exp. Pharmacol. 309–35 (2012).

289. Beglinger, S. et al. Role of fat hydrolysis in regulating glucagon-like Peptide-1 secretion. J. Clin. Endocrinol. Metab. 95, 879–86 (2010).

290. Poreba, M. A. et al. Role of fatty acid transport protein 4 in oleic acid-induced glucagon-like peptide-1 secretion from murine intestinal L cells. Am. J. Physiol. Endocrinol. Metab. 303, E899–907 (2012).

291. Edfalk, S., Steneberg, P. & Edlund, H. Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes 57, 2280–7 (2008).

292. Hirasawa, A. et al. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat. Med. 11, 90–4 (2005).

293. Oya, M. et al. The G protein-coupled receptor family C group 6 subtype A (GPRC6A) receptor is involved in amino acid-induced glucagon-like peptide-1 secretion from GLUTag cells. J. Biol. Chem. 288, 4513–21 (2013).

294. Cani, P. D. et al. GLUT2 and the incretin receptors are involved in glucose-induced incretin secretion. Mol. Cell. Endocrinol. 276, 18–23 (2007).

Page 198: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

185

295. Parker, H. E. et al. Molecular mechanisms underlying bile acid-stimulated glucagon-like peptide-1 secretion. Br. J. Pharmacol. 165, 414–23 (2012).

296. Katsuma, S., Hirasawa, A. & Tsujimoto, G. Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem. Biophys. Res. Commun. 329, 386–90 (2005).

297. Bucinskaite, V. et al. Receptor-mediated activation of gastric vagal afferents by glucagon-like peptide-1 in the rat. Neurogastroenterol. Motil. 21, 978–e78 (2009).

298. Williams, D. L., Baskin, D. G. & Schwartz, M. W. Evidence that intestinal glucagon-like peptide-1 plays a physiological role in satiety. Endocrinology 150, 1680–7 (2009).

299. Abbott, C. R. et al. The inhibitory effects of peripheral administration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 1044, 127–31 (2005).

300. Sisley, S. et al. Neuronal GLP1R mediates liraglutide’s anorectic but not glucose-lowering effect. J. Clin. Invest. 124, 2456–2463 (2014).

301. Turton, M. D. et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 69–72 (1996).

302. Tang-Christensen, M. et al. Central administration of GLP-1-(7-36) amide inhibits food and water intake in rats. Am. J. Physiol. 271, R848–56 (1996).

303. Meeran, K. et al. Repeated intracerebroventricular administration of glucagon-like peptide-1-(7-36) amide or exendin-(9-39) alters body weight in the rat. Endocrinology 140, 244–50 (1999).

304. Rupprecht, L. E. et al. Hindbrain GLP-1 receptor-mediated suppression of food intake requires a PI3K-dependent decrease in phosphorylation of membrane-bound Akt. Am. J. Physiol. Endocrinol. Metab. 305, E751–9 (2013).

305. Hayes, M. R. et al. Intracellular signals mediating the food intake-suppressive effects of hindbrain glucagon-like peptide-1 receptor activation. Cell Metab. 13, 320–30 (2011).

306. Hayes, M. R. Neuronal and intracellular signaling pathways mediating GLP-1 energy balance and glycemic effects. Physiol. Behav. 106, 413–6 (2012).

307. Fehmann, H. C., Janssen, M. & Göke, B. Interaction of glucagon-like peptide-I (GLP-I) and galanin in insulin (beta TC-1)- and somatostatin (RIN T3)-secreting cells and evidence that both peptides have no receptors on glucagon (INR1G9)-secreting cells. Acta Diabetol. 32, 176–81 (1995).

308. MacDonald, P. E., Salapatek, A. M. F. & Wheeler, M. B. Glucagon-like peptide-1 receptor activation antagonizes voltage-dependent repolarizing K(+) currents in beta-

Page 199: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

186

cells: a possible glucose-dependent insulinotropic mechanism. Diabetes 51 Suppl 3, S443–7 (2002).

309. Light, P. E., Manning Fox, J. E., Riedel, M. J. & Wheeler, M. B. Glucagon-like peptide-1 inhibits pancreatic ATP-sensitive potassium channels via a protein kinase A- and ADP-dependent mechanism. Mol. Endocrinol. 16, 2135–44 (2002).

310. Suga, S. et al. cAMP-independent decrease of ATP-sensitive K+ channel activity by GLP-1 in rat pancreatic beta-cells. Pflugers Arch. 440, 566–72 (2000).

311. Suga, S., Kanno, T., Dobashi, Y. & Wakui, M. GLP-1 (7-36) amide activates L-type Ca2+ channels of pancreatic B-cells through c-AMP signaling. Jpn. J. Physiol. 47 Suppl 1, S13–4 (1997).

312. Ding, W. G., Kitasato, H. & Matsuura, H. Involvement of calmodulin in glucagon-like peptide 1(7-36) amide-induced inhibition of the ATP-sensitive K+ channel in mouse pancreatic beta-cells. Exp. Physiol. 86, 331–9 (2001).

313. Orskov, C., Holst, J. J. & Nielsen, O. V. Effect of truncated glucagon-like peptide-1 [proglucagon-(78-107) amide] on endocrine secretion from pig pancreas, antrum, and nonantral stomach. Endocrinology 123, 2009–13 (1988).

314. Hare, K. J. et al. The glucagonostatic and insulinotropic effects of glucagon-like peptide 1 contribute equally to its glucose-lowering action. Diabetes 59, 1765–70 (2010).

315. Hare, K. J. et al. Preserved inhibitory potency of GLP-1 on glucagon secretion in type 2 diabetes mellitus. J.Clin.Endocrinol.Metab 94, 4679–4687 (2009).

316. Ritzel, R., Orskov, C., Holst, J. J. & Nauck, M. A. Pharmacokinetic, insulinotropic, and glucagonostatic properties of GLP-1 [7-36 amide] after subcutaneous injection in healthy volunteers. Dose-response-relationships. Diabetologia 38, 720–5 (1995).

317. De Heer, J., Rasmussen, C., Coy, D. H. & Holst, J. J. Glucagon-like peptide-1, but not glucose-dependent insulinotropic peptide, inhibits glucagon secretion via somatostatin (receptor subtype 2) in the perfused rat pancreas. Diabetologia 51, 2263–70 (2008).

318. Tornehave, D., Kristensen, P., Rømer, J., Knudsen, L. B. & Heller, R. S. Expression of the GLP-1 receptor in mouse, rat, and human pancreas. J. Histochem. Cytochem. 56, 841–51 (2008).

319. Nauck, M. A. et al. Effects of glucagon-like peptide 1 on counterregulatory hormone responses, cognitive functions, and insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp experiments in healthy volunteers. J. Clin. Endocrinol. Metab. 87, 1239–46 (2002).

320. Burcelin, R., Da Costa, A., Drucker, D. J. & Thorens, B. Glucose Competence of the Hepatoportal Vein Sensor Requires the Presence of an Activated Glucagon-Like Peptide-1 Receptor. Diabetes 50, 1720–1728 (2001).

Page 200: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

187

321. Sandoval, D. A., Bagnol, D., Woods, S. C., D’Alessio, D. A. & Seeley, R. J. Arcuate glucagon-like peptide 1 receptors regulate glucose homeostasis but not food intake. Diabetes 57, 2046–2054 (2008).

322. Knauf, C. et al. Brain glucagon-like peptide – 1 increases insulin secretion and muscle insulin resistance to favor hepatic glycogen storage. J. Clin. Invest. 115, 17–20 (2005).

323. Nauck, M. A. et al. Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. Am. J. Physiol. 273, E981–8 (1997).

324. Brennan, I. M. et al. Evaluation of interactions between CCK and GLP-1 in their effects on appetite, energy intake, and antropyloroduodenal motility in healthy men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 288, R1477–85 (2005).

325. Schirra, J. et al. Effects of glucagon-like peptide-1(7-36)amide on antro-pyloro-duodenal motility in the interdigestive state and with duodenal lipid perfusion in humans. Gut 46, 622–31 (2000).

326. Schirra, J. et al. Endogenous glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-pyloro-duodenal motility in humans. Gut 55, 243–51 (2006).

327. Brubaker, P. L. et al. Circulating and tissue forms of the intestinal growth factor, glucagon-like peptide-2. Endocrinology 138, 4837–43 (1997).

328. Xiao, Q., Boushey, R. P., Drucker, D. J. & Brubaker, P. L. Secretion of the intestinotropic hormone glucagon-like peptide 2 is differentially regulated by nutrients in humans. Gastroenterology 117, 99–105 (1999).

329. Parker, H. E. et al. Predominant role of active versus facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia 55, 2445–55 (2012).

330. Thomas, C. et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 10, 167–77 (2009).

331. Orskov, C. et al. Glucagon-like peptides GLP-1 and GLP-2, predicted products of the glucagon gene, are secreted separately from pig small intestine but not pancreas. Endocrinology 119, 1467–75 (1986).

332. Drucker, D. J. & Yusta, B. Physiology and Pharmacology of the Enteroendocrine Hormone Glucagon-Like Peptide-2. Annu. Rev. Physiol. (2013).

333. Dong, C. X. & Brubaker, P. L. Ghrelin, the proglucagon-derived peptides and peptide YY in nutrient homeostasis. Nat. Rev. Gastroenterol. Hepatol. 9, 705–15 (2012).

334. Baldassano, S., Bellanca, A. L., Serio, R. & Mulè, F. Food intake in lean and obese mice after peripheral administration of glucagon-like peptide 2. J. Endocrinol. 213, 277–84 (2012).

Page 201: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

188

335. Schmidt, P. T. et al. Peripheral administration of GLP-2 to humans has no effect on gastric emptying or satiety. Regul. Pept. 116, 21–5 (2003).

336. Sørensen, L. B. et al. No effect of physiological concentrations of glucagon-like peptide-2 on appetite and energy intake in normal weight subjects. Int. J. Obes. Relat. Metab. Disord. 27, 450–6 (2003).

337. Lovshin, J., Estall, J., Yusta, B., Brown, T. J. & Drucker, D. J. Glucagon-like peptide (GLP)-2 action in the murine central nervous system is enhanced by elimination of GLP-1 receptor signaling. J. Biol. Chem. 276, 21489–99 (2001).

338. Tang-Christensen, M., Larsen, P. J., Thulesen, J., Rømer, J. & Vrang, N. The proglucagon-derived peptide, glucagon-like peptide-2, is a neurotransmitter involved in the regulation of food intake. Nat. Med. 6, 802–7 (2000).

339. Guan, X. et al. GLP-2 receptor in POMC neurons suppresses feeding behavior and gastric motility. Am. J. Physiol. Endocrinol. Metab. 303, E853–64 (2012).

340. Wøjdemann, M., Wettergren, A., Hartmann, B. & Holst, J. J. Glucagon-like peptide-2 inhibits centrally induced antral motility in pigs. Scand. J. Gastroenterol. 33, 828–32 (1998).

341. Nagell, C. F., Wettergren, A., Pedersen, J. F., Mortensen, D. & Holst, J. J. Glucagon-like peptide-2 inhibits antral emptying in man, but is not as potent as glucagon-like peptide-1. Scand. J. Gastroenterol. 39, 353–8 (2004).

342. Amato, A., Baldassano, S., Serio, R. & Mulè, F. Glucagon-like peptide-2 relaxes mouse stomach through vasoactive intestinal peptide release. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G678–84 (2009).

343. McDonagh, S. C., Lee, J., Izzo, A. & Brubaker, P. L. Role of glial cell-line derived neurotropic factor family receptor alpha2 in the actions of the glucagon-like peptides on the murine intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G461–8 (2007).

344. De Heer, J., Pedersen, J., Orskov, C. & Holst, J. J. The alpha cell expresses glucagon-like peptide-2 receptors and glucagon-like peptide-2 stimulates glucagon secretion from the rat pancreas. Diabetologia 50, 2135–42 (2007).

345. Meier, J. J. et al. Glucagon-like peptide 2 stimulates glucagon secretion, enhances lipid absorption, and inhibits gastric acid secretion in humans. Gastroenterology 130, 44–54 (2006).

346. Bahrami, J., Longuet, C., Baggio, L. L., Li, K. & Drucker, D. J. Glucagon-like peptide-2 receptor modulates islet adaptation to metabolic stress in the ob/ob mouse. Gastroenterology 139, 857–68 (2010).

347. Shi, X. et al. Central GLP-2 Enhances Hepatic Insulin Sensitivity via Activating PI3K Signaling in POMC Neurons. Cell Metab. 18, 86–98 (2013).

Page 202: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

189

348. Bataille, D. et al. Bioactive enteroglucagon (oxyntomodulin): present knowledge on its chemical structure and its biological activities. Peptides 2 Suppl 2, 41–4 (1981).

349. Kervran, A., Blache, P. & Bataille, D. Distribution of oxyntomodulin and glucagon in the gastrointestinal tract and the plasma of the rat. Endocrinology 121, 704–13 (1987).

350. Chaudhri, O. B., Field, B. C. T. & Bloom, S. R. Gastrointestinal satiety signals. Int. J. Obes. (Lond). 32 Suppl 7, S28–31 (2008).

351. Pocai, A. Unraveling oxyntomodulin, GLP1’s enigmatic brother. J. Endocrinol. 215, 335–46 (2012).

352. Jorgensen, R., Kubale, V., Vrecl, M., Schwartz, T. W. & Elling, C. E. Oxyntomodulin differentially affects glucagon-like peptide-1 receptor beta-arrestin recruitment and signaling through Galpha(s). J. Pharmacol. Exp. Ther. 322, 148–54 (2007).

353. Dakin, C. L. et al. Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 145, 2687–95 (2004).

354. Cohen, M. A. et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J. Clin. Endocrinol. Metab. 88, 4696–701 (2003).

355. Baggio, L. L., Huang, Q., Brown, T. J. & Drucker, D. J. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 127, 546–58 (2004).

356. Schjoldager, B., Mortensen, P. E., Myhre, J., Christiansen, J. & Holst, J. J. Oxyntomodulin from distal gut. Role in regulation of gastric and pancreatic functions. Dig. Dis. Sci. 34, 1411–9 (1989).

357. Maida, A., Lovshin, J. A., Baggio, L. L. & Drucker, D. J. The glucagon-like peptide-1 receptor agonist oxyntomodulin enhances beta-cell function but does not inhibit gastric emptying in mice. Endocrinology 149, 5670–8 (2008).

358. Jarrousse, C., Bataille, D. & Jeanrenaud, B. A pure enteroglucagon, oxyntomodulin (glucagon 37), stimulates insulin release in perfused rat pancreas. Endocrinology 115, 102–5 (1984).

359. Ballantyne, G. H. Peptide YY(1-36) and peptide YY(3-36): Part I. Distribution, release and actions. Obes. Surg. 16, 651–8 (2006).

360. Grandt, D. et al. Two molecular forms of peptide YY (PYY) are abundant in human blood: characterization of a radioimmunoassay recognizing PYY 1-36 and PYY 3-36. Regul. Pept. 51, 151–9 (1994).

361. Grandt, D. et al. Characterization of two forms of peptide YY, PYY(1-36) and PYY(3-36), in the rabbit. Peptides 15, 815–20 (1994).

Page 203: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

190

362. Feltrin, K. L. et al. Effects of lauric acid on upper gut motility, plasma cholecystokinin and peptide YY, and energy intake are load, but not concentration, dependent in humans. J. Physiol. 581, 767–77 (2007).

363. Degen, L. et al. Effect of CCK-1 receptor blockade on ghrelin and PYY secretion in men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292, R1391–9 (2007).

364. Adrian, T. E. et al. Human distribution and release of a putative new gut hormone, peptide YY. Gastroenterology 89, 1070–1077 (1985).

365. Keire, D. A. et al. Primary structures of PYY, [Pro(34)]PYY, and PYY-(3-36) confer different conformations and receptor selectivity. Am. J. Physiol. Gastrointest. Liver Physiol. 279, G126–31 (2000).

366. Koda, S. et al. The role of the vagal nerve in peripheral PYY3-36-induced feeding reduction in rats. Endocrinology 146, 2369–2375 (2005).

367. Browning, K. N. & Travagli, R. A. Modulation of inhibitory neurotransmission in brainstem vagal circuits by NPY and PYY is controlled by cAMP levels. Neurogastroenterol. Motil. 21, 1309–e126 (2009).

368. Boey, D. et al. Peptide YY ablation in mice leads to the development of hyperinsulinaemia and obesity. Diabetologia 49, 1360–70 (2006).

369. Böttcher, G., Ahrén, B., Lundquist, I. & Sundler, F. Peptide YY: intrapancreatic localization and effects on insulin and glucagon secretion in the mouse. Pancreas 4, 282–8 (1989).

370. Nieuwenhuizen, A. G., Karlsson, S., Fridolf, T. & Ahrén, B. Mechanisms underlying the insulinostatic effect of peptide YY in mouse pancreatic islets. Diabetologia 37, 871–8 (1994).

371. Burcelin, R., Brunner, H., Seydoux, J., Thorensa, B. & Pedrazzini, T. Increased insulin concentrations and glucose storage in neuropeptide Y Y1 receptor-deficient mice. Peptides 22, 421–7 (2001).

372. Chandarana, K. et al. Peripheral activation of the Y2-receptor promotes secretion of GLP-1 and improves glucose tolerance. Mol. Metab. 2, 142–52 (2013).

373. Moran, T. H. et al. Peptide YY(3-36) inhibits gastric emptying and produces acute reductions in food intake in rhesus monkeys. Am. J. Physiol. Regul. Integr. Comp. Physiol. 288, R384–8 (2005).

374. Witte, A.-B. et al. Differential effect of PYY1-36 and PYY3-36 on gastric emptying in man. Regul. Pept. 158, 57–62 (2009).

Page 204: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

191

375. Chelikani, P. K., Haver, A. C. & Reidelberger, R. D. Comparison of the inhibitory effects of PYY(3-36) and PYY(1-36) on gastric emptying in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 287, R1064–70 (2004).

376. Fujino, T., Kang, M. J., Suzuki, H., Iijima, H. & Yamamoto, T. Molecular characterization and expression of rat acyl-CoA synthetase 3. J.Biol.Chem. 271, 16748–16752 (1996).

377. Breen, D. M., Yang, C. S. & Lam, T. K. T. Gut-brain signalling: how lipids can trigger the gut. Diabetes. Metab. Res. Rev. 27, 113–9 (2011).

378. Caspi, L., Wang, P. Y. & Lam, T. K. A balance of lipid-sensing mechanisms in the brain and liver. Cell Metab 6, 99–104 (2007).

379. Wang, P. Y. et al. Upper intestinal lipids trigger a gut-brain-liver axis to regulate glucose production. Nature 452, 1012–1016 (2008).

380. Greenberg, D., Kava, R. A., Lewis, D. R., Greenwood, M. R. & Smith, G. P. Time course for entry of intestinally infused lipids into blood of rats. Am.J.Physiol 269, R432–R436 (1995).

381. Van Horn, C. G. et al. Characterization of recombinant long-chain rat acyl-CoA synthetase isoforms 3 and 6: identification of a novel variant of isoform 6. Biochemistry 44, 1635–1642 (2005).

382. Lam, C. K. L. et al. Activation of N-methyl-D-aspartate (NMDA) receptors in the dorsal vagal complex lowers glucose production. J. Biol. Chem. 285, 21913–21 (2010).

383. Kokorovic, A. et al. Duodenal mucosal protein kinase C-δ regulates glucose production in rats. Gastroenterology 141, 1720–7 (2011).

384. Cheung, G. W., Kokorovic, A., Lam, C. K., Chari, M. & Lam, T. K. Intestinal cholecystokinin controls glucose production through a neuronal network. Cell Metab 10, 99–109 (2009).

385. Mellor, H. & Parker, P. J. The extended protein kinase C superfamily. Biochem.J. 332 ( Pt 2, 281–292 (1998).

386. Furness, J. B. et al. The distribution of PKC isoforms in enteric neurons, muscle and interstitial cells of the human intestine. Histochem. Biol. 126, 537–548 (2006).

387. Poole, D. P., Hunne, B., Robbins, H. L. & Furness, J. B. Protein kinase C isoforms in the enteric nervous system. Histochem. Biol. 120, 51–61 (2003).

388. Duran-Sandoval, D. et al. The farnesoid X receptor modulates hepatic carbohydrate metabolism during the fasting-refeeding transition. J.Biol.Chem. 280, 29971–29979 (2005).

Page 205: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

192

389. Kokorovic, A. et al. Duodenal Mucosal Protein Kinase C-delta Regulates Glucose Production in Rats. Gastroenterology 141, 1720–1727 (2011).

390. Takahashi, A. et al. Involvement of calmodulin and protein kinase C in cholecystokinin release by bombesin from STC-1 cells. Pancreas 21, 231–239 (2000).

391. Breen, D. M. et al. Duodenal PKC-δ and cholecystokinin signaling axis regulates glucose production. Diabetes 60, 3148–53 (2011).

392. Wang, J. et al. Overfeeding rapidly induces leptin and insulin resistance. Diabetes 50, 2786–2791 (2001).

393. Chari, M., Lam, C. K., Wang, P. Y. & Lam, T. K. Activation of central lactate metabolism lowers glucose production in uncontrolled diabetes and diet-induced insulin resistance. Diabetes 57, 836–840 (2008).

394. Ono, H., Pocai, A. & Wang, Y. Activation of hypothalamic S6 kinase mediates diet-induced hepatic insulin resistance in rats. J. … 118, 2959–2968 (2008).

395. Spiller, R. C. et al. The ileal brake--inhibition of jejunal motility after ileal fat perfusion in man. Gut 25, 365–74 (1984).

396. Lin, H. C., Doty, J. E., Reedy, T. J. & Meyer, J. H. Inhibition of gastric emptying by sodium oleate depends on length of intestine exposed to nutrient. Am. J. Physiol. 259, G1031–6 (1990).

397. Lin, H. C., Zhao, X. T. & Wang, L. Fat absorption is not complete by midgut but is dependent on load of fat. Am. J. Physiol. 271, G62–7 (1996).

398. Meyer, J. H., Elashoff, J. D., Doty, J. E. & Gu, Y. G. Disproportionate ileal digestion on canine food consumption. A possible model for satiety in pancreatic insufficiency. Dig. Dis. Sci. 39, 1014–24 (1994).

399. Rodriguez, M. D., Kalogeris, T. J., Wang, X. L., Wolf, R. & Tso, P. Rapid synthesis and secretion of intestinal apolipoprotein A-IV after gastric fat loading in rats. Am. J. Physiol. 272, R1170–7 (1997).

400. Holgate, A. M. & Read, N. W. Relationship between small bowel transit time and absorption of a solid meal. Influence of metoclopramide, magnesium sulfate, and lactulose. Dig. Dis. Sci. 28, 812–9 (1983).

401. Jian, R., Pecking, A., Najean, Y. & Bernier, J. J. [Study of the progression of an ordinary meal in the human small bowel by a scintigraphic method (author’s transl)]. Gastroentérologie Clin. Biol. 3, 755–62 (1979).

402. Breen, D. M. et al. Jejunal nutrient sensing is required for duodenal-jejunal bypass surgery to rapidly lower glucose concentrations in uncontrolled diabetes. Nat. Med. 18, 950–5 (2012).

Page 206: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

193

403. Maggard, M. A. et al. Meta-analysis: surgical treatment of obesity. Ann. Intern. Med. 142, 547–59 (2005).

404. Pories, W. J. et al. Who would have thought it? An operation proves to be the most effective therapy for adult-onset diabetes mellitus. Ann. Surg. 222, 339–50 (1995).

405. Sjöström, L. et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N. Engl. J. Med. 351, 2683–93 (2004).

406. Rubino, F. From bariatric to metabolic surgery: definition of a new discipline and implications for clinical practice. Curr. Atheroscler. Rep. 15, 369 (2013).

407. Cohen, R. V et al. Effects of gastric bypass surgery in patients with type 2 diabetes and only mild obesity. Diabetes Care 35, 1420–8 (2012).

408. Booth, H. et al. Incidence of type 2 diabetes after bariatric surgery: population-based matched cohort study. Lancet Diabetes Endocrinol. 2, 963–968 (2014).

409. Koshy, A. a, Bobe, A. M. & Brady, M. J. Potential mechanisms by which bariatric surgery improves systemic metabolism. Transl. Res. 161, 63–72 (2013).

410. Buchwald, H. et al. Bariatric surgery: a systematic review and meta-analysis. JAMA 292, 1724–37 (2004).

411. Ward, M. & Prachand, V. Surgical treatment of obesity. Gastrointest. Endosc. 70, 985–90 (2009).

412. Silecchia, G. et al. Effectiveness of laparoscopic sleeve gastrectomy (first stage of biliopancreatic diversion with duodenal switch) on co-morbidities in super-obese high-risk patients. Obes. Surg. 16, 1138–44 (2006).

413. Hess, D. S. & Hess, D. W. Biliopancreatic diversion with a duodenal switch. Obes. Surg. 8, 267–82 (1998).

414. Scopinaro, N., Gianetta, E., Civalleri, D., Bonalumi, U. & Bachi, V. Bilio-pancreatic bypass for obesity: II. Initial experience in man. Br. J. Surg. 66, 618–20 (1979).

415. DeMeester, T. R. et al. Experimental and clinical results with proximal end-to-end duodenojejunostomy for pathologic duodenogastric reflux. Ann. Surg. 206, 414–26 (1987).

416. Prachand, V. N., Davee, R. T. & Alverdy, J. C. Duodenal switch provides superior weight loss in the super-obese (BMI > or =50 kg/m2) compared with gastric bypass. Ann. Surg. 244, 611–9 (2006).

417. Mingrone, G. & Castagneto-Gissey, L. Mechanisms of early improvement/resolution of type 2 diabetes after bariatric surgery. Diabetes Metab. 35, 518–23 (2009).

Page 207: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

194

418. Korner, J. et al. Differential effects of gastric bypass and banding on circulating gut hormone and leptin levels. Obesity (Silver Spring). 14, 1553–61 (2006).

419. Le Roux, C. W. et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann. Surg. 243, 108–14 (2006).

420. Morínigo, R. et al. Glucagon-like peptide-1, peptide YY, hunger, and satiety after gastric bypass surgery in morbidly obese subjects. J. Clin. Endocrinol. Metab. 91, 1735–40 (2006).

421. Michalakis, K. & le Roux, C. Gut hormones and leptin: impact on energy control and changes after bariatric surgery--what the future holds. Obes. Surg. 22, 1648–57 (2012).

422. Clements, R. H., Gonzalez, Q. H., Long, C. I., Wittert, G. & Laws, H. L. Hormonal changes after Roux-en Y gastric bypass for morbid obesity and the control of type-II diabetes mellitus. Am. Surg. 70, 1–4 (2004).

423. Rubino, F. et al. The early effect of the Roux-en-Y gastric bypass on hormones involved in body weight regulation and glucose metabolism. Ann. Surg. 240, 236–42 (2004).

424. Le Roux, C. W. et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann. Surg. 246, 780–5 (2007).

425. Morínigo, R. et al. Circulating peptide YY, weight loss, and glucose homeostasis after gastric bypass surgery in morbidly obese subjects. Ann. Surg. 247, 270–5 (2008).

426. Cummings, D. E. et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N. Engl. J. Med. 346, 1623–30 (2002).

427. Peterli, R. et al. Metabolic and hormonal changes after laparoscopic Roux-en-Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial. Obes. Surg. 22, 740–8 (2012).

428. Sundbom, M., Holdstock, C., Engström, B. E. & Karlsson, F. A. Early changes in ghrelin following Roux-en-Y gastric bypass: influence of vagal nerve functionality? Obes. Surg. 17, 304–10 (2007).

429. Laferrère, B. et al. Effect of weight loss by gastric bypass surgery versus hypocaloric diet on glucose and incretin levels in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 93, 2479–85 (2008).

430. Whitson, B. A. et al. Entero-endocrine changes after gastric bypass in diabetic and nondiabetic patients: a preliminary study. J. Surg. Res. 141, 31–9 (2007).

431. Lee, W.-J. et al. Changes in postprandial gut hormones after metabolic surgery: a comparison of gastric bypass and sleeve gastrectomy. Surg. Obes. Relat. Dis. 7, 683–90 (2011).

Page 208: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

195

432. Borg, C. M. et al. Progressive rise in gut hormone levels after Roux-en-Y gastric bypass suggests gut adaptation and explains altered satiety. Br. J. Surg. 93, 210–5 (2006).

433. Liu, Y., Zhou, Y., Wang, Y., Geng, D. & Liu, J. Roux-en-Y gastric bypass-induced improvement of glucose tolerance and insulin resistance in type 2 diabetic rats are mediated by glucagon-like peptide-1. Obes. Surg. 21, 1424–31 (2011).

434. Mokadem, M., Zechner, J. F., Margolskee, R. F., Drucker, D. J. & Aguirre, V. Effects of Roux-en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse models of functional glucagon-like peptide-1 deficiency. Mol. Metab. 1–11 (2013).

435. Habegger, K. M. et al. GLP-1R responsiveness predicts individual gastric bypass efficacy on glucose tolerance in rats. Diabetes 63, 505–513 (2013).

436. Meirelles, K. et al. Mechanisms of glucose homeostasis after Roux-en-Y gastric bypass surgery in the obese, insulin-resistant Zucker rat. Ann. Surg. 249, 277–85 (2009).

437. Patel, R. T., Shukla, A. P., Ahn, S. M., Moreira, M. & Rubino, F. Surgical control of obesity and diabetes: The role of intestinal vs. gastric mechanisms in the regulation of body weight and glucose homeostasis. Obesity (Silver Spring). 22, 159–69 (2014).

438. Chambers, A. P. et al. The effects of vertical sleeve gastrectomy in rodents are ghrelin independent. Gastroenterology 144, 50–52.e5 (2013).

439. Wilson-Pérez, H. E. et al. Vertical sleeve gastrectomy is effective in two genetic mouse models of glucagon-like Peptide 1 receptor deficiency. Diabetes 62, 2380–5 (2013).

440. Rubino, F. & Marescaux, J. Effect of duodenal-jejunal exclusion in a non-obese animal model of type 2 diabetes: a new perspective for an old disease. Ann.Surg. 239, 1–11 (2004).

441. Lee, H. C., Kim, M. K., Kwon, H. S., Kim, E. & Song, K.-H. Early changes in incretin secretion after laparoscopic duodenal-jejunal bypass surgery in type 2 diabetic patients. Obes. Surg. 20, 1530–5 (2010).

442. Cohen, R. V, Schiavon, C. a, Pinheiro, J. S., Correa, J. L. & Rubino, F. Duodenal-jejunal bypass for the treatment of type 2 diabetes in patients with body mass index of 22-34 kg/m2: a report of 2 cases. Surg. Obes. Relat. Dis. 3, 195–7 (2007).

443. Geloneze, B. et al. Metabolic surgery for non-obese type 2 diabetes: incretins, adipocytokines, and insulin secretion/resistance changes in a 1-year interventional clinical controlled study. Ann. Surg. 256, 72–8 (2012).

444. Cohen, R. et al. Glycemic control after stomach-sparing duodenal-jejunal bypass surgery in diabetic patients with low body mass index. Surg.Obes.Relat Dis. 8, 375–380 (2012).

445. Troy, S. et al. Intestinal gluconeogenesis is a key factor for early metabolic changes after gastric bypass but not after gastric lap-band in mice. Cell Metab. 8, 201–11 (2008).

Page 209: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

196

446. Dixon, J. B., le Roux, C. W., Rubino, F. & Zimmet, P. Bariatric surgery for type 2 diabetes. Lancet 379, 2300–11 (2012).

447. Madsbad, S. & Holst, J. J. GLP-1 as a Mediator in the Remission of Type 2 Diabetes After Gastric Bypass and Sleeve Gastrectomy Surgery. Diabetes 63, 3172–3174 (2014).

448. Salinari, S., le Roux, C. W., Bertuzzi, A., Rubino, F. & Mingrone, G. Duodenal-Jejunal Bypass and Jejunectomy Improve Insulin Sensitivity in Goto-Kakizaki Diabetic Rats Without Changes in Incretins or Insulin Secretion. Diabetes 63, 1069 (2014).

449. Thomas, C., Pellicciari, R., Pruzanski, M., Auwerx, J. & Schoonjans, K. Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug Discov. 7, 678–93 (2008).

450. Pournaras, D. J. et al. The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology 153, 3613–9 (2012).

451. Habegger, K. M. et al. Duodenal nutrient exclusion improves metabolic syndrome and stimulates villus hyperplasia. Gut 0, 1–9 (2013).

452. Ryan, K. K. et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature 509, 183–188 (2014).

453. Scully, T. Diabetes in numbers. Nature 485, S2–S3 (2012).

454. Brownlee, M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes 54, 1615–1625 (2005).

455. Yang, W. et al. Prevalence of diabetes among men and women in China. N. Engl. J. Med. 362, 1090–101 (2010).

456. Lam, T. K. T. China needs to boost funding for graduate students to stay competitive. Nat. Med. 17, 655 (2011).

457. Lam, T. K. Neuronal regulation of homeostasis by nutrient sensing. Nat.Med. 16, 392–395 (2010).

458. Badman, M. K. & Flier, J. S. The gut and energy balance: visceral allies in the obesity wars. Science (80-. ). 307, 1909–1914 (2005).

459. Coll, A. P., Farooqi, I. S. & O’Rahilly, S. The hormonal control of food intake. Cell 129, 251–62 (2007).

460. Noble, F. et al. International Union of Pharmacology. XXI. Structure, distribution, and functions of cholecystokinin receptors. Pharmacol.Rev. 51, 745–781 (1999).

461. Williams, J. A. & Blevins Jr., G. T. Cholecystokinin and regulation of pancreatic acinar cell function. Physiol Rev. 73, 701–723 (1993).

Page 210: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

197

462. Berridge, M. J. Inositol trisphosphate and diacylglycerol as second messengers. Biochem. J. 220, 345–60 (1984).

463. Marino, C. R., Leach, S. D., Schaefer, J. F., Miller, L. J. & Gorelick, F. S. Characterization of cAMP-dependent protein kinase activation by CCK in rat pancreas. FEBS Lett. 316, 48–52 (1993).

464. Simonsson, E., Karlsson, S. & Ahrén, B. Involvement of phospholipase A2 and arachidonic acid in cholecystokinin-8-induced insulin secretion in rat islets. Regul. Pept. 65, 101–7 (1996).

465. McBain, C. J. & Mayer, M. L. N-methyl-D-aspartic acid receptor structure and function. Physiol. Rev. 74, 723–60 (1994).

466. Rong, W. et al. Jejunal afferent nerve sensitivity in wild-type and TRPV1 knockout mice. J. Physiol. 560, 867–81 (2004).

467. Rong, W., Winchester, W. J. & Grundy, D. Spontaneous hypersensitivity in mesenteric afferent nerves of mice deficient in the sst2 subtype of somatostatin receptor. J. Physiol. 581, 779–86 (2007).

468. Li, Y., Hao, Y. & Owyang, C. High-affinity CCK-A receptors on the vagus nerve mediate CCK-stimulated pancreatic secretion in rats. Am.J.Physiol 273, G679–G685 (1997).

469. Smith, R. D. & Goldin, A. L. Potentiation of rat brain sodium channel currents by PKA in Xenopus oocytes involves the I-II linker. Am.J.Physiol Cell Physiol 278, C638–C645 (2000).

470. Basbaum, A. I., Bautista, D. M., Scherrer, G. & Julius, D. Cellular and molecular mechanisms of pain. Cell 139, 267–84 (2009).

471. Neelands, P. J. & Clandinin, M. T. Diet fat influences liver plasma-membrane lipid composition and glucagon-stimulated adenylate cyclase activity. Biochem.J. 212, 573–583 (1983).

472. Myers Jr., M. G. & Olson, D. P. Central nervous system control of metabolism. Nature 491, 357–363 (2012).

473. Morton, G. J. et al. Leptin regulates insulin sensitivity via phosphatidylinositol-3-OH kinase signaling in mediobasal hypothalamic neurons. Cell Metab. 2, 411–420 (2005).

474. Buettner, C. et al. Critical role of STAT3 in leptin’s metabolic actions. Cell Metab. 4, 49–60 (2006).

475. German, J. P. et al. Leptin activates a novel CNS mechanism for insulin-independent normalization of severe diabetic hyperglycemia. Endocrinology 152, 394–404 (2011).

Page 211: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

198

476. Fujikawa, T., Chuang, J.-C., Sakata, I., Ramadori, G. & Coppari, R. Leptin therapy improves insulin-deficient type 1 diabetes by CNS-dependent mechanisms in mice. Proc. Natl. Acad. Sci. U. S. A. 107, 17391–6 (2010).

477. Hayes, M. R. et al. Endogenous leptin signaling in the caudal nucleus tractus solitarius and area postrema is required for energy balance regulation. Cell Metab 11, 77–83 (2010).

478. Attele, A. S., Shi, Z. Q. & Yuan, C. S. Leptin, gut, and food intake. Biochem.Pharmacol. 63, 1579–1583 (2002).

479. Picó, C., Oliver, P., Sánchez, J. & Palou, A. Gastric leptin: a putative role in the short-term regulation of food intake. Br. J. Nutr. 90, 735 (2007).

480. Guo, X. et al. Leptin signaling in intestinal epithelium mediates resistance to enteric infection by Entamoeba histolytica. Mucosal Immunol. 4, 294–303 (2011).

481. Duggal, P. et al. A mutation in the leptin receptor is associated with Entamoeba histolytica infection in children. 121, 1191–1198 (2011).

482. Iqbal, J. et al. An intrinsic gut leptin-melanocortin pathway modulates intestinal microsomal triglyceride transfer protein and lipid absorption. J. Lipid Res. 51, 1929–42 (2010).

483. Lostao, M. P., Urdaneta, E., Martinez-Anso, E., Barber, A. & Martinez, J. A. Presence of leptin receptors in rat small intestine and leptin effect on sugar absorption. FEBS Lett. 423, 302–306 (1998).

484. Yuan, C. S., Attele, A. S., Wu, J. A., Zhang, L. & Shi, Z. Q. Peripheral gastric leptin modulates brain stem neuronal activity in neonates. Am.J.Physiol 277, G626–G630 (1999).

485. Sheng, H., Shao, J., Townsend, C. M. & Evers, B. M. Phosphatidylinositol 3-kinase mediates proliferative signals in intestinal epithelial cells. Gut 52, 1472–8 (2003).

486. Schwartz, M. W. & Porte Jr., D. Diabetes, obesity, and the brain. Science (80-. ). 307, 375–379 (2005).

487. Rasmussen, B. A. et al. Duodenal activation of cAMP-dependent protein kinase induces vagal afferent firing and lowers glucose production in rats. Gastroenterology 142, 834–843 (2012).

488. Ishizuka, T. et al. Phenotypic consequences of a nonsense mutation in the leptin receptor gene (fak) in obese spontaneously hypertensive Koletsky rats (SHROB). J. Nutr. 128, 2299–306 (1998).

489. Takaya, K. et al. Nonsense mutation of leptin receptor in the obese spontaneously hypertensive Koletsky rat. Nat. Genet. 14, 130–1 (1996).

Page 212: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

199

490. Koletsky, S. Pathologic findings and laboratory data in a new strain of obese hypertensive rats. Am. J. Pathol. 80, 129–42 (1975).

491. Friedman, J. E. et al. Reduced insulin receptor signaling in the obese spontaneously hypertensive Koletsky rat. Am. J. Physiol. 273, E1014–23 (1997).

492. Lee, G. H. et al. Abnormal splicing of the leptin receptor in diabetic mice. Nature 379, 632–635 (1996).

493. Hummel, K. P., Dickie, M. M. & Coleman, D. L. Diabetes, a new mutation in the mouse. Science 153, 1127–8 (1966).

494. Chen, H. et al. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell 84, 491–5 (1996).

495. Coleman, D. L. Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice. Diabetologia 14, 141–8 (1978).

496. Szkudelski, T. The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol. Res. 50, 537–46 (2001).

497. Hosokawa, M., Dolci, W. & Thorens, B. Differential sensitivity of GLUT1- and GLUT2-expressing beta cells to streptozotocin. Biochem. Biophys. Res. Commun. 289, 1114–7 (2001).

498. Morton, N. M., Emilsson, V., Liu, Y. L. & Cawthorne, M. A. Leptin action in intestinal cells. J.Biol.Chem. 273, 26194–26201 (1998).

499. Zhang, J. & Scarpace, P. J. The soluble leptin receptor neutralizes leptin-mediated STAT3 signalling and anorexic responses in vivo. Br.J.Pharmacol. 158, 475–482 (2009).

500. Koletsky, S. Obese spontaneously hypertensive rats--a model for study of atherosclerosis. Exp.Mol.Pathol. 19, 53–60 (1973).

501. Wang, M. Y. et al. Leptin therapy in insulin-deficient type I diabetes. Proc.Natl.Acad.Sci.U.S.A 107, 4813–4819 (2010).

502. Chinookoswong, N., Wang, J. L. & Shi, Z. Q. Leptin restores euglycemia and normalizes glucose turnover in insulin-deficient diabetes in the rat. Diabetes 48, 1487–1492 (1999).

503. Gong, Y. et al. The long form of the leptin receptor regulates STAT5 and ribosomal protein S6 via alternate mechanisms. J.Biol.Chem. 282, 31019–31027 (2007).

504. Tong, Q., Booth, R. E., Worrell, R. T. & Stockand, J. D. Regulation of Na+ transport by aldosterone: signaling convergence and cross talk between the PI3-K and MAPK1/2 cascades. Am. J. Physiol. Renal Physiol. 286, F1232–8 (2004).

Page 213: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

200

505. De Lartigue, G., Barbier de la Serre, C., Espero, E., Lee, J. & Raybould, H. E. Diet-induced obesity leads to the development of leptin resistance in vagal afferent neurons. Am. J. Physiol. Endocrinol. Metab. 301, E187–95 (2011).

506. Liddle, R. A., Goldfine, I. D., Rosen, M. S., Taplitz, R. A. & Williams, J. A. Cholecystokinin bioactivity in human plasma. Molecular forms, responses to feeding, and relationship to gallbladder contraction. J.Clin.Invest 75, 1144–1152 (1985).

507. Herrmann, C. et al. Glucagon-like peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in response to nutrients. Digestion 56, 117–126 (1995).

508. Cote, C. D., Zadeh-Tahmasebi, M., Rasmussen, B. a., Duca, F. a. & Lam, T. K. T. Hormonal Signaling in the Gut. J. Biol. Chem. 289, 11642–11649 (2014).

509. Lawton, C. L., Delargy, H. J., Brockman, J., Smith, F. C. & Blundell, J. E. The degree of saturation of fatty acids influences post-ingestive satiety. Br. J. Nutr. 83, 473–82 (2000).

510. Card, W. I. A comparison of the inhibitory action of different fats and fatty acids introduced into the duodenum on gastric contractions. Am. J. Dig. Dis. 8, 47–53 (1941).

511. Kozimor, A., Chang, H. & Cooper, J. a. Effects of dietary fatty acid composition from a high fat meal on satiety. Appetite 69, 39–45 (2013).

512. Ballinger, A. B. & Clark, M. L. L-phenylalanine releases cholecystokinin (CCK) and is associated with reduced food intake in humans: evidence for a physiological role of CCK in control of eating. Metabolism. 43, 735–8 (1994).

513. Wang, Y. et al. Amino acids stimulate cholecystokinin release through the Ca2+-sensing receptor. Am. J. Physiol. Gastrointest. Liver Physiol. 300, G528–37 (2011).

514. Yoshikawa, T. et al. Comparative expression of hexose transporters (SGLT1, GLUT1, GLUT2 and GLUT5) throughout the mouse gastrointestinal tract. Histochem. Cell Biol. 135, 183–94 (2011).

515. Sakar, Y. et al. Positive regulatory control loop between gut leptin and intestinal GLUT2/GLUT5 transporters links to hepatic metabolic functions in rodents. PLoS One 4, e7935 (2009).

516. Yue, J. T. & Lam, T. K. Lipid sensing and insulin resistance in the brain. Cell Metab 15, 646–655 (2012).

517. Gao, S. et al. Leptin activates hypothalamic acetyl-CoA carboxylase to inhibit food intake. Proc.Natl.Acad.Sci.U.S.A 104, 17358–17363 (2007).

518. Walker, J. et al. 5-aminoimidazole-4-carboxamide riboside (AICAR) enhances GLUT2-dependent jejunal glucose transport: a possible role for AMPK. Biochem.J. 385, 485–491 (2005).

Page 214: Intestinal cholecystokinin and leptin signaling and the ...€¦ · vii List of Abbreviations AA Arachidonic acid ACC Acetyl-CoA carboxylase ACS Acyl-CoA synthetase AG Acylated-ghrelin

201

519. Washington, L., Cook, G. A. & Mansbach, C. M. Inhibition of carnitine palmitoyltransferase in the rat small intestine reduces export of triacylglycerol into the lymph. J.Lipid Res. 44, 1395–1403 (2003).

520. Nelson, D. W., Sharp, J. W., Brownfield, M. S., Raybould, H. E. & Ney, D. M. Localization and activation of glucagon-like peptide-2 receptors on vagal afferents in the rat. Endocrinology 148, 1954–62 (2007).

521. Williams, J. A. et al. Cholecystokinin activates a variety of intracellular signal transduction mechanisms in rodent pancreatic acinar cells. Pharmacol. Toxicol. 91, 297–303 (2002).

522. Broberger, C., Holmberg, K., Shi, T. J., Dockray, G. & Hokfelt, T. Expression and regulation of cholecystokinin and cholecystokinin receptors in rat nodose and dorsal root ganglia. Brain Res. 903, 128–140 (2001).

523. Duca, F. A., Côté, C. D., Rasmussen, B. A., Zadeh-tahmasebi, M. & Lam, T. Metformin activates duodenal AMPK and a neuronal network to lower glucose production. Nat. Med. In Press (2015).

524. Côté, C. D. et al. Resveratrol activates duodenal Sirt1 to reverse insulin resistance through a neuronal network in rats. Nat. Med. In Press (2015).

525. Saeidi, N. et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science 341, 406–10 (2013).

526. Mumphrey, M. B., Patterson, L. M., Zheng, H. & Berthoud, H.-R. Roux-en-Y gastric bypass surgery increases number but not density of CCK-, GLP-1-, 5-HT-, and neurotensin-expressing enteroendocrine cells in rats. Neurogastroenterol. Motil. 25, e70–9 (2013).

527. Filippi, B. M., Yang, C. S., Tang, C. & Lam, T. K. Insulin activates Erk1/2 signaling in the dorsal vagal complex to inhibit glucose production. Cell Metab 16, 500–510 (2012).

528. Patterson, L. M., Zheng, H. & Berthoud, H. R. Vagal afferents innervating the gastrointestinal tract and CCKA-receptor immunoreactivity. Anat.Rec. 266, 10–20 (2002).

529. Qiu, J., Fang, Y., Rønnekleiv, O. K. & Kelly, M. J. Leptin excites proopiomelanocortin neurons via activation of TRPC channels. J. Neurosci. 30, 1560–5 (2010).

530. Samuel, V. T. et al. Fasting hyperglycemia is not associated with increased expression of PEPCK or G6Pc in patients with Type 2 Diabetes. Proc. Natl. Acad. Sci. U. S. A. 106, 12121–6 (2009).