+ All Categories
Home > Documents > The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10...

The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10...

Date post: 12-Aug-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
103
The Role of the Glucagon-Like Peptide-1 Receptor in Atherosclerosis by Naim Panjwani A thesis submitted in conformity with the requirements for the degree of Masters of Science Institute of Medical Science University of Toronto © Copyright by Naim Panjwani (2012)
Transcript
Page 1: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

The Role of the Glucagon-Like Peptide-1 Receptor in Atherosclerosis

by

Naim Panjwani

A thesis submitted in conformity with the requirements for the degree of Masters of Science

Institute of Medical Science University of Toronto

© Copyright by Naim Panjwani (2012)

Page 2: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

ii

The Role of the Glucagon-Like Peptide-1 Receptor in

Atherosclerosis

Naim Panjwani

Masters of Science

Institute of Medical Science

University of Toronto

2012

Abstract

Objective: Glucagon-like peptide-1 receptor (GLP-1R) agonists have been shown to reduce

atherosclerosis in non-diabetic mice. We hypothesized that treatment with GLP-1R agonists

would reduce the development of atherosclerosis in diabetic Apoe-/-

mice.

Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice

for 22 weeks did not significantly reduce oral glucose tolerance (P=0.62) or HbA1c (P=0.85),

and did not reduce plaque size at the aortic sinus (P = 0.35). Taspoglutide treatment for 12 weeks

(0.4-mg tablet/month) of diabetic mice reduced body weight (P<0.05), food intake (P<0.05), oral

glucose tolerance (P<0.05), intrahepatic triglycerides (P<0.05) and cholesterol (P<0.001), and

plasma IL-6 levels (P<0.01); increased insulin:glucose (P<0.05); and unaltered oral lipid

tolerance (P=0.21), plasma triglycerides (P=0.45) or cholesterol (P=0.92). Nonetheless,

taspoglutide unaltered aortic atherosclerosis (P=0.18, sinus; P=0.19, descending aorta) or

macrophage infiltration (P=0.45, sinus; P=0.26, arch).

Conclusions: GLP-1R activation in either glucose-intolerant or diabetic mice does not

significantly modify the development of atherosclerosis.

Page 3: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

iii

Acknowledgments and Contributions

First and foremost, I would like to thank my supervisor, Dr. Daniel Drucker for having given me

the opportunity to work in his lab. He offered remarkable support, guidance, and encouragement

throughout my graduate studies. It has been a true honour to work under his supervision and an

intellectual challenge that motivated me. I would also like to extend my gratitude to my

committee members, Dr. Mansoor Husain, Dr. Myron Cybulsky, and Dr. Michelle Bendeck for

their valuable guidance, support and discussions.

This thesis would not have been possible without the help from everyone in the Drucker lab. I am

especially thankful for the outstanding mentorship from Laurie Baggio, Erin Mulvihill and

Christine Longuet. I am also grateful for the help from Laurie, Xiemin and Minsuk Kim with

glucose and lipid tolerance tests; Christine also helped with lipid tolerance tests.

I am also grateful for the friendship and support from everyone at the Drucker lab, as well as

intellectual discussions with Adriano Maida, Ben Lamont, John Ussher, Bernardo Yusta and

Marc Angeli.

I would also like to acknowledge several funding agencies—Natural Sciences and Engineering

Research Council of Canada, Banting and Best Diabetes Centre, Roche Pharmaceuticals and

Institute of Medical Science.

Finally, I would like to thank my parents, Nooruddin and Gulshan Panjwani for their love and

support.

Page 4: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

iv

Table of Contents

Abstract ..................................................................................................................................... ii

Acknowledgments ..................................................................................................................... iii

Table of Contents .......................................................................................................................iv

List of Abbreviations..................................................................................................................vi

Chapter 1. Introduction ............................................................................................................... 1

1.1 General background....................................................................................................... 1

1.2 Glucagon-like peptide-1 ................................................................................................ 1

1.2.1 Proglucagon and proglucagon-derived peptides .................................................. 1

1.2.2 GLP-1 action ...................................................................................................... 2

1.2.3 GLP-1 secretion, metabolism and clearance ....................................................... 4

1.2.3.1 Secretion .............................................................................................. 4

1.2.3.2 Metabolism .......................................................................................... 6

1.2.3.3 Clearance ............................................................................................. 7

1.2.4 The GLP-1 receptor ........................................................................................... 7

1.2.5 Structural and functional characteristics of GLP-1R agonists exendin-4 and

taspoglutide ..................................................................................................... 8

1.2.5.1 Exendin-4 ............................................................................................. 8

1.2.5.2 Taspoglutide ......................................................................................... 8

1.3 Atherosclerosis .............................................................................................................. 9

1.3.1 The development of atherosclerosis .................................................................... 9

1.3.1.1 Initiating events in atherosclerosis ........................................................ 9

1.3.1.2 Lesion propagation ............................................................................. 12

1.3.1.3 Vulnerable plaques and lesion disruption ............................................ 12

1.3.2 Atherosclerosis in diabetes ............................................................................... 13

1.3.2.1 Hyperglycemia ................................................................................... 14

1.3.2.2 Insulin resistance and hyperinsulinemia .............................................. 16

1.3.3 Lipoprotein metabolism ................................................................................... 17

1.3.4 Adipokines in athersoclerosis ........................................................................... 18

1.3.4.1 Leptin ................................................................................................. 19

1.3.4.2 Resistin .............................................................................................. 20

1.3.4.3 Interleukin-6 ....................................................................................... 20

1.3.5 The Apoe-/- mouse model of atherosclerosis ..................................................... 21

1.3.6 Current treatments for atherosclerosis .............................................................. 22

1.3.6.1 Lipid lowering .................................................................................... 22

1.3.6.2 Hypertension ...................................................................................... 23

1.3.6.3 Diabetes ............................................................................................. 24

1.4 GLP-1R actions on atherogenesis ................................................................................ 25

1.4.1 Indirect actions ................................................................................................. 25

1.4.1.1 Lipoprotein metabolism ...................................................................... 25

1.4.1.2 Blood pressure .................................................................................... 27

1.4.1.3 Adiposity and adipokines.................................................................... 27

Page 5: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

v

1.4.1.4 Insulin resistance, hyperinsulinemia and hyperglycemia ..................... 28

1.4.2 Direct actions ................................................................................................... 29

1.4.2.1 Endothelial cells ................................................................................. 29

1.4.2.2 Vascular smooth muscle cells ............................................................. 31

1.4.2.3 Monocytes and monocyte-derived cells .............................................. 33

1.5 Project rationale and hypothesis ................................................................................... 34

Chapter 2. Materials and methods .............................................................................................. 35

2.1 Exendin-4 project ........................................................................................................ 35

2.1.1 Animals, diets and drug treatments ................................................................... 35

2.1.2 Metabolic measurements .................................................................................. 36

2.1.3 Histological analysis of atherosclerotic lesions ................................................. 37

2.2 Taspoglutide project .................................................................................................... 38

2.2.1 Animals, diets and drug treatments ................................................................... 38

2.2.2 Metabolic measurements .................................................................................. 39

2.2.3 Histological analysis of atherosclerotic lesions ................................................. 40

2.2.4 Immunohistochemical analysis of macrophage infiltration ............................... 40

2.2.5 Plasma adipokine measurements ...................................................................... 41

2.3 Statistical analyses ....................................................................................................... 41

Chapter 3. Results ..................................................................................................................... 42

3.1 Exendin-4 project ........................................................................................................ 42

3.1.1 Metabolic phenotype ........................................................................................ 42

3.1.2 Atherosclerosis................................................................................................. 42

3.2 Taspoglutide project .................................................................................................... 46

3.2.1 Metabolic phenotype ........................................................................................ 46

3.2.2 Lipid metabolism ............................................................................................. 46

3.2.3 Plasma adipokines ............................................................................................ 53

3.2.4 Atherosclerosis................................................................................................. 54

3.2.5 Blood Pressure ................................................................................................. 55

Chapter 4. Discussion ................................................................................................................ 60

4.1 Effect of GLP-1R activation on body weight, adipocyte mass, adipokines and

atherogenesis ............................................................................................................. 60

4.2 Effect of GLP-1R activation on glucose homeostasis and atherogenesis ...................... 62

4.3 Effect of GLP-1R activation on lipoprotein metabolism ............................................... 64

4.4 Effect of GLP-1R activation on atheroma development ............................................... 66

4.5 Expression of the GLP-1R in liver and atheroma ......................................................... 70

Chapter 5. Conclusions ............................................................................................................. 71

Chapter 6. Future Directions ..................................................................................................... 72

6.1 Atherosclerosis ............................................................................................................ 72

6.2 Fatty liver and lipoprotein metabolism ......................................................................... 73

References ................................................................................................................................ 75

Page 6: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

vi

List of Abbreviations

ABCA1 ATP-binding cassette transporter A1

ABCG1 ATP-binding cassette subfamily G member 1

ACC Acetyl CoA carboxylase

ACCORD Action to Control Cardiovascular Risk in Diabetes

ACE Angiotensin-converting enzyme

ACS Acute coronary syndromes

ADVANCE Action in Diabetes and Vascular Disease Preterax and Diamicron Modified Release Controlled Evaluation

AGE Advanced glycation endproduct

Aib -Aminoisobutyric acid

ALE Advanced lipoxidation endproduct

AMP Adenosine monophosphate

AMPK Adenosine monophosphate-activated protein kinase

APO(a) Apolipoprotein(a)

APO-AI Apolipoprotein A-I

APO-AII Apolipoprotein A-II

APOB APO-B48 and/or APO-B100

APO-B100 Apolipoprotein B-48

APO-B48 Apolipoprotein B-100

APOC2 Apolipoprotein C-II

APOE Apolipoprotein E

ARB Angiotensin receptor blocker

ATP Adenosine triphosphate

AUC Area under the curve

BAEC Bovine aortic endothelial cell

BBB Blood-brain barrier

BCA Brachiocephalic artery

BH4 Tetrahydrobiopterin

BP Blood pressure

CAD Coronary artery disease

cAMP Cyclic adenosine monophosphate

CCL2 Chemokine C-C motif ligand-2

Page 7: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

vii

CCR2 Chemokine C-C motif receptor 2

cDNA Complementary DNA

CE Cholesteryl ester

CETP Cholesteryl ester transfer protein

cGMP Cyclic guanine monophosphate

CIMT Carotid intima-media thickness

CPT-I Carnitine palmitoyltransferase-I

CREB cAMP response element binding

CRP C-reactive protein

CVD Cardiovascular disease

DAB diaminobenzidine

DAG 1,2-diacylglycerol

DC Dendritic cell

DCCT Diabetes Control and Complications Trial

DDP-4 Dipeptidyl peptidase-4

DNA Deoxyribonucleic acid

EC Endothelial cell

ECM Extracellular matrix

EIRAKO Endothelial insulin receptor and Apoe knockout

ENHANCE Ezetimibe and Simvastatin in Hypercholesterolemia Enhances Atherosclerosis Regression

eNOS Endothelial nitric oxide synthase

EPAC2 Exchange protein directly activated by cAMP 2

ER Endoplasmic reticulum

ERK1/2 Extracellular-signal-regulated kinase-1/2

ESL1 E-selectin ligand-1

ET-1 Endothelin-1

FAS Fatty acid synthase

FC Free cholesterol

FMD Flow-mediated vasodilation

FFA Free fatty acids

FFAR1 Free fatty acid receptor-1

GIP Glucose-dependent insulinotropic polypeptide

GIPR GIP receptor

Page 8: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

viii

GLP-1 Glucagon-like peptide-1

GLP-2 Glucagon-like peptide-2

GLP1R Glucagon-like peptide-1 receptor

GP Glycoprotein

GP2B Platelet glycoprotein IIb of IIb/IIIa complex

GP3A Platelet glycoprotein IIIa of IIb/IIIa complex

GPAT Glycerophosphate acyltransferase

GPCR Guanine nucleotide-binding (G-protein)-coupled receptor

GPR119 GPCR 119

GPR120 GPCR 120

GRP Gastrin releasing peptide

GRPP Glicentin-related pancreatic polypeptide

HbA1c Hemoglobin A1c

HCAEC Human coronary artery endothelial cells

HDL High density lipoprotein

HMG-CoA 3-hydroxy-3-methylglutaryl-coenzyme A reductase

HOMA-IR Homeostatic model assessement of insulin resistance

HTGL Hepatic triglyceride lipase

HUVEC Human umbilical vein endothelial cells

hVSMC Human vascular smooth muscle cells

I/R Ischemia-reperfusion injury

ICAM-1 Intracellular adhesion molecule-1

IFN Interferon

IB I-kappaB

IL-1 Interleukin-1

IL1 Interleukin-1 beta

IL1Ra Interleukin-1 receptor antagonist

IL6 Interleukin-6

IL12 Interleukin-12

i.p. Intraperitoneal

IP1 Intervening polypeptide-1

IP2 Intervening polypeptide-2

ISI Insulin sensitivity index (glucose infusion rate to plasma insulin concentration)

Page 9: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

ix

JNK c-Jun N-terminal kinase

LAD Left anterior descending

LCAT Lecithin:cholesterol acyltransferase

LDL Low-density lipoprotein

LDLR LDL receptor

Lp(a) Lipoprotein(a)

LPS Lipopolysaccharide

LRP LDL receptor related protein

LVDP Left ventricular developed pressure

MAC-1 Macrophage-1 antigen

MAC-2 Macrophage-2 antigen

MAPK Mitogen activated protein kinase

MCD Malonyl CoA decarboxylase

MCP-1 Macrophage chemoattractant molecule-1

MHC Major histocompatibility complex

MMP Matrix metalloproteinase

MPGF Major proglucagon fragment

mRNA Messenger ribonucleic acid

MTTP Microsomal triglyceride transfer protein

NAFLD Non-alcoholic fatty liver disease

NASH Non-alcoholic steatohepatitis

NEFA Non-esterified fatty acid

NFB Nuclear factor kappa-B

NO Nitric oxide

NOD Nonobese diabetic

O-GlcNAc N-acetylglucosamine

OGTT Oral glucose tolerance test

oxLDL Oxidized LDL

PC1 Prohormone convertase-1

PC2 Prohormone convertase-2

PC3 Prohormone convertase-3

PCR Polymerase chain reaction

PDGF Platelet-derived growth factor

Page 10: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

x

PGDP Proglucagon-derived peptides

PI3K Phosphatidylinositol-3 kinase

PKA Phosphokinase A

PKC Protein kinase C

PLC Phospholipase C

PPAR Peroxisome proliferator-activated receptor

RAAS Renin-angiotensin-aldosterone system

RAGE Receptor for AGEs

RCT Reverse cholesterol transport

RNA Ribonucleic acid

ROS Reactive oxygen species

SAA Serum amyloid A

SCD Stearoyl CoA desaturase-1

SGLT1 Sodium/glucose co-transporter-1

SMC Smooth muscle cell

SOCS-3 Suppressor of cytokine signaling-3

SPT Serine palmitoyltransferase

SRA Scavenger receptor A

SR-B1 Scavenger receptor class B1 (also known as CD36)

sTNFR Soluble tumor necrosis factor receptor

STZ Streptozotocin

SVC Stromal vascular cells

T1DM Type 1 diabetes mellitus

T2DM Type 2 diabetes mellitus

T3 Triiodothyronine

TED 500,000 IU/ml Trasylol, 1.2 mg/ml EDTA, and 0.1 mM Diprotin A

TG Triglyceride (triacylglycerol)

TIMP Tissue inhibitor of metalloproteinases

tGLP-1 GLP-1(9-36)NH2 and GLP-1(9-37)

TNF- Tumor necrosis factor alpha

Tregs Regulatory T cells

TRL Triglyceride-rice lipoprotein

TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling

Page 11: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

xi

UKPDS The United Kingdom Prospective Diabetes Study

VADT Veterans Administration Diabetes Trial

VALT Vascular-associated lymphoid tissue

VCAM-1 Vascular cell adhesion molecule-1

VDC Vascular dendritic cell

VEGF Vascular endothelial growth factor

VLDL Very low density lipoprotein

VSMC Vascular smooth muscle cell

WAT White adipose tissue

XBP1 X-box-binding protein-1

ZDF Zucker diabetic fatty

Page 12: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

1

Chapter 1 Introduction

1.1 General background

Diabetes is at epidemic proportions with an estimated 366 million people living with the disease

in 2011, and by 2030 this figure is expected to rise to 552 million (Whiting et al. 2011).

Moreover, macrovascular disease (mainly myocardial infarction and stroke) is the principal

cause of death in patients with diabetes with an increased risk that is two to six times higher than

in the general population (Ruderman and Haudenschild 1984). The main cause for

macrovascular disease is atherosclerosis (Jarrett 1981), and the World Health Organization also

predicts that a worldwide epidemic of atherosclerosis will evolve as developing countries

acquire the dietary and sedentary lifestyle habits of the western world (Molecular Mechanisms

of Atherosclerosis 2005). The severity of atherosclerosis is worse in patients with diabetes as

cardiovascular events occur at an earlier age and at much higher incidences than patients with

no diabetes (Ruderman and Haudenschild 1984). Furthermore, the relative lower incidence of

cardiovascular events in pre-menopausal females is abolished by diabetes (Ruderman and

Haudenschild 1984).

There is currently no cure for atherosclerosis, and therapies today aim at reducing and thus

decelerating atherosclerosis in order to decrease cardiovascular events. While therapies such as

statins, fibrates and anti-hypertensive drugs are effective at reducing cardiovascular events in

patients at risk, there remains a significant event rate, especially in patients with diabetes and

insulin resistance (Rubenfire, Brook, and Rosenson 2010; Tenenbaum et al. 2006). The aim of

this thesis is to investigate whether the new class of anti-diabetic drugs that activate the

glucagon-like peptide-1 receptor (GLP-1R) play a role in the development of atherosclerosis in

the setting of diabetes.

1.2 Glucagon-like peptide-1

1.2.1 Proglucagon and proglucagon-derived peptides

Glucagon-like peptide-1 (GLP-1) is a 30-amino acid protein that belongs to the glucagon

superfamily of peptide hormones due to its considerable sequence homology with glucagon

Page 13: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

2

(Kieffer and Habener 1999). GLP-1 is derived from the proglucagon gene, which is expressed in

the pancreas, intestine and brain (Kieffer and Habener 1999; Baggio and Drucker 2007). In the

pancreatic cells of mammals, proglucagon is cleaved posttranslationally by prohormone

convertase-2 (PC-2) into glicentin-related pancreatic polypeptide (GRPP), glucagon, intervening

peptide-1 (IP-1) and major proglucagon fragment (MPGF); whereas in enteroendocrine L cells

and brain, proglucagon is cleaved by PC1/3 to yield oxyntomodulin, glicentin, GLP-1,

intervening peptide-2 (IP-2) and glucagon-like peptide-2 (GLP-2) (Baggio and Drucker 2007;

Kieffer and Habener 1999).

The physiologic actions of MPGF, GRPP, IP-1 or IP-2 are yet to be uncovered (Baggio and

Drucker 2007). Glucagon, on the other hand, is known for its counterregulatory actions to

insulin. Glucagon is essential for the maintenance of glucose homeostasis in the fasting state by

regulating hepatic glucose production via activation of glycogenolysis and gluconeogenesis and

inhibition of glycolysis (Ramnanan et al. 2011; Baggio and Drucker 2007). Glucagon mediates

its actions through the glucagon receptor (Rodbell et al. 1971). Oxyntomodulin reduces food

intake and hunger in both humans (Cohen et al. 2003) and rodents (Dakin et al. 2004) and

inhibits gastric acid secretion (Schjoldager et al. 1988; Schjoldager et al. 1989), gastric

emptying (Schjoldager et al. 1989), and gut motility (Pellissier et al. 2004). Glicentin is also

known to reduce gastric acid secretion and gut motility (Pellissier et al. 2004). GLP-2 modulates

gastrointestinal structure and function by stimulation of crypt cell proliferation and inhibition of

epithelial cell apoptosis (Baggio and Drucker 2007; Brubaker 2006). GLP-2 also enhances

intestinal glucose transport, increases intestinal blood flow, reduces gastric emptying and

secretion, and enhances epithelial barrier function (Baggio and Drucker 2007; Brubaker 2006).

1.2.2 GLP-1 action

GLP-1 acts on the GLP-1R (Göke and Conlon 1988; Drucker et al. 1987), a G-protein coupled

receptor that is expressed by many tissues of the body (Mayo et al. 2003). Upon nutrient

ingestion, GLP-1 is secreted from the enteroendocrine L cells of the ileum and colon and acts on

the endocrine pancreas through the GLP-1R to stimulate glucose-dependent insulin secretion

and biosynthesis. Nutrient ingestion gives rise to higher insulin levels than glucose delivery by

intravenous infusion partly due to the action of GLP-1 on the GLP-1R (Gutniak et al. 1994;

Page 14: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

3

Kreymann et al. 1987; Mojsov, Weir, and Habener 1987; Drucker et al. 1987; Unger et al.

1968). This increase in insulin secretion has been termed the “incretin effect” and is mediated by

both GLP-1 and another incretin, GIP (glucose-dependent insulinotropic polypeptide), in an

independent and additive fashion through the GLP-1R and the GIP receptor (GIPR),

respectively (Baggio and Drucker 2007; Kreymann et al. 1987; Mojsov, Weir, and Habener

1987; Gutniak et al. 1994; Drucker et al. 1987). Incretin action is potentiated for the treatment of

type 2 diabetes mellitus (T2DM) through use of agonists for the GLP-1R, such as exenatide and

liraglutide (marketed as Byetta and Victoza, respectively).

GLP-1R agonists exert actions in the cardiovascular system that are cardioprotective (Best et al.

2011; Ban et al. 2009; Noyan-Ashraf et al. 2009). GLP-1R agonists also exert actions on lipid

metabolism (Farr and Adeli 2012; Hsieh et al. 2010; Matikainen et al. 2006; Meier et al. 2006)

that are potentially anti-atherogenic (Rizzo et al. 2009). Briefly, twice-daily intraperitoneal (i.p.)

injections of 75 g/kg liraglutide for 7 days in 12-14-week old C57BL6 male non-diabetic and

diabetic (streptozotocin-induced) mice improved survival and reduced cardiac rupture after left

anterior descending artery (LAD) ligation (to induce experimental myocardial infarction)

(Noyan-Ashraf et al. 2009); survival of mice was associated with increases in the expression of

cardioprotective genes both before and after LAD ligation; and, liraglutide pre-treatment in vivo

for 1 or 7 days improved recovery of hearts (as measured by left ventricle developed pressure)

after ischemia-reperfusion injury ex vivo. In humans, a retrospective database analysis found

exenatide therapy was associated with a reduced number of cardiovascular events in patients

with T2DM compared to other standard forms of treatment for diabetes (hazard ratio 0.81). The

reduction in cardiovascular events was observed despite a higher prevalence of ischemic heart

disease, hyperlipidemia, obesity and hypertension at baseline in the group prescribed exenatide

(Best et al. 2011). GLP-1R agonists also reduce body weight through actions in the brain

(Gutzwiller et al. 1999; Alhadeff, Rupprecht, and Hayes 2012; Turton et al. 1996), reduce blood

pressure (Hirata et al. 2009) and improve lipid profile (Farr and Adeli 2012; Hsieh et al. 2010;

Meier et al. 2006; Matikainen et al. 2006) (discussed in section 1.4); these extra-pancreatic

effects all predict a reduction in atherosclerosis (Rizzo et al. 2009). Furthermore, one study in

non-diabetic mice has suggested potential direct effects of exenatide on the initiation of

atherosclerosis by action on monocytes (Arakawa et al. 2010). This thesis aims to investigate the

in vivo role of two GLP-1R agonists, exenatide and taspoglutide, on the development of

atherosclerosis in high-fat fed glucose intolerant mice and streptozotocin-induced diabetic mice.

Page 15: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

4

1.2.3 GLP-1 secretion, metabolism and clearance

1.2.3.1 Secretion

Secretion of GLP-1 is regulated by nutrients, endocrine and neural factors. GLP-1 is secreted

from enterendocrine L cells located in the distal ileum, colon, and duodenum (Kauth and Metz

1987; Theodorakis et al. 2006; Eissele et al. 1992). While a higher density of L cells is found in

the ileum and colon, human duodenum has about the same number of L cells as K cells (as

assessed by endoscopy of human volunteers followed by immunofluorescence with anti-GLP-1

antibodies on duodenal tissue) (Theodorakis et al. 2006). Food intake increases plasma levels of

GLP-1 within 15-30 minutes, well before nutrients reach the ileum. A second peak then follows

at 90-120 minutes after a meal, when nutrients have reached the ileum (Brubaker 2006;

Herrmann et al. 1995).

The first release of GLP-1 is likely mediated by both direct and/or indirect mechanisms. A direct

mechanism similar to the secretion of GIP may occur for GLP-1; GIP is released from K cells

(primarily found in the duodenum) by direct nutrient stimulation (Kreymann et al. 1987).

Secretion of GLP-1 from duodenal L cells may account for most of the early phase increase of

GLP-1 in plasma after nutrient ingestion (Theodorakis et al. 2006). At the same time, an indirect

stimulation of the L cells in the ileum and colon may also occur. The vagus nerve plays an

important role in a proximal-distal loop pathway for early stimulation of the L cell via a

cholinergic pathway; infusion of 3-4 ml of corn oil directly into the duodenum of male Wistar

rats with bilateral subdiaphragmatic vagotomy plus gut transection 10 cm from the infusion site

abolished the GLP-1 rise in plasma above basal, while electrical stimulation of the celiac

branches of the vagus that innervate the jejunum, ileum and colon resulted in a peak GLP-1

increase of 71 pg/ml above basal after 10 minutes (Rocca and Brubaker 1999). In humans, the

role of the vagus nerve in the early release of GLP-1 had been demonstrated with administration

of atropine, a muscarinic receptor antagonist, which attenuated the increase of GLP-1 in plasma

above basal (Balks et al. 1997). In rats, GIP also plays a role in the early release of GLP-1;

intravenous infusion of GIP to achieve post-prandial plasma GIP concentrations induced a 2-

fold increase in plasma proglucagon-derived peptide (PGDP) levels, including GLP-1 (Roberge

and Brubaker 1993). Furthermore, infusion of fat into the duodenum of rats whose duodena

were surgically ligated increased GIP in plasma followed by increased levels of proglucagon-

Page 16: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

5

derived peptides, including GLP-1; however, resection of the ileum and colon abolished the

increase of proglucagon-derived peptides in plasma (Rocca and Brubaker 1999). In humans,

however, GIP infusion during a hyperglycemic clamp did not raise plasma GLP-1 levels above

basal in either normal or T2DM subjects (Nauck et al. 1993). Finally, infusion of gastrin-

releasing peptide (GRP) (470 ng/h) in rats also stimulated an increase in proglucagon-derived

peptides in plasma, while blocking the GRP receptor with an antagonist failed to increase the

levels of proglucagon-derived peptides in plasma (Roberge, Gronau, and Brubaker 1996). In

addition, GRP receptor knockout mice demonstrated impaired glucose tolerance and reduced

insulin response at 10 minutes after gastric glucose administration (50 or 150 mg) (Persson et al.

2000). Thus, nutrients activate the L cell to secrete GLP-1 via a cholinergic pathway that is

dependent on the GRP receptor in rodents (Brubaker 2006).

The second phase of GLP-1 secretion is stimulated by direct contact of nutrients with L cells in

the ileum and colon. Carbohydrates and amino acids can stimulate the L cell in vitro, and may

be the main stimulators for the early phase of GLP-1 secretion by acting on the L cells in the

duodenum and jejunum. In contrast, fatty acids reaching the ileum may be the main stimulus for

the second phase of GLP-1 secretion (Brubaker 2006; Iakoubov et al. 2011).

Our current knowledge of the molecular mechanism(s) for GLP-1 release is advancing in part

due to the development of transgenic mice in which GLP-1-producing cells are labeled with a

fluorescent marker, enabling the identification of murine L cells (Diakogiannaki, Gribble, and

Reimann 2011; Parker, Reimann, and Gribble 2010; Tolhurst, Reimann, and Gribble 2008). The

L cell is equipped to detect carbohydrates, protein and fat and respond to each macronutrient

accordingly. L cells are open-type cells with apical processes extending to the gut lumen, a

morphology that facilitates sensing of nutrients (Diakogiannaki, Gribble, and Reimann 2011;

Parker, Reimann, and Gribble 2010; Tolhurst, Reimann, and Gribble 2008). Glucose is a potent

secretagogue for GLP-1, and is currently believed to act through the sodium/glucose co-

transporter-1 (SGLT1). SGLT1 transports two sodium ions per glucose molecule across the

membrane, depolarizing the cell and triggering action potentials that elevate cytosolic calcium

ions, which activate voltage-gated calcium channels (Diakogiannaki, Gribble, and Reimann

2011; Parker, Reimann, and Gribble 2010; Tolhurst, Reimann, and Gribble 2008; Gorboulev et

al. 2012). Fat is a good stimulant of GLP-1 secretion as well, and the response is proportional to

the caloric content of the ingested lipid. Fatty acid receptors like the G-protein coupled receptor

Page 17: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

6

120 (GPR120) and free fatty acid receptor-1 (FFAR1) are G-protein coupled receptors (GPCRs)

that trigger GLP-1 release by coupling to Gq, activating protein kinase C (PKC) (among which,

PKC zeta is essential) and inositol triphosphate (IP3)-induced calcium release (Diakogiannaki,

Gribble, and Reimann 2011; Iakoubov et al. 2011). The G-protein coupled receptor 119

(GPR119) is another GPCR activated by oleoylethanolamide or 2-monoacylglycerol that

couples to Gs rather than Gq, stimulating adenylyl cyclase and intracellular cyclic adenosine

monophosphate (cAMP) production (Diakogiannaki, Gribble, and Reimann 2011; Hansen et al.

2011). Finally, male Sprague-Dawley rats given a lipid stimulus (Liposyn) plus a surfactant

(Pluronic L-81) known to inhibit chylomicron formation in the intestine while not interfering

with uptake of lipolytic products and reesterification to triglyceride (TG), showed a 75%

decrease in GLP-1 secretion compared to rats given Liposyn alone during the first 30 minutes of

lipid infusion; this evidence suggests a role for chylomicron formation in the secretion of GLP-1

(Lu et al. 2012).

1.2.3.2 Metabolism

GLP-1 is processed from proglucagon and secreted as GLP-1(1-36)NH2 and GLP-1(1-37),

which are thought to be inactive (Ghiglione et al. 1984); and GLP-1(7-36)NH2 and GLP-1(7-

37), which are the bioactive equipotent peptides liberated from proglucagon by PC1/3 cleavage

in the intestine and brain (Orskov, Wettergren, and Holst 1993; Dhanvantari, Seidah, and

Brubaker 1996).

The half-life of GLP-1 is less than 2 minutes due to rapid degradation by the serine protease

dipeptidyl peptidase-4 (DPP-4), which cleaves GLP-1 in the alanine position 2 yielding GLP-

1(9-36)NH2 or GLP-1(9-37) (Deacon et al. 1995), which are also thought to play a role in the

cardiovascular system (Ban et al. 2009). DPP-4 is expressed in many tissues, including the

surface of endothelial cells that line the intestinal mucosa adjacent to the L cell; DPP-4 also

exists as a soluble form in the circulation. Thus, more than half of GLP-1 that enters the portal

circulation is in the cleaved form (Hansen et al. 1999; Mentlein 1999). Intravenous infusion of

GLP-1(7-36)NH2 in both healthy and T2DM patients results in rapid cleavage by DPP-4

(Deacon et al. 1995); infusion of [125

I]GLP-1(7-36)NH2 in male Wistar rats results in 50% of the

peptide being cleaved by DPP-4 within 2 minutes to [125

I]GLP-1(9-36)NH2 metabolite, while

Page 18: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

7

infusion of the non-truncated tracer into DPP-4-deficient animals results in the absence of

[125

I]GLP-1(9-36)NH2 metabolite (Kieffer, McIntosh, and Pederson 1995). Inhibition of DPP-4

or its absence in DPP-4-deficient animals greatly increases the half-life of non-truncated GLP-1

(Marguet et al. 2000).

1.2.3.3 Clearance

GLP-1(9-36)NH2 and GLP-1(9-37) (truncated GLP-1 or tGLP-1) are cleared from the

circulation by the kidneys by glomerular filtration, tubular uptake and catabolism (Ruiz-Grande

et al. 1993). Due to renal clearance, the plasma half-life of tGLP-1 is about 5 minutes.

Furthermore, patients with renal failure or chronic renal insufficiency have an increased half-life

of tGLP-1 in the circulation (Meier et al. 2004; Orskov, Andreasen, and Holst 1992).

1.2.4 The GLP-1 receptor

The GLP-1R belongs to the class B family of guanine nucleotide-binding (G-protein)-coupled

receptors (GPCRs) known as the secretin receptor family of 7 transmembrane receptors,

subfamily B1, which consists of multiple receptors for peptide hormones (Mayo et al. 2003).

The rat and human receptors are structurally similar, 463 amino acids in length with 90%

sequence identity at the amino acid level (Mayo et al. 2003).

Agonists for the GLP-1R include GLP-1(7-37), GLP-1(7-36)NH2, and the Heloderma

suspectum peptides exendin-3 and exendin-4 (Mayo et al. 2003). A known antagonist for the

GLP-1R is the truncated lizard peptide exendin(9-39) (Mayo et al. 2003).

The GLP-1R is expressed in many cells and tissues such as -cells of pancreatic islets

(Tornehave et al. 2008), lung, brain, heart, kidney and stomach of rodents and humans as

assessed by RNAse protection assay (Bullock, Heller, and Habener 1996; Wei and Mojsov

1995). Although it has been shown that the GLP-1R can be desensitized and internalized by

phosphorylation of three serine doublets within the cytoplasmic tail in vitro (Mayo et al. 2003),

GLP-1R desensitization has not been observed in vivo in the endocrine pancreas of mice treated

with exendin-4 (2 g twice daily), even after long-term (26-week) administration (Hadjiyanni et

Page 19: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

8

al. 2008). Similarly, twice-daily administration of exenatide in human patients with T2DM was

effective in significantly reducing HbA1c and glycemic deterioration after approximately 5

years of treatment (Gallwitz et al. 2012).

1.2.5 Structural and functional characteristics of GLP-1R agonists exendin-4 and taspoglutide

The short plasma half-lives of GLP-1(7-37) and GLP-1(7-36)NH2 (2 min) makes the use of

these peptides suboptimal for treatment. One major reason for the short plasma half-lives is

cleavage at the 8th residue (alanine) by DPP-4. Thus, GLP-1R agonists resistant to cleavage by

DPP-4 have been developed for pharmaceutical use. Two GLP-1R agonists have been used to

investigate GLP-1R activation in the development of atherosclerosis in diabetic mice in this

study: exendin-4 and taspoglutide.

1.2.5.1 Exendin-4

Exendin-4 is a 39-residue peptide produced in the salivary gland of the Gila monster

(Heloderma suspectum) that is not cleaved by DPP-4 (Baggio and Drucker 2007). Exendin-4

has 53% identity to GLP-1(7-36)NH2 with 16 identical amino acids in the first 30, and an

additional C-terminal extension of nine amino acid residues. Exendin-4 is equipotent to GLP-1

in binding the GLP-1R and acts as a GLP-1 mimetic (Thorens et al. 1993). The synthetic version

of exendin-4 is exenatide and has a plasma half-life of 4-6 hours after subcutaneous

administration in patients with T2DM with the kidney being the major route of elimination

(Kolterman et al. 2005). Mice and rats show similar plasma half-lives (Copley et al. 2006).

1.2.5.2 Taspoglutide

Taspoglutide is a peptide very similar to human GLP-1(7-36)NH2 with the exception of

substitutions at position 8 and 35 with -aminoisobutyric acid (Aib), which render the peptide

resistant to degradation by DPP-4 (Dong et al. 2011). This drug was formulated by Roche in the

form of a microtablet for mice that releases peptide slowly and is suitable for once-monthly

dosing. In humans, the drug was formulated as a sustained-release once-weekly formulation

Page 20: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

9

with zinc chloride. Subcutaneous administration of taspoglutide precipitates and forms a depot

that slowly dissolves into the circulation (Dong et al. 2011). Unfortunately, taspoglutide was

withdrawn from human clinical testing after results from phase 3 clinical trials revealed a high

rate of withdrawal from treatment (11-13% versus 3.3% in placebo) and hypersensitivity

reactions in four patients, two of which withdrew (Raz et al. 2012).

1.3 Atherosclerosis

1.3.1 The development of atherosclerosis

Atherosclerosis is a disease of very complex etiology (Weber and Noels 2011) and this chapter

does not aim to review all aspects of its development and/or progression. Rather, the aim of the

chapter is to selectively review important aspects of atherogenesis and how diabetes impacts its

development.

Atherosclerosis is a disease characterized by a chronic inflammatory process in the large arteries

that is progressive and involves excess deposition of low-density lipoprotein (LDL) cholesterol

and fibrous elements in the intima of the artery (Lusis 2000; Ross 1997, 1999). Some of the

known factors that influence development and progression of atherosclerosis include

hyperlipidemia, high blood pressure, diabetes, smoking and genetic factors (Molecular

Mechanisms of Atherosclerosis 2005). In humans, the first signs of atherosclerosis may be

found in the first decade of life in the aorta and the lesions progress with age (Lusis 2000). In

this section, a brief description of the different stages of atherogenesis is discussed.

1.3.1.1 Initiating events in atherosclerosis

Atherosclerosis is thought to be initiated by a combination of endothelial dysfunction, which

upregulates adhesion molecules and chemokines by endothelial cells to attract inflammatory

cells such as lymphocytes and monocytes; a non-confluent luminal elastin layer such as those

found at branching points of arteries that facilitates entry of lipids and immune cells; and

exposure of negatively-charged proteoglycans at the luminal surface of arteries that promote

binding of APOB100-containing lipoproteins (Kwon et al. 2008). The accumulation and

oxidative modification of lipids and LDL in the intima of the artery is mediated by reactive

Page 21: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

10

oxygen species (ROS) and/or enzymes released from inflammatory cells such as

myeloperoxidase and lipoxygenases (Weber and Noels 2011). Atherogenesis is largely mediated

by a very complex interaction between the cells of the inflammatory immune system that may

be analogous to an autoimmune reaction (Bobryshev 2000; Weber and Noels 2011; Hansson

and Hermansson 2011).

Accumulation of lipoproteins and their aggregates in the intima occurs mainly at sites of

predilection, where blood flow is turbulent rather than uniform and laminar. Turbulent flow

occurs most commonly at branching and curvature sites (eg. aortic arch) of arteries (Collins and

Cybulsky 2001). This occurs because endothelial cells take an ellipsoid shape reflecting the

flow’s uniform laminar directionality, but where flow is disturbed, the cells have polygonal

shapes with no particular orientation, and are more permeable (Collins and Cybulsky 2001).

Interestingly, these sites also have intimal thickenings of vascular smooth muscle cells

(VSMCs) that are thought to be important in the initiation and late stages of atherogenesis due to

their ability to proliferate, migrate and produce extracellular matrix (ECM) (Doran, Meller, and

McNamara 2008).

LDL accumulation in the intima itself has limited atherogenicity; it is the modification of LDL,

such as its oxidation, that initiates the inflammatory process. This involves recruitment of T

cells and monocyte-derived cells that engulf the lipoproteins at a rate higher than it can be

eliminated, thus forming lipid-laden cells called “foam cells” (Lusis 2000; Bobryshev 2000;

Cybulsky and Jongstra-Bilen 2010; Paulson et al. 2010; Weber and Noels 2011). VSMCs can

also take up lipid and appear foamy under the microscope (Doran, Meller, and McNamara

2008). Thus, hypercholesterolemia is associated with higher rates of atherosclerosis initiation

and progression, and cardiovascular events (Collins and Cybulsky 2001).

Factors that accelerate the development of atherosclerosis include both genetic and

environmental components (Lusis 2000; Weber and Noels 2011). Some examples include:

elevated levels of LDL and/or very low-density lipoprotein (VLDL), reduced levels of HDL (the

“good cholesterol”), elevated levels of lipoprotein(a) (Lp(a)), elevated blood pressure, elevated

homocysteine, diabetes, elevated levels of haemostatic factors, depression, gender (men develop

more atherosclerosis than pre-menopausal women), systemic inflammation, and insulin

Page 22: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

11

resistance (Lusis 2000). Some environmental factors include: high-fat diet, smoking, low

antioxidant levels and lack of exercise (Lusis 2000).

Adherence and transmigration (diapedesis) of leukocytes from the blood circulation is mediated

by activated endothelial cells (ECs) (Molecular Mechanisms of Atherosclerosis 2005). EC

activation occurs under a variety of conditions, such as exposure to tumor necrosis factor alpha

(TNF-, interleukin-1 beta (IL-1, endotoxin, or substances that induce oxidative stress that is

overwhelming to the protective antioxidant mechanisms of the cell (Molecular Mechanisms of

Atherosclerosis 2005). EC activation results in the activation of nuclear factor kappa-B (NFB)

transcription factor, which mediates the upregulation of adhesion molecules for increased

leukocyte rolling, firm adherence, and diapedesis (Molecular Mechanisms of Atherosclerosis

2005; Collins and Cybulsky 2001). Families of adhesion molecules include the selectins (E-

selectin, P-selectin), integrins (heterodimeric molecules like vitronectin, macrophage antigen-1

(MAC-1), GP IIb/IIIa), and immunoglobulin superfamily members: intracellular adhesion

molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) (Molecular

Mechanisms of Atherosclerosis 2005). Leukocyte rolling is mediated by E-selectin and

leukocyte-expressed E-selectin ligand-1 (ESL-1); firm adhesion happens through VCAM-1 and

ICAM-1 on ECs with MAC-1, and other 4, D-integrins on leukocytes; and, transmigration is

induced by chemotactic factors such as macrophage chemoattractant molecule-1 (MCP-1),

which acts on the CCR2 receptor (4).

Defense mechanisms have also been identified that counteract the formation of atherosclerotic

lesions, mainly at the initiation stage. One such mechanism is mediated by the antioxidant

protein serum paraoxonase, an esterase carried by HDL that degrades oxidized phospholipids

(Lusis 2000). In addition, reverse cholesterol transport by HDL through the ATP-binding

cassette transporter A1 (ABCA1) and the ATP-binding cassette subfamily G member 1

(ABCG1) transporters, which mediate cholesterol efflux on macrophages is another means of

diminishing deposited LDL in the intima and thus, prevent lesion initiation and/or progression

(Yvan-Charvet, Wang, and Tall 2010; Yvan-Charvet et al. 2010). Secretion of APOE by

macrophages also promotes cholesterol efflux as evidenced by bone marrow transplantation

experiments to wildtype mice with Apoe-deficient marrow (Lusis 2000; Fazio et al. 1997; Basu

et al. 1982; Basu et al. 1981). Mice transplanted bone marrow deficient in APOE developed

atherosclerosis without inducing a change in the lipoprotein profile (Fazio et al. 1997). APOE is

Page 23: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

12

well known to be atheroprotective due to its role in pro-atherogenic lipoprotein remnant uptake

by the liver, but it is also involved in the generation and maturation of APOE-containing HDL

particles in conjunction with ABCA1 and lecithin:cholesterol acyltransferase (LCAT), which is

an important pathway for the secretion of cholesterol by macrophage foam cells (Kypreos and

Zannis 2007). Thus, ABCA1, ABCG1, LCAT and APOE may all work in conjunction for the

efflux of cholesterol and formation of HDL particles (Yvan-Charvet, Wang, and Tall 2010;

Basu et al. 1981; Basu et al. 1982; Fazio et al. 1997; Kypreos and Zannis 2007; Yvan-Charvet et

al. 2010).

1.3.1.2 Lesion propagation

Oxidized LDL (OxLDL) induces EC oxidative stress leading to expression of genes under the

transcriptional regulation of NFB, such as the adhesion molecules ICAM-1 and VCAM-1

(Molecular Mechanisms of Atherosclerosis 2005; Collins and Cybulsky 2001). The

overexpression of adhesion molecules and chemotactic factors recruit leukocytes from the

circulation to the site of EC activation. Leukocytes recruited include neutrophils, monocytes, T

cells, B cells, dendritic cells (DCs) and mast cells (Weber and Noels 2011). The recruitment of

diverse cell subsets into the plaque lesion hints to the complexity, robustness and specificity of

the chemokine system (Koenen and Weber 2010). When the rate of endocytosis exceeds that of

cholesterol efflux, the macrophages accumulate significant amounts of cholesterol to become

foam cells and appear morphologically distinct under the microscope. Foam cells express IL-1

and TNF-, which further activate ECs, secrete proteoglycans that further induce APOB100-

containing lipoprotein binding, and induce VSMCs to produce matrix proteins (Molecular

Mechanisms of Atherosclerosis 2005; Lusis 2000; Moore and Tabas 2011).

1.3.1.3 Vulnerable plaques and lesion disruption

Cytokines and growth factors released by macrophages and T cells induce VSMC migration,

proliferation and ECM protein production such as collagen and fibronectin (Molecular

Mechanisms of Atherosclerosis 2005; Lusis 2000). As the lesion develops, a central necrotic

core of deposited lipid (mostly esterified cholesterol) and cell debris is covered by the migrating

VSMCs and secreted ECM, which form a fibrous cap that covers the growing lesion. As the

Page 24: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

13

lesion grows, it obstructs blood flow and may produce ischaemic syndromes, especially when

blood flow demand is high (Molecular Mechanisms of Atherosclerosis 2005; Lusis 2000).

The fibrous cap prevents thrombus formation by separating the dense lipid core from luminal

blood flow. In certain cases, however, the inflammatory response results in the rupture and

erosion of the plaque, leading to acute coronary syndromes (ACS) (Virmani et al. 2006). A

recent study showed the presence of thin-cap atherosclerosis lesions preceding acute coronary

events (Virmani et al. 2006). Upon rupture, a superimposed thrombus may form and occlude

blood flow, leading to infarction of the affected tissue (Molecular Mechanisms of

Atherosclerosis 2005; Lusis 2000). It is currently unclear, largely due to the lack of animal

models, as to how these inflammatory events are initiated resulting in thinning of the fibrous cap

and its subsequent rupture (Molecular Mechanisms of Atherosclerosis 2005; Lusis 2000).

1.3.2 Atherosclerosis in diabetes

While atherosclerosis is accelerated in both type 1 and 2 diabetes, the factors involved are not

the same. In T2DM, the increased prevalence of obesity, hypertension, dyslipidemia,

hyperinsulinemia, and insulin resistance are associated factors that contribute to the increased

risk for macrovascular disease. These factors usually precede the onset of type 2 diabetes

(Molecular Mechanisms of Atherosclerosis 2005).

In contrast, in T1DM, the increased cardiovascular risk (10-fold compared to the general

population) is thought to be due in part to increased lipids and lack of perfect glycemic control,

but controlling for these factors still leaves an exceptionally increased risk that cannot be fully

explained (Nadeau and Reusch 2011). A follow-up of subjects with T1DM from the Diabetes

Control and Complications Trial (DCCT) treated intensively for 6.5 years to achieve tight

glucose control showed that this period of optimal glycemic control reduced the incidence of

cardiovascular disease by 57% even with deteriorating glycemia thereafter (Nathan et al. 2005).

However, the increased cardiovascular risk cannot be completely explained by hyperglycemia.

Whole-body insulin resistance is present in T1DM patients despite intensive glucose control (as

in the DCCT) and appears to correlate with cardiac and vascular defects (Orchard et al. 2003;

Nadeau et al. 2010). Furthermore, in most patients with T1DM, nephropathy and hypertension

also greatly accelerate atherosclerosis (Molecular Mechanisms of Atherosclerosis 2005).

Page 25: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

14

Overall, a good understanding of the factors contributing to cardiovascular dysfunction in

T1DM is currently lacking.

1.3.2.1 Hyperglycemia

A predisposing factor in patients with diabetes that accelerates atherosclerosis was long thought

to be hyperglycemia. Hyperglycemia induces endothelial cell dysfunction and increases

nitrosative and oxidative stress (Ceriello et al. 2002). Hyperglycemia mediates accelerated

atherosclerosis by oxidative stress, which results in the formation of advanced glycation

endproducts (AGEs), synthesis of 1,2-diacylglycerol (DAG) and PKC activation, NFB-

mediated gene transcription, increase in the polyol and hexosamine pathways, and apoptosis

(Brownlee 2001). These changes due to hyperglycemia-induced oxidative stress lead to EC

activation and acceleration of atherosclerosis.

Clinical trials, however, reveal a weak association between blood glucose levels and

macrovascular outcomes. The United Kingdom Prospective Diabetes Study (UKPDS) trial

consisted of 5,102 newly-diagnosed T2DM patients with fasting plasma glucose concentrations

between 6.1 and 15.0 mM and randomized to either conventional glucose control (primarily

with diet) or intensive glucose control with sulphonylurea or insulin for an average of 10 years

(Stratton et al. 2000). The aim for the conventional control group was a fasting glucose of ≤15

mM and the intensive group <6 mM. The control group achieved a median hemoglobin A1c

(HbA1c) of 7.9% (HbA1c is a marker of average glucose control over 2-3 months measured by

the level of glycosylation of red blood cells) and the intensive group achieved a median HbA1c

of 7.0%. The intensive control group had a reduced risk of combined fatal or nonfatal

myocardial infarction and sudden death of 16% (P = 0.052), suggesting that hyperglycemia

alone is a small factor in the associated increased risk in people with diabetes as there is still a

significant event rate (Stratton et al. 2000).

Three other trials that specifically focused on glucose control and its association with

cardiovascular outcomes also failed to show a significant reduction in cardiovascular risk

(Macisaac and Jerums 2011; Control et al. 2009). The Action to Control Cardiovascular Risk in

Diabetes (ACCORD) trial was a randomized study of 10,251 patients living with type 2 diabetes

for 10 years and with established cardiovascular disease or additional cardiovascular risk factors

Page 26: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

15

(anatomical evidence of significant atherosclerosis, albuminuria, left ventricular hypertrophy, or

two risk factors such as dyslipidemia, hypertension, status as a smoker or obesity) and a median

HbA1c of 8.1% at baseline. Patients in the ACCORD trial were assigned to either intensive

glucose control treatment to achieve a HbA1c below 6.0% or to standard treatment to achieve a

HbA1c of 7.0-7.9%. Treatments to achieve these targets included metformin, sulfonylureas,

meglitinides, thiazolidinediones, -glucosidase inhibitors, insulin, insulin analogues and

lifestyle intervention. After a 3.5-year follow-up, the ACCORD study’s intensive treatment

group had increased rates of severe hypoglycemia requiring assistance. But more importantly,

increased mortality was observed in the intensive treatment group and the trial was discontinued

(Gerstein et al. 2008).

The Action in Diabetes and Vascular Disease Preterax and Diamicron Modified Release

Controlled Evaluation (ADVANCE) trial was a randomized study of 11,140 patients with type 2

diabetes with an age ≥55 with a median follow-up of 5 years. The study also consisted of an

intensive glucose lowering group aiming for an HbA1c of ≤6.5% and a standard glucose therapy

group. Patients in the intensive therapy group were treated with gliclazide MR, a sulphonylurea,

plus any additional therapy required to achieve the HbA1c target; patients in the standard

therapy group were treated according to local guidelines with any required medication other

than gliclazide. Patients in the ADVANCE trial achieved a mean HbA1c of 6.5% in the

intensive therapy group for 2 years, but the study did not find a significant difference in the

number of macrovascular events between the groups (hazard ratio 0.94 [0.84-1.06], P = 0.32)

after a 5-year follow-up (Heller 2009).

Finally, the Veterans Affairs Diabetes Trial (VADT) trial was a study with 1791 randomized

patients with poorly-controlled type 2 diabetes with a median HbA1c of 9.4% who were

diagnosed a mean of 11.5 years earlier. Patients were allocated into an intensive treatment or

standard treatment group for glucose lowering and achieved a median HbA1c of 6.9% and

8.4%, respectively. Patients were treated with metformin plus rosiglitazone or glimepiride plus

rosiglitazone, with the intensive group started on maximal doses and the standard group on half-

maximal doses. Insulin was also added if the intensive group did not achieve a HbA1c of <6%

or the standard group patient was above 9%. As in the ACCORD and ADVANCE trials, the

VADT trial also failed to show a significant reduction in cardiovascular risk after a median

follow-up of 5.6 years in the veterans population (Duckworth et al. 2009).

Page 27: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

16

1.3.2.2 Insulin resistance and hyperinsulinemia

The observation that cardiovascular risk increases well before the onset and diagnosis of type 2

diabetes has led to the belief that perhaps insulin resistance or hyperinsulinemia or both

contribute to the increased risk (Molecular Mechanisms of Atherosclerosis 2005; Bornfeldt and

Tabas 2011). Insulin resistance is often accompanied by hyperinsulinemia, and each contribute

independently to the risk for vascular disease; the relative contribution of each, however, has

been a matter of debate (Christian and George 2007). The term “insulin resistance” can either

mean decreased or increased insulin signaling through the insulin receptor; most studies,

however, have examined the effect of reduced insulin signaling (Bornfeldt and Tabas 2011).

Hypercholesterolemic (Apoe-/) mice with a knockout of the insulin receptor specifically in

endothelial cells (EIRAKO) showed a marked increase in atherosclerosis (as seen in the en face

flat preparation of the aorta) when fed a low-fat diet (9 kcal% with 0.221 ppm cholesterol) for

24 and 52 weeks (Rask-Madsen et al. 2010). Similar increases in atherosclerosis in mice are

obtained when the insulin receptor is eliminated in monocyte-derived cells (Seongah et al.

2006). A subsequent report showed hypercholesterolemic (Apoe-/-

) mice with one allele of the

insulin receptor knocked out in all tissues resulted in hyperinsulinemia due to reduced insulin

clearance (50% increase in plasma insulin without altering insulin resistance in other tissues),

and this hyperinsulinemic state did not change atherosclerosis burden when measured as en face

lesion area, cross-sectional plaque area in the aortic sinus or cholesterol abundance in the

brachiocephalic artery compared to control after 24 and 52 weeks of age (always fed a 9 kcal%

low-fat diet with 0.221 ppm cholesterol) (Rask-Madsen et al. 2012). Other studies show that

insulin resistant subjects often have elevated free fatty acids (FFAs) in the plasma, which impair

the action of insulin on the endothelium (Molecular Mechanisms of Atherosclerosis 2005;

Hamilton, Chew, and Watts 2007). These studies suggest that insulin signaling in ECs and

monocytes may play an important role in atherosclerosis, while hyperinsulinemia may have a

less significant role.

Page 28: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

17

1.3.3 Lipoprotein metabolism

Lipids are of central importance in the initiation and progression of atherosclerosis. This section

briefly describes the important aspects of lipoprotein metabolism.

Lipids are hydrophobic and thus, are carried within lipoprotein complexes surrounding the

hydrophobic particles within a spherical core and exposing hydrophilic residues to solubilize

nonpolar lipids in plasma (Vance, Vance, and Bernardi 2002). In the intestine, bile salts

synthesized from cholesterol in the liver are secreted through the gallbladder following ingestion

of fat. Bile salts facilitate the emulsification of fat for lipolysis by pancreatic lipases. Once

absorbed by enterocytes, the FFAs are re-synthesized into TGs. TGs in the enterocytes are

incorporated into chylomicrons (which also have some cholesteryl ester (CE), free cholesterol

(FC) and phospholipids) on the APOB48 template and secreted into lymph, where they drain

into the circulation at the left subclavian vein. In the circulation, chylomicrons acquire APOE,

(apolipoprotein A-I) APO-AI, (apolipoprotein A-II) APO-AII, and (apolipoprotein C-II) APO-

CII from HDL (among other lipoproteins). Another key function of apolipoproteins is binding to

specific receptors. For example, APO-CII in chylomicrons activates lipoprotein lipase (LPL) on

ECs to release FFAs to peripheral cells such as muscle and adipose. As chylomicrons lose TGs,

they lose APO-AI, APO-AII and APO-CII and are termed chylomicron remnants. Chylomicron

remnants are internalized by the liver through the LDL-receptor related protein (LRP) and the

LDL receptor (LDLR) by endocytosis mediated by APOE (Vance, Vance, and Bernardi 2002).

Endogenous TGs are secreted in VLDL by the liver and contain apolipoprotein B-100 (APO-

B100), and later acquire APO-AI, APO-AII, APOC, and APOE from HDL (Vance, Vance, and

Bernardi 2002). Analogous to chylomicrons, VLDL particles also have greater TGs within their

core and deliver them to the periphery through LPL. As VLDL particles shrink, they become

VLDL remnants and are taken up by the liver through APOE binding to LDLR and/or LRP

(Vance, Vance, and Bernardi 2002).

After internalization of remnants in the liver, excess TGs in remnants undergo lipolysis by

hepatic triglyceride lipase (HTGL), remnants lose APO-CII and APOE (transferred back to

HDL), decrease in size, and are secreted as LDL into the circulation (Vance, Vance, and

Bernardi 2002). LDL also forms in the circulation from the lipolysis of TGs within VLDL. Also,

because LDL lacks APOE, its residence time is longer in the circulation than remnant particles

Page 29: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

18

as its uptake must be mediated by APO-B100 alone, which has less affinity for the LDLR.

Furthermore, LDL is richer in cholesterol and serves as an extracellular cholesterol depot for

peripheral cells, which take up LDL through LDLR-mediated endocytosis. The most

atherogenic lipoproteins are cholesterol-rich remnant lipoproteins and LDL due to their smaller

size, which eases penetration into the subendothelium, and the presence of APOB, which bind

matrix proteoglycans through electrostatic interactions (Vance, Vance, and Bernardi 2002).

To complete the cycle, reverse cholesterol transport (RCT) is mediated by HDL and is

characterized by removal of excess cholesterol from peripheral tissues, including cholesterol-

laden macrophages, to the liver for excretion via the bile into feces (Vance, Vance, and Bernardi

2002; Brufau, Groen, and Kuipers 2011). While the traditional view of HDL function has been

in the removal of cholesterol, HDL also contributes to the extracellular cholesterol pool by

exchange of cholesterol through cholesteryl ester transfer protein (CETP) with VLDL and

remnant particles (Dominiczak and Caslake 2011). HDL is assembled on the APO-AI template,

which is secreted by both the liver and the intestine (Vance, Vance, and Bernardi 2002). Once

secreted, nascent HDL consists of phospholipid-rich discoid particles that bind to ABCA1

triggering cholesterol efflux from cells. Free cholesterol is then esterified by LCAT within the

HDL particle. As nascent HDL acquires cholesterol, it enlarges to become spherical HDL3.

HDL3 transfer cholesteryl esters to VLDL, chylomicrons and remnants in exchange for TGs.

HDL3 thus enlarges into HDL2 and offloads its cholesteryl esters into the liver by docking into

scavenger receptor B1 (SR-B1). Thus, HDL is considered anti-atherogenic as it removes

cholesterol from the body (Vance, Vance, and Bernardi 2002).

1.3.4 Adipokines in athersoclerosis

Obesity is associated with a predisposition to developing atherosclerosis, diabetes and non-

alcoholic fatty liver disease (NAFLD) (McGill et al. 2002). This association may relate to the

complex interaction of adipocytes with immune cells (stromal vascular cells or SVCs) through

secretion of adipokines. Obese individuals have more macrophages in adipocytes and they

secrete adipokines and cytokines that are pro-inflammatory (Tilg and Moschen 2006). Known

adipokines include adiponectin, leptin, resistin and visfatin. TNF-, interleukin-6 (IL-6) and

MCP-1 (also known as chemokine C-C motif ligand 2 or CCL2) are secreted by both adipocytes

Page 30: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

19

and resident macrophages, and are considered cytokines. The role of leptin, resistin and IL-6 in

obesity are discussed.

1.3.4.1 Leptin

Leptin is a 16-kDa hormone secreted by adipocytes in proportion to adipocyte mass and is

virtually undetectable in the SVC fraction that contains the immune cells (Maury and Brichard

2010). Leptin signals energy deficiency to the brain and its circulating levels parallel adipose

tissue mass and nutritional status (leptin levels decrease during fasting). Obesity is associated

with high leptin levels and leptin resistance due to diminished transport of leptin across the

blood-brain barrier (BBB) and elevated suppressor of cytokine signaling-3 (SOCS-3) and

endoplasmic reticulum (ER) stress in the hypothalamus. Low leptin, leptin deficiency (ob/ob

mice), and/or leptin receptor deficiency (db/db mice) results in overfeeding, suppression of

energy expenditure, suppression of triiodothyronine (T3) thyroid hormone levels, decreased

testosterone and leutenizing hormone pulsatility, and a reduction in the number of CD4+ T cells

(Maury and Brichard 2010; Chan et al. 2003; Farooqi et al. 2002). Patients with lipodystrophy

have low circulating leptin levels and this results in severe insulin resistance, hyperinsulinemia,

hyperglycemia, and an enlarged fatty liver; these symptoms are ameliorated with exogenous

leptin supplementation in patients with lipodystrophy (Oral et al. 2002).

Leptin receptors have been detected in human umbilical vein endothelial cells (HUVECs) and

within human aortic atherosclerotic plaques by immunohistochemistry (Park et al. 2001; Sierra-

Honigmann et al. 1998). Increased leptin in humans is associated with impaired arterial

distensibility measured by vascular ultrasound in healthy adolescents aged 13-16 years, and this

result is independent of fat mass, blood pressure, C-reactive protein (CRP), fasting insulin, or

LDL cholesterol concentrations (Singhal et al. 2002). Leptin (10 ng/ml) induces expression of

pro-inflammatory markers such as NF-B and MCP-1, as well as the generation of ROS in

cultured HUVECs and bovine aortic endothelial cells (BAECs) possibly by increasing fatty acid

oxidation by stimulation of carnitine palmitoyltransferase-1 (CPT-1) activity and inhibition of

acetyl-CoA carboxylase (ACC) activity as shown in BAECs treated with 10 ng/ml leptin

(Bouloumie et al. 1999; Yamagishi et al. 2001). One report showed that recombinant murine

leptin administration (125 g) for 4 weeks in 16-week-old Apoe-/-

mice significantly enhanced

Page 31: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

20

atherosclerotic lesion development despite significant reductions in body weight (Bodary et al.

2005). It is thus postulated that hyperleptinemia may be atherogenic (Ritchie et al. 2004).

1.3.4.2 Resistin

Resistin is a 114-amino-acid protein that was originally shown to induce insulin resistance in

mice (Tilg and Moschen 2006). Resistin synthesis in mice occurs only in adipose tissue;

whereas in humans, adipocytes, muscle, pancreatic cells and macrophages also synthesize the

protein. Resistin expression increases when induced by pro-inflammatory cytokines and

lipopolysaccharide (LPS). Resistin is thought to be atherogenic (Tedgui and Mallat 2006) as it

stimulates synthesis of pro-inflammatory cytokines TNF-, interleukin-1 (IL-1), IL-6 and

interleukin-12 (IL-12) through NF-B in various cell types. It also upregulates VCAM-1,

ICAM-1 and MCP-1 in human ECs. Lastly, macrophages infiltrating human atherosclerotic

aneurysms secrete resistin (Tilg and Moschen 2006).

1.3.4.3 Interleukin-6

IL-6 is a cytokine produced by several cells (fibroblasts, ECs, monocytes, SMCs, T cells and

adipocytes) and adipose tissue contributes about 15-35% of the systemic IL-6 in humans (Maury

and Brichard 2010; Tedgui and Mallat 2006). IL-6 levels predict future coronary artery disease

(Harris et al. 1999) and have been shown to be an independent predictor of mortality among

patients with unstable coronary artery disease (Lindmark et al. 2001). IL-6 is thought to be

implicated in insulin resistance, and wildtype mice fed a high-fat diet increase IL-6 production

by adipose tissue, which induces hepatic insulin resistance. This insulin resistance is thought to

be due to the upregulation of SOCS-3, which results in the ubiquitination of insulin receptors for

proteosomal degradation (Maury and Brichard 2010). While chronic IL-6 elevation has no effect

in muscle in vivo, acute IL-6 elevation from exercise enhances insulin action in skeletal muscle

(Maury and Brichard 2010). IL-6 secreted from visceral adipose tissue also regulates hepatic

production of acute-phase reactants such as CRP; evidence for this is the 50% higher levels of

IL-6 detected in the portal vein than the radial artery of obese subjects, which correlated with

systemic CRP (Maury and Brichard 2010). In turn, CRP also predicts cardiovascular disease

(CVD) and atherosclerotic plaque instability in humans and has been shown to enhance

Page 32: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

21

atherosclerosis in Apoe-/-

mice overexpressing human CRP (Paffen and DeMaat 2006). IL-6

protein and mRNA is detected in human atherosclerotic plaques by immunohistochemistry, and

reverse transcription and PCR amplification, respectively at 10- to 40-fold higher levels than in

non-atherosclerotic lesions (Rus, Vlaicu, and Niculescu 1996; Seino et al. 1994).

IL-6 has been shown to have both inflammatory and anti-inflammatory roles in atherogenesis.

Exogenous administration of recombinant IL-6 (5000 U) to wildtype C57BL6 mice fed a high-

fat diet (20% fat, 1.5% cholesterol), or Apoe-/-

mice on either normal chow or high-fat diets for

6 or 21 weeks (started at 3 weeks of age) increased fatty streak size at the aortic sinus 5.1-fold in

C57BL6 mice and atherosclerotic plaque area 2.4- and 1.9-fold in Apoe-/-

mice on the low-fat or

high-fat diets, respectively (Huber et al. 1999). Conversely, 1-year-old Il6-/-

Apoe-/-

mice on

normal chow have enhanced plaque formation with more calcified lesions but decreased

macrophage and leukocyte infiltration than Il6+/+

Apoe-/-

littermate control mice (Elhage et al.

2001; Schieffer et al. 2004). However, younger mice, 16-week-old Il6+/-

Apoe-/-

or Il6-/-

Apoe-/-

,

did not show differences in fatty streak lesion size (Elhage et al. 2001). Thus, the role of IL-6 in

atherosclerosis is unclear. While IL-6 can be viewed as a pro-inflammatory cytokine, it also has

anti-inflammatory actions (reviewed in (Barton 1996)). For example, continuous intravenous

administration of IL-6 in humans for 120 hours leads to an increase in IL-1 receptor antagonist

(IL-1Ra) and soluble TNF- receptors (sTNFR) (Tilg et al. 1994). IL-1Ra antagonizes the

effects of IL-1 by blocking the binding of IL-1 to cell surface receptors, thus protecting against

septic shock and inflammatory bowel disease. Soluble forms of TNF- receptors block LPS-

mediated lethality. In another experiment, i.p. administration of a sublethal dose of endotoxin (4

g/g) resulted in higher TNF- and interferon-gamma inflammatory cytokines in serum of Il6-/-

mice compared to Il6+/+

mice, and recombinant IL-6 administration to Il6-/-

mice abolished the

difference in circulating pro-inflammatory cytokines (Xing et al. 1998).

1.3.5 The Apoe-/- mouse model of atherosclerosis

Unlike humans, wildtype mice are highly resistant to developing atherosclerosis even when

given diets very high in cholesterol (Meir and Leitersdorf 2004). Also unlike humans, mice

carry most of their cholesterol in anti-atherogenic HDL, whereas humans carry 75% of

cholesterol in LDL (Jawien, Nastalek, and Korbut 2004). To circumvent these challenges, Apoe-

Page 33: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

22

deficient mice were created (Plump et al. 1992). As discussed earlier, APOE is synthesized by

the liver for the clearance of chylomicron and VLDL remnant lipoprotein particles via the

LDLR and LRP (Meir and Leitersdorf 2004). Mice deficient in APOE and fed a low fat, low

cholesterol diet exhibit much higher plasma cholesterol than wildtype control mice (494 versus

60 mg/dl); and when fed a Western diet with 0.15% cholesterol (developed by Hayek et al),

Apoe-deficient mice have extremely high levels of plasma cholesterol (1821 versus 132 mg/dl)

(Plump et al. 1992). In addition, Apoe-deficient mice have a lipid profile that is more similar to

humans: a shift for much higher cholesterol in chylomicrons, VLDL and LDL, plus a reduction

in HDL cholesterol 45% of that compared to wildtype mice (Jawien, Nastalek, and Korbut

2004). But most important of all, these mice develop atherosclerotic lesions that are similar to

human lesions (Nakashima et al. 1994). Another advantage is that lesions in these mice develop

early and spontaneously even when fed a regular chow diet. Monocytic adhesion is observed in

5-6 weeks, fatty streaks by 3 months, intermediate lesions by 15 weeks, fibrous plaques at 20

weeks, and calcification and wall thinning after 32 weeks of age (Meir and Leitersdorf 2004).

For these reasons, the Apoe-deficient mouse model has been used extensively for the study of

atherogenesis (Meir and Leitersdorf 2004). In this study, we used the Apoe-knockout mouse

model with streptozotocin-induced diabetes to study the effects of GLP-1R activation on

atherogenesis.

1.3.6 Current treatments for atherosclerosis

1.3.6.1 Lipid lowering

Therapy with 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors

(statins) has shown significant benefits for CVD (Ray, Cannon, and Braunwald 2007). HMG-

CoA reductase inhibitors inhibit the rate-limiting enzyme in cholesterol synthesis, HMG-CoA

reductase. These inhibitors thus upregulate LDLRs in hepatocytes and increase LDL uptake and

clearance from the circulation. With the fall in LDL levels, HDL levels rise slightly (Molecular

Mechanisms of Atherosclerosis 2005). They have also been discovered to exert a number of

immunomodulatory, potentially anti-inflammatory effects. HMG-CoA reductase inhibitors

inhibit interferon gamma (IFN--induced major histocompatibility complex-2 (MHC II)-

mediated T-cell activation, and monocyte adhesion molecule expression (Molecular

Mechanisms of Atherosclerosis 2005). Akt and Rho/Rho-kinase systems are also induced by

Page 34: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

23

statins, resulting in potentiation of nitric oxide (NO) by endothelial nitric oxide synthase (eNOS)

phosphorylation, inhibition of EC tissue factor, inhibition of VSMC migration, and mobilization

of endothelial progenitor cells (Molecular Mechanisms of Atherosclerosis 2005). Thus, it is not

surprising that HMG-CoA reductase inhibitors still reduce plaque burden even in the absence of

lipid lowering ("MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in

20,536 high-risk individuals: a randomised placebo-controlled trial" 2002).

Fibric acid compounds also improve the lipid profile, but they have not shown significant

benefits in the reduction of cardiovascular risk in patients with diabetes either alone (Zoungas

and Patel 2010) or in combination with HMG-CoA reductase inhibitors (Farmer 2011).

Although dietary cholesterol is an independent risk factor for atherosclerosis, the attributable

risk is low. Increasing evidence points more at cholesterol-rich post-prandial chylomicron

remnants being most atherogenic, where dietary cholesterol contributes only about one third,

and the rest is derived endogenously from de novo synthesis (Huff 2003). To address the

intestinal pathway, the drug ezetimibe was developed, which inhibits carrier-mediated

cholesterol absorption in the intestine. Despite ezetimibe’s effect on LDL-cholesterol lowering,

its effect on atherosclerosis remains indeterminate. The ENHANCE (Ezetimibe and Simvastatin

in Hypercholesterolemia Enhances Atherosclerosis Regression) trial consisted of 720 patients

with familial hypercholesterolemia treated with 80 mg simvastatin (an HMG-CoA reductase

inhibitor) plus either placebo or 10 mg of ezetimibe given daily for 2 years. The ENHANCE

trial assessed carotid and femoral intima-media thickness by ultrasonography after 24 months

and found a trend suggesting a negative effect of ezetimibe treatment as the mean intima-media

thickness of the carotid and femoral arteries showed a trend toward greater thickness (0.0033

mm for simvastatin monotherapy versus 0.0182 mm for simvastatin plus ezetimibe therapy, P =

0.15) (Guyton 2010).

1.3.6.2 Hypertension

Therapeutic options for the treatment of hypertension include several major classes of drugs:

diuretics, β-adrenoceptor antagonists (β-blockers), angiotensin-converting enzyme (ACE)

inhibitors, angiotensin II type 1 receptor antagonists (ARBs), renin inhibitors, calcium channel

blockers, and central sympatholytics, alone or in combination (Chrysant 2010). Patients with

Page 35: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

24

diabetes often have elevated blood pressure (BP) that adds an additional risk factor for

accelerated atherosclerosis and cardiovascular events (Taylor and Bakris 2010). ACE inhibitors

reduce the risk of coronary events by regulation of blood pressure. These drugs regulate blood

pressure by reducing the activity of components of the renin-angiotensin-aldosterone system

(RAAS). It has been suggested that these drugs not only have effects on lowering blood

pressure, but may also have other effects that might help further reduce progression of

atherosclerosis (Koh et al. 2010). For example, the ARB drug candesartan used to treat

hypertension also improves markers of oxidative stress, inflammation, and fibrinolysis

independent of BP lowering effects; losartan, another ARB, decreases myocardial collagen

content and corrects the altered endothelial structure and resistance of arteries; and valsartan,

another ARB, has BP-independent effects on left ventricular hypertrophy, reactive oxygen

species formation by monocytes, and C-reactive protein in hypertensive patients with left

ventricular hypertrophy when compared with amlodipine, a calcium channel blocker also used

to treat hypertension (Koh et al. 2010).

1.3.6.3 Diabetes

Despite current therapies to lower lipids and blood pressure, there remains a significant event

rate for cardiovascular disease especially in patients with diabetes and/or insulin resistance

(Rubenfire, Brook, and Rosenson 2010; Tenenbaum et al. 2006). Patients with diabetes often

present with more risk for atherosclerosis; obesity, dyslipidemia, hypertension, hyperglycemia

and a chronic inflammatory state may all be present in diabetes (Taylor and Bakris 2010).

Simultaneous management of diabetes and other risk factors for atherosclerosis that are usually

present in patients with diabetes requires administration of several drugs and makes control

challenging. Treatment is often associated with side effects and further progression to

developing type 2 diabetes (Taylor and Bakris 2010). For example, less than 33% of patients

with diabetes treated for hypertension attained controlled levels of BP (Taylor and Bakris 2010).

In this regard, GLP-1R agonists may hold the key to further reducing the rate of cardiovascular

events. GLP-1R agonists have been shown to reduce body weight, blood pressure and improve

lipid profile, all of which contribute to a reduction in atherosclerosis (Rizzo et al. 2009). In

addition, the GLP-1R agonist liraglutide reduced infarct size in experimental murine myocardial

infarction (Noyan-Ashraf et al. 2009), and the GLP-1R agonist exenatide was associated with a

Page 36: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

25

reduced incidence for cardiovascular disease events in a retrospective analysis of type 2 diabetic

patients treated with exenatide twice daily (hazard ratio 0.81) (Best et al. 2011). It remains to be

shown, however, whether GLP-1R agonists can exert direct effects on atherogenesis in the

setting of diabetes.

1.4 GLP-1R actions on atherogenesis

Atherogenesis is influenced by many indirect factors such as lipoprotein concentrations, blood

pressure, visceral adiposity, insulin resistance and hyperglycemia which do not directly act on

the plaque but influence its progression and/or regression. Atherosclerosis also involves the

interplay of many cell types (neutrophils, monocytes, T cells, B cells, DCs, mast cells, ECs and

smooth muscle cells (SMCs)) (Weber and Noels 2011). In this section, the indirect and direct

actions of GLP-1R agonists on atherogenesis are discussed.

1.4.1 Indirect actions

1.4.1.1 Lipoprotein metabolism

The effects of atherogenic plasma LDL and anti-atherogenic HDL in atherogenesis are well-

known (Weber and Noels 2011). Several lines of evidence show that activation of the GLP-1R

in either rats (Qin et al. 2005), mice (Hsieh et al. 2010; Parlevliet et al. 2009), hamsters (Hsieh

et al. 2010) or humans (Koska et al. 2010; Matikainen et al. 2006; Meier et al. 2006; Tremblay

et al. 2011) results in a decrease in postprandial lipids. In adult rats, GLP-1(7-36)NH2 infusion

through the jugular vein (20 pmol/kg/min) with simultaneous direct infusion of lipids (3 ml/h)

into the duodenum of lymph duct-cannulated rats reduced triglyceride absorption as measured

by lymphatic triolein recovery and levels of lymphatic APOB lipoproteins after overnight

fasting (Qin et al. 2005). In hamsters and mice, treatment with the DPP-4 inhibitor sitagliptin

both acutely (10 g/kg) and chronically (5 g/kg for 2 to 3 weeks), resulted in marked

reductions in APO-B48-containing lipoproteins after olive oil gavage followed by Triton

WR1339 (0.5 g/kg) administration to inhibit peripheral and hepatic lipase. This reduction in

APO-B48-containing lipoproteins after olive oil gavage was abolished in GLP-1R knockout

mice (Hsieh et al. 2010). An ex vivo experiment using cultured chow-fed hamsters’ enterocytes

Page 37: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

26

showed exendin-4 (100 pM) inhibited the secretion of newly-synthesized APO-B48 (Hsieh et al.

2010). In the absence of lipase inhibition by Triton, 8-week vildagliptin treatment (another DPP-

4 inhibitor) (1 mol/ml supplied in drinking water) also resulted in a significant reduction in

plasma triglycerides and cholesterol in high-fat-fed (45 kcal%) male C57BL6 wildtype mice

after refeeding from an overnight fast with a 45 kcal% high-fat diet (Flock et al. 2007). In

healthy (male) humans, intravenous administration of GLP-1(7-36)NH2 (1.2 pmol/kg/min) over

390 minutes (-30 to 360 min) with a mixed test meal (250 kcal) ingested at 0 min was shown to

abolish the rise in plasma triglycerides (area under the triglyceride concentration curve for

placebo, 7331±1850 mg dl-1

min and GLP-1, 1263±436 mg dl-1

min) and non-esterified fatty

acids (NEFA) (39% lower in the fasting state and 31±5% lower throughout the meal test);

however, these effects are in part due to delayed gastric emptying for GLP-1-treated individuals

versus placebo (50% of the initial content was still in the stomach compared to only 20% for

placebo) (Meier et al. 2006). In patients with recent-onset T2DM (<3 years), acute subcutaneous

administration of exenatide (10 g) also reduced plasma triglycerides (3.8 mM for placebo

versus 2.5 mM after exenatide) while improving endothelial function (measured by peripheral

arterial tonometry or PAT index; PAT index was higher in the exenatide group than placebo)

after a high-fat meal (45% fat) (Koska et al. 2010). To circumvent the effects of gastric

emptying and other hormones or FFAs on lipoprotein metabolism, a recent study in healthy

humans (15 males) administered a liquid formula (49 kcal% from fat) directly into the

duodenum through a nasoduodenal tube, and showed exenatide (10 g subcutaneous)

suppressed intestinal lipoprotein particle production (24-37% decrease in triglyceride-rich

lipoprotein (TRL)-APOB-48, and a 38% reduction in TRL-APOB-48 production rate) during

constant infusion of somatostatin during a standard pancreatic clamp (Xiao et al. 2012). Finally,

treatment with the DPP-4 inhibitors vildagliptin (50 mg twice daily for 4 weeks) or sitagliptin

(100 mg/day for 6 weeks) in T2DM patients decreased post-prandial (over 8 hours) APOB-

containing lipoproteins of both intestinal and hepatic origin (5.1 to 7.8% APOB reduction)

(Matikainen et al. 2006; Tremblay et al. 2011). In short, while a molecular mechanism of action

is currently not known, these data suggest that, in general, GLP-1R activation reduces intestinal

lipoprotein secretion independent of gastric emptying, which may lead to reductions in TRL-

APOB-48 lipoprotein particles.

Page 38: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

27

1.4.1.2 Blood pressure

Hypertension promotes atherogenesis and is a risk factor for cardiovascular events (Zanchetti et

al. 1998). Studies in animals have shown that acute activation of the GLP-1R increases BP

(Ussher and Drucker 2012). Chronic treatment with agonists for the GLP-1R and DPP-4

inhibitors in rodents and humans, however, results in reductions in BP that can be observed

before significant weight loss is recorded (Ussher and Drucker 2012).

1.4.1.3 Adiposity and adipokines

The role of select adipokines in atherogenesis was discussed in section 1.3.4. Here, current

findings on how GLP-1R activation affects adipokine secretion and/or action is reviewed.

Treatment in both animals (Li et al. 2008) and humans (Bunck et al. 2010) with GLP-1R

agonists often results in a decrease in body weight with changes in adipokine expression. In

humans (Bunck et al. 2010), rats (Li et al. 2008) and mice (Samson et al. 2008), plasma

adiponectin has consistently been shown to be increased in vivo with exendin-4 treatment. In

vitro, 3T3-L1 adipocytes cultured with 2.5 or 5 nM exendin-4 for 8 hours also showed an

increase in adiponectin mRNA (2-fold) and protein in media (by 20%) (Kim Chung et al. 2009).

The decrease in adiponectin levels in the setting of obesity has been associated with insulin

resistance and cardiovascular risk (Kim et al. 2007; Menzaghi, Trischitta, and Doria 2007; Hotta

et al. 2000; Pischon et al. 2004); however, in vivo studies in mice have not shown a direct role of

adiponectin in atherogenesis (Nawrocki et al. 2010).

Exendin-4 treatment of 3T3-L1 adipocytes also decreased protein expression of IL-6 and MCP-

1 (Kim Chung et al. 2009). Furthermore, clinical studies have revealed a consistent decrease in

CRP after chronic treatment with exenatide in patients with T2DM, although plasma IL-6,

MCP-1 and resistin levels were unaffected (Bunck et al. 2010; Horton et al. 2009; Courrèges et

al. 2008). The observed reductions in CRP, however, could be due to the significant decrease in

body weight. A recent study, however, examined the effects of 12 weeks of treatment with

exenatide in T2DM patients. These patients did not lose weight during this period, yet showed a

marked reduction in plasma concentrations of MCP-1, matrix metalloproteinase-9 (MMP-9),

serum amyloid A (SAA), and IL-6 (Chaudhuri et al. 2012). It is thus not clear whether

Page 39: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

28

activation of the GLP-1R has a direct role in vivo in the expression and secretion of IL-6 and

MCP-1 from adipocytes.

Long-term GLP-1R activation (15 weeks) by constitutive expression of exendin-4 through a

helper-dependent adenoviral vector expressed in liver did not change plasma leptin levels in

C57BL6 mice fed a 42 kcal% high-fat diet (Samson et al. 2008). In T2DM patients, liraglutide

treatment for 14 weeks did not change plasma leptin levels despite significant reductions in

body weight (Courrèges et al. 2008). On the other hand, reductions in resistin levels were seen

in mice treated with constitutive expression of exendin-4 through a helper-dependent adenoviral

vector in liver, but the reduction was linearly correlated to decreases in body fat (Samson et al.

2008).

Thus, current evidence suggests that the overall effect of activation of the GLP-1R on

adipokines appears to be indirect through the loss of adipocyte mass due to the anorectic effects

of the GLP-1R in the brain. Reductions in plasma CRP from GLP-1R activation also occur, but

a clear direct role of CRP in atherogenesis is currently lacking; there is only data on the

association of CRP with CVD and plaque instability (Paffen and DeMaat 2006).

1.4.1.4 Insulin resistance, hyperinsulinemia and hyperglycemia

The contributions of insulin resistance, hyperinsulinemia and hyperglycemia to atherogenesis

were discussed in section 1.3.2. Briefly, insulin resistance (decreased insulin signaling) in

endothelial cells and macrophages resulted in marked increases in atherogenesis in mice (Rask-

Madsen et al. 2010; Seongah et al. 2006); whereas hyperinsulinemia did not alter atherogenesis

(Rask-Madsen et al. 2012). Furthermore, clinical evidence pointed to moderate effects of

improved glucose control in the reduction of cardiovascular events in the UKPDS trial after 10

years of treatment (Stratton et al. 2000).

Activation of the GLP-1R in pancreatic -cells stimulates glucose-dependent insulin secretion to

promote glucose homeostasis (Baggio and Drucker 2007; Drucker et al. 1987; Lamont et al.

2012). The improved blood glucose control from GLP-1R activation may have modest effects

on reducing atherogenesis as proved by the UKPDS (Stratton et al. 2000), whereas the increased

Page 40: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

29

insulin secretion may not play an important role as proved in hyperinsulinemic mice prone to

atherosclerosis (Rask-Madsen et al. 2012).

While the GLP-1R is not expressed in muscle, fat or liver, studies suggest prolonged GLP-1R

activation may improve insulin sensitivity. Non-diabetic Zucker fa/fa rats treated with exenatide

(3 g/kg twice daily) for 6 weeks had a 61% increase in insulin sensitivity index (ISI—glucose

infusion rate to plasma insulin concentration) as assessed in a hyperinsulinemic euglycemic

clamp compared to pair-fed controls (Gedulin et al. 2005). Similarly, infusion of GLP-1(7-

36)NH2 (4.8 pmol/kg/min) for 6 weeks in T2DM patients improved the insulinogenic index

(area under the curve, AUC insulin to AUC glucose) during a hyperinsulinemic euglycemic

clamp (Zander et al. 2002). Nonetheless, increased insulin sensitivity by GLP-1R activation is

controversial; GLP-1 (1.2 pmol/kg/min) or exendin-4 (0.12 pmol/kg/min) in eight healthy

subjects failed to show changes in glucose infusion rate, glucose disappearance or endogenous

glucose production during a hyperinsulinemic euglycemic clamp with infusion of somatostatin

(120 ng/kg/min) to inhibit endogenous insulin, while maintaining growth hormone (3

ng/kg/min) and glucagon (0.65 ng/kg/min) basal levels (Vella et al. 2002). It is likely that

studies showing a positive effect on insulin sensitivity are due to a period of improved glucose

control due to glucose-stimulated insulin secretion from GLP-1R activation. In turn, improved

glucose control independently improves insulin sensitivity of peripheral tissues (Yki-Jarvinen

1990, 1997). Thus, while GLP-1R activation does not directly modulate insulin sensitivity, it

may do so indirectly in diabetic patients through improvements in glucose homeostasis. Thus,

improved glucose control through GLP-1R activation may indirectly ameliorate insulin

resistance and thus, reduce the accelerated development of atherosclerosis in diabetic patients.

1.4.2 Direct actions

1.4.2.1 Endothelial cells

In vitro studies: Activation of the GLP-1R by liraglutide (0.01 to 100 g/ml) in HUVECs

induced NO formation dose-dependently through eNOS phosphorylation and suppressed high

glucose- (27.5 mM) or TNF--mediated (10 ng/ml) activation of NF-B and I-kappaB (IB)

degradation. HUVECs treated with liraglutide (1 g/ml) for 24 hours also reduced TNF--

induced upregulation of MCP-1, VCAM-1, ICAM-1 and E-selectin mRNA (Hattori et al. 2010).

Page 41: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

30

In human coronary artery endothelial cells (HCAECs) incubated at euglycemic conditions (5

mM glucose), 1-10 nM exendin-4 treatment for 48 hours also resulted in a greater than 2-fold

phosphorylation of eNOS at Ser-1177 (activating eNOS) by a PI3K-Akt-dependent pathway,

and an increase in eNOS protein expression (Erdogdu et al. 2010). The study also showed that

incubation with 1 nM exendin-4 for 48 hours increased the rate of [3H]thymidine incorporation

to 190% and 10 nM further augmented the rate to 264%. The proliferation of HCAECs was

through Akt and MAPK phosphorylation, and the proliferative effects were abolished with

PI3K, Akt, PKA or eNOS inhibitors (Erdogdu et al. 2010). Another study, however, found no

Akt phosphorylation in HUVECs treated with 10 nM GLP-1, although cAMP response element

binding (CREB) phosphorylation was increased by 52% in a PKA-dependent manner (effect

was abolished by PKA inhibition) (Oeseburg et al. 2010). The same group treated 10-week-old

ZDF rats with the DPP-4 inhibitor vildagliptin (3 mg/kg/day) for 15 weeks which resulted in a

6-fold increase in plasma GLP-1 levels and reduced senescence of abdominal aortic endothelial

cells as assessed by -galactosidase staining (Oeseburg et al. 2010).

Incubation of HUVECs with 0.03-0.3 nM GLP-1(7-36)NH2 for 4 hours resulted in inhibition of

AGE-induced ROS generation and the inhibition of AGE-induced VCAM1 mRNA upregulation;

incubation of HUVECs with 0.3 nM GLP-1 for 4 hours also downregulated mRNA levels of the

receptor for AGEs (RAGE) (Ishibashi et al. 2010). In the spontaneously-transformed C11-STH

human umbilical vein endothelial cell line, incubation with 100 nM liraglutide for 1 hour

inhibited TNF--induced (10 ng/ml) or hyperglycemia-induced (10 M) upregulation of PAI-1,

ICAM-1 and VCAM-1 mRNA and protein (Liu et al. 2009). Furthermore, incubation of C11-

STH endothelial cells with 100 nM liraglutide incubated with 10 ng/ml TNFfor 16 hours

inhibited TNF-mediated induction of ICAM-1 and VCAM-1 mRNA and protein and NFkB

mRNA expression, which was independent of PKA signaling as use of the PKA inhibitor H89

(10 M) did not attenuate this effect (Gaspari et al. 2011).

In vivo studies: Apoe-/-

mice fed a 22% fat and 0.15% cholesterol diet for 12 weeks and

subsequently treated twice daily with 80 nmol/kg liraglutide for four weeks show significant

improvements in endothelial function as assessed by endothelial-dependent vasodilation of

abdominal aortic rings (using acetylcholine) pre-constricted ex vivo with a thromboxane A2

analogue (Rmax = 78.45% versus 55.36% in controls) (Gaspari et al. 2011). In addition, eNOS

and ICAM-1 were shown to be upregulated by liraglutide by immunohistochemical analysis in

Page 42: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

31

intermediate aortic cross-sections compared to control mice (Gaspari et al. 2011). Activating the

GLP-1R in pulmonary artery rings isolated from male Sprague-Dawley rats also showed a dose-

dependent relaxation with GLP-1(7-36)NH2 (10-8

to 10-5

M) when arteries were pre-constricted

using 10-7

M norepinephrine (16.2% of maximal relaxation by acetylcholine) (Richter et al.

1993; Golpon et al. 2001).

In healthy non-diabetic humans, GLP-1 infusion improved acetylcholine-induced vasodilation

and forearm blood flow measured by venous occlusion plethysmography (9.1 versus 6.5

ml/100ml/min) (Basu et al. 2007). Similarly, in T2DM patients with established coronary artery

disease (CAD), a hyperinsulinemic euglycemic clamp with GLP-1(7-36)NH2 infusion (2

pmol/kg/min) increased brachial artery flow-mediated vasodilation (FMD) (6.6% versus 3.1%)

(Nyström et al. 2004). A third study with both healthy and T2DM patients infused with GLP-

1(7-36)NH2 at 0.4 pmol/kg/min (to reach physiological postprandial GLP-1 levels) also showed

improvements in flow-mediated vasodilation in both groups during a hyperglycemic clamp

(healthy subjects, 9.6% versus 7.0%; T2DM subjects, 3.8% versus 0.5%) (Ceriello et al. 2011).

While these experiments were conducted using the native GLP-1 peptide, DPP-4-resistant

exenatide (0.03 pM to 0.3 M) did not induce vasodilation or cyclic guanine monophosphate

(cGMP) release from mouse mesenteric arteries pre-constricted with 3 M phenylephrine ex

vivo. However, GLP-1(7-36)NH2 and the truncated peptide GLP-1(9-36) did induce vasodilation

and cGMP release (Ban et al. 2008). A similar result in male Sprague-Dawley rats’ femoral

arteries demonstrated that exenatide did not exert an effect after infusion with intralipid, but

GLP-1 and GLP-1(9-36) produced concentration-dependent vasorelaxation after pre-

constriction with phenylephrine (10-5

M) (Nathanson et al. 2009). Thus, it is not clear whether

vasodilatory effects can be mediated by degradation-resistant GLP-1R agonists as current data

suggests the effects are predominantly mediated by the truncated GLP-1(9-36) metabolite.

1.4.2.2 Vascular smooth muscle cells

VSMCs are important in both early and late stages of atherosclerosis (Doran, Meller, and

McNamara 2008). GLP-1R binding sites are known to be expressed in rat pulmonary VSMCs as

detected by incubation of pulmonary arteries with 125

I-GLP-1(7-36)NH2 and slide-mount

autoradiography (Richter et al. 1993). The receptor was also shown to be expressed by Western

Page 43: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

32

blotting using the GLP-1R antibody from Abcam using extracts from murine aortic, rat and

human primary coronary artery SMCs (Goto et al. 2011). Isolated and cultured murine VSMCs

also express the GLP-1R as detected using the same antibody from Abcam (Arakawa et al.

2010). Little is known about the role of the GLP-1R in VSMCs. It has been reported that

exendin-4 treatment for 4 weeks (24 nmol/kg/day infused through an osmotic pump) reduces

neointimal formation (by about 50% and intima/media ratio by about 3-fold) in 9-week-old

C57BL6 mice subjected to endothelial denudation at the femoral artery (Goto et al. 2011). Ten-

week-old female ZDF rats subjected to balloon catheter injury in the carotid artery and

descending aorta showed a marked reduction in intimal hyperplasia when treated with exendin-4

(5.0 g/kg for one week prior to injury, then resuming exendin-4 treatment for another 22 days

after injury; intima/media ratio = 1.0 in controls versus 0.2 in exendin-treated rats). Examination

by Western blotting of the aortic tissue revealed no differences in the level of total eNOS

(endothelial nitric oxide synthase), but did show a trend for decreased protein levels of NFB

(0.11 versus 0.045 density units) (Murthy et al. 2010). In vitro, platelet-derived growth factor

(PDGF)-induced proliferation (measured by BrdU incorporation) of cultured VSMCs of the

mouse aorta was inhibited with 12-hour pre-incubation with 10 nM exendin-4. The inhibitory

effect by exendin-4 was not due to changes in intracellular cAMP, phosphorylation of mitogen-

activated protein kinase (MAPK), Akt, cAMP response element-binding (CREB) or

P70S6kinase (a serine/threonine kinase that phosphorylates S6 ribosome to induce protein

synthesis) as Western blotting did not reveal significant changes from control (Goto et al. 2011).

1.4.2.3 Monocytes and monocyte-derived cells

The GLP-1R has been shown to be expressed by Western blotting in human circulating

monocytes, in the THP-1 monocyte cell line, and in murine peritoneal macrophages using the

antibody from Abcam (Kodera et al. 2011; Arakawa et al. 2010). The GLP-1R was also shown

to be present in atherosclerotic lesions of Apoe-/-

mice using fluorescent staining with the GLP-

1R antibody from MBL International (LS-A1205) (Arakawa et al. 2010). Detection of the GLP-

1R using today’s commercially-available antibodies or the use of RT-PCR, however, is

controversial (please refer to section 4.5 for an expanded explanation). In vitro treatment of

human monocytes with 0.3-3 nM exendin-4 reduced CD11b surface receptors (which bind

ICAM-1) as assessed by flow cytometry (Arakawa et al. 2010). In cultured THP-1 monocyte

Page 44: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

33

cells, exendin-4 was shown to reduce the mRNA expression (by RT-PCR) and protein secretion

(by ELISA on supernatant) of TNF- and IL-1 (Kodera et al. 2011). In thioglycolate-induced

peritoneal macrophages from wildtype C57BL6 mice, 0.03-3 nM exendin-4 pre-treatment for 1

hour reduced 10 g/ml LPS-induced expression of MCP-1 and TNF- through adenylate

cyclase/cAMP/PKA pathway as explored by use of adenylate cyclase and PKA inhibitors or

activators (Arakawa et al. 2010). Cholesteryl ester accumulation in peritoneal macrophages

from Apoe-/-

mice incubated with GLP-1(7-36) was reduced compared to control in association

with reduced protein levels of CD36 (a scavenger receptor) and ACAT-1 (a cholesteryl ester

transferase) (Nagashima et al. 2011). THP-1 monocyte-derived macrophages also express the

GLP-1R as shown by RT-PCR and Western blotting using the antibody from Santa Cruz

Biotechnology (Matsubara et al. 2012). Treatment with GLP-1 (10 pM) in THP-1 cells reduced

LPS-induced (10 ng/ml) IL-6 production through inhibition of LPS-induced extracellular-signal-

regulated kinase-1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), and increased cAMP as

assessed with MDL-12330A adenylyl cyclase inhibitor (5 M) and H89 PKA inhibitor (5 M)

(Matsubara et al. 2012).

1.4.2.4 Plaque formation

The effect of GLP-1R activation on plaque development has been evaluated in non-diabetic

mice. The first report identified a moderate reduction in atherogenesis in association with

reduced monocyte adhesion to the endothelium as assessed by en face immunohistochemical

staining using MAC-2 antibody on the thoracic aorta after 28-day continuous infusion of

exendin-4 with an osmotic pump (24 nmol/kg/day) in both wildtype C57BL6 and Apoe-/-

mice

(wildtype, 7 versus 23 cells/mm2; Apoe

-/-, 23 versus 46 cells/mm

2) (Arakawa et al. 2010). Plaque

size at the aortic sinus (stained with oil red O) of Apoe-/-

mice treated with exendin-4 was

significantly reduced, as was Icam1 and Vcam1 mRNA expression in the thoracic aorta

(Arakawa et al. 2010). Continuous infusion through implanted mini-osmotic pumps of active

GLP-1(7-36)NH2 for 4 weeks at 2.2 nmol/kg/day in Apoe-/-

mice aged 17 weeks fed an

atherogenic diet (30% fat, unknown amount of cholesterol) for 4 weeks resulted in markedly

reduced plaque size and macrophage infiltration in cross-sections of the aortic root (ORO and

MOMA-2 antibody staining, respectively), and reduced lesion size in the entire aorta (from root

to abdominal area) compared to control and independent of cholesterol levels (Nagashima et al.

Page 45: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

34

2011). Furthermore, co-infusion of GLP-1(7-36)NH2 with exendin(9-39) antagonist (22

nmol/kg/day) did not reduce plaque size in the entire aorta (including the aortic root) nor

reduced macrophage infiltration beyond that of control (Nagashima et al. 2011).

1.5 Project rationale and hypothesis

While the effects of activating the GLP-1R on plaque formation have been examined in non-

diabetic mice, it is currently not known whether the effects of activating the GLP-1R on

reduction of plaque development and monocyte recruitment will remain in the presence of

diabetes. The focus of this thesis is to examine the effects of activation of the GLP-1R by two

agonists, exendin-4 and taspoglutide, on atherogenesis in glucose intolerant and diabetic Apoe-/-

mice.

The main reason for choosing the Apoe-/-

mouse model over the Ldlr-/-

mouse model (another

well-known mouse model for atherosclerosis) is to avoid the need to utilize a high-cholesterol

diet for inducing atherosclerosis. Ldlr-/-

mice have physiological levels of cholesterol unless

cholesterol is supplemented exogenously in the diet. As mentioned in section 1.3.5, mice are

highly resistant to developing atherosclerosis unless hypercholesterolemia is induced (Meir and

Leitersdorf 2004). Apoe-/-

mice, on the other hand, are hypercholesterolemic from birth and

develop atherosclerosis spontaneously even when fed a regular chow diet (Meir and Leitersdorf

2004). Because GLP-1R activation was shown to inhibit intestinal lipoprotein secretion (Qin et

al. 2005; Hsieh et al. 2010), we wanted to avoid the effect of cholesterol lowering on

atherosclerosis lesion size in order to be able to examine the more direct actions of GLP-1R

activation in atherosclerosis.

Hypothesis: We predict that activation of the GLP-1R in glucose intolerant and diabetic Apoe-/-

mice will slow the progression of atherosclerosis, similar to that shown in non-diabetic mice

(Arakawa et al. 2010; Nagashima et al. 2011).

Page 46: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

35

Chapter 2 Materials and methods

All experimental procedures on animals conformed to the guidelines and policies approved by

the Mount Sinai Hospital Animal Care Committee. Mice were maintained on a 12-hour light

(7:00 A.M.)/dark (19:00) cycle in a pathogen-free environment and had ad libitum access to food

and water, except where noted. Two sets of experiments were carried out and will be

categorized as the “exendin-4 project” and the “taspoglutide project”, where exendin-4 and

taspoglutide are the GLP-1R agonists being tested in each project.

2.1 Exendin-4 project

2.1.1 Animals, diets and drug treatments

Six-week-old male Apoe-/-

mice were purchased from Jackson Laboratory (Stock Number

002052, Bar Harbor, ME) and housed at the animal facility of the Toronto Centre for

Phenogenomics at Mount Sinai Hospital. Upon arrival, mice were acclimatized for one week

and fed a standard chow diet with 18% of calories from fat (2018S, Harlan Teklad Laboratories,

Mississauga, Ontario, Canada) before taking basal measurements. At 8 weeks of age, all mice

were started on a 60 kcal% high-fat diet (HFD) with 0.003% by weight cholesterol (D12492,

Research Diets, New Brunswick, NJ). The reason for choosing this diet low in cholesterol and

not the Western diet with 0.15% cholesterol was because exendin-4 is known to inhibit

intestinal lipoprotein secretion (Hsieh et al. 2010); development of atherosclerosis in Apoe-/-

mice is dependent on hypercholesterolemia (Whitman 2004) and the possibility for a reduction

in excess dietary cholesterol by exendin-4 treatment would be the main factor driving lesion

size. We were more interested in the effects of exendin-4 treatment in atherosclerosis

independent of changes in lipid parameters. The other reason for choosing a high-fat diet was in

order to render the mice glucose intolerant and hyperinsulinemic, two important aspects that are

observed in insulin-resistant T2DM patients. It had been reported that male Apoe-/-

mice fed this

diet for 17 weeks exhibit obesity, glucose intolerance, inflammation and increased aortic sinus

plaque size with increased levels of serum amyloid A without inducing changes in lipoprotein

profile or adipokines (King et al. 2010).

Page 47: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

36

Mice were divided into 3 groups and treatments started at 8 weeks of age (N = 8 per group, but

staggered in time into two groups with N = 4). Group 1 was administered PBS (i.p.) twice a day

(9:00 and 17:00); group 2 was treated with metformin supplied in food (sterilized and premixed

by Research Diets) (225 mg/kg/day; food intake was measured weekly per cage and the

metformin diet was appropriately diluted with the 60 kcal% control diet using a blender to

match the dose) and also injected with PBS twice daily; and, group 3 was administered 10

nmol/kg/day exendin-4 (i.p.) dissolved in PBS and injected twice daily (5 nmol/kg per

injection). The reason for having the metformin group in the study was to have a control for the

glucose-lowering effect of exendin-4. Mice were treated for 22 weeks and euthanized by carbon

dioxide asphyxiation.

2.1.2 Metabolic measurements

Body composition. Total body fat and lean mass were measured using a mouse whole-body

magnetic resonance analyzer (Echo Medical Systems, Houston, TX) after 5 and 10 weeks of

treatment.

Oral glucose tolerance tests and measurement of plasma insulin levels. Oral glucose tolerance

tests (OGTTs) were carried out 5, 9, and 22 weeks after the start of treatments following a 6-

hour fast (7:00-13:00). No drugs were administered in the morning of the day of the test.

Glucose (1.5 mg/g of body weight) was administered orally through a gavage tube. Blood was

drawn from the tail vein at 0, 10, 20, 30, 60, 90, and 120 min after glucose administration, and

blood glucose levels were measured by the glucose oxidase method using the Contour

glucometer (Bayer Healthcare, Toronto, Ontario, Canada). For plasma insulin determinations,

blood samples (50 μl) were drawn from the tail vein at the 0- and 10-minute time points

following glucose administration in a heparinized tube. Blood was mixed with 10% (v/v) TED

(500,000 IU/ml Trasylol, 1.2 mg/ml EDTA, and 0.1 mM Diprotin A) and plasma separated by

centrifugation at 4 °C and stored at –80°C until assayed. Plasma was assayed for insulin using a

mouse insulin ELISA kit (Alpco).

Systolic blood pressure. Systolic blood pressure was measured in awake mice using an

automated tail cuff system (BP-2000, Visitech Systems, Apex, NC) after 11 weeks of treatment.

Measurements were taken twice in order to train the mice on the apparatus the first day, and

Page 48: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

37

measurements from the second day were recorded for analysis. During measurements, mice

were restrained in a dark chamber, in which the floor was heated to 38°C. Results from the first

10 inflation cycles were discarded, and the average obtained from the next 10 cycles was

recorded. Mice were in the chamber for about 15 min. On each study day, systolic blood

pressure and heart rate were measured between 10:30–15:30. While the apparatus measured

heart rate and diastolic blood pressure values, it failed to do so accurately and consistently (a

high variability in heart rate and erroneous values were observed; for example, diastolic blood

pressure was very frequently not measured and reported by the apparatus, and heart rates varied

from 70 to 900 beats per minute in repeated measurements).

2.1.3 Histological analysis of atherosclerotic lesions

The aortic root was isolated for quantification of atherosclerosis lesion size as described

(Daugherty and Whitman 2003) with the following modifications: the aorta was flushed with

cold PBS (about 8-10 ml) by perfusion with a needle syringe inserted into the left ventricle of

the heart. Prior to perfusion, the right atrium was nicked. The aorta was dissected while attached

to the heart and a scalpel was used to cut out two thirds of the bottom ventricles. The aortic arch

region was cleared of periadventitial adipose tissue and other surrounding vessels under a

dissecting microscope and the aorta was cut about 1 mm above the atria. This upper part of the

heart with the aortic root was embedded in Tissue-Tek OCT Compound (Sakura Finetek,

Torrence, CA), wrapped in clear plastic wrap and frozen in liquid nitrogen. Frozen samples

were processed by Lily Morikawa from the Centre for Modeling Human Disease (CMHD), a

division of the Toronto Centre for Phenogenomics (TCP). Frozen samples were sectioned with a

cryostat with 5-m thickness and mounted on slides. The first emergence of the three valve

leaflets was taken as reference point and the 1st, 14

th and 21

st sections were taken for analysis of

plaque size (each section being 5 m apart). Slides were stained with oil red O and

counterstained with hematoxylin and eosin for determination of plaque size and lipid within the

plaque. ImageScope software version 11.0.2.716 (Aperio) was used to manually draw a

perimeter around the plaques and quantify plaque area and lipids by thresholding function

available in Aperio on the three sections. The plaque areas from all three sections were then

added. Plaques that formed directly on the leaflets of the aortic sinus were not quantified; only

plaques that developed on the aortic wall were quantified.

Page 49: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

38

2.2 Taspoglutide project

2.2.1 Animals, diets and drug treatments

Four-week-old male Apoe-/-

mice were purchased from Jackson Laboratory (Bar Harbor, ME)

and housed at the animal facility of the Toronto Centre for Phenogenomics at Mount Sinai

Hospital. Mice were acclimatized for one week before taking basal measurements. Upon arrival,

mice were immediately started on a 45 kcal% high-fat diet (HFD) (D12451, Research Diets,

New Brunswick, NJ). The reason for choosing this diet is the same as for the exendin-4 project;

this diet is low in cholesterol (unlike Western diets with 0.15% cholesterol) to avoid the

possibility for a reduction in ingested cholesterol due to taspoglutide treatment, as cholesterol is

a key mediator of atherogenesis (see section 1.5). In addition, a change from 60 to 45 kcal% was

made because the protocol to induce mild diabetes with streptozotocin (STZ, a beta-cell toxin)

(as described next) was optimized with the 45 kcal% diet by other members of our lab.

To render the mice diabetic (plasma glucose between 15-25 mM), we did not want to induce

total pancreatic beta-cell destruction, hence we employed a specific combination of diet and

STZ. After 4 weeks on the HFD, mice were given an intraperitoneal injection of 75 mg/kg STZ

in 0.05 M citrate buffer, pH 4.5, or citrate buffer alone as control. After 10 days, a second dose

with an additional 125 mg/kg STZ was administered. Plasma glucose was measured 10 days

after the second dose of STZ between 9-10 A.M. and mice in the desired range (15-25 mM) were

used in the study. This procedure rendered, as expected, 40-50% of mice in the desired glucose

range and only these mice were selected for the study. Insulin supplementation was not required

for these mice.

Mice were divided into 3 groups and treatments started at 13 weeks of age (n = 15 per group).

All mice were rendered diabetic with STZ. Group 1 mice were administered a placebo

microtablet subcutaneously (the test drug, taspoglutide, was formulated in the form of a

microtablet); group 2 was treated with metformin supplied in autoclaved water (water intake

was monitored weekly and concentration adjusted to correspond to a dose of 400 mg/kg/day;

this dose was chosen based on a previous study from our lab (Sauve et al. 2010)) and also given

the placebo microtablet; and, group 3 was made diabetic and administered with 0.4-mg dose of

taspoglutide microtablet subcutaneously (provided by Roche Pharmaceuticals). The dose of

Page 50: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

39

metformin was changed in the taspoglutide study as the previous dose of 225 mg/kg/day in the

exendin-4 study did not improve oral glucose tolerance. The dose of taspoglutide was chosen

after results from a 4-week pilot study in 6-week-old C57BL6 wildtype mice where the 0.4-mg

and 1.0-mg doses were administered and assessed for the least change in body weight while still

lowering blood glucose compared to chow-fed controls during an oral glucose tolerance test.

The reason for the metformin group in this study was also to have a control for the glucose-

lowering effect of taspoglutide. Mice were treated for 12 weeks (30-week old) with placebo or

taspoglutide microtablets administered every 4 weeks and euthanized by carbon dioxide

asphyxiation.

Administration of microtablets: placebo or taspoglutide microtablets were administered

subcutaneously. Due to the thickness of needles loaded with the microtablets, mice were

anaesthetized with isofluorane and shaved for administration of the microtablet. Antibiotic

(Polysporin) was then applied in the area to avoid infection.

2.2.2 Metabolic measurements

Assessment of food intake and stool collection. Food intake was measured three days prior to

euthanasia, and stools were collected. Mice were placed in single cages without bedding and

pre-weighed food. On the last day, food and stool weights were recorded.

Body composition. Total body fat and lean mass were measured using a mouse whole-body

magnetic resonance analyzer (Echo Medical Systems, Houston, TX) after 5 and 10 weeks of

treatment.

Oral glucose tolerance test and measurement of plasma insulin levels. An oral glucose tolerance

test was carried out after 5 weeks from start of treatments as described in section 2.1.2 except

for the differences described here. A lower glucose concentration (0.75 mg/g of body weight)

was administered orally through a gavage tube. A lower glucose concentration than that

employed in the exendin-4 study was used to avoid inducing a glucose change above 33.3 mM,

which is the upper limit value measured by the glucometers.

Systolic blood pressure. The same procedure as described in section 2.1.2 was followed.

Page 51: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

40

Lipid tolerance test. After 6-week treatment, mice were fasted overnight (19:00-9:00) before a

lipid tolerance test. Olive oil (5 l/g) was administered orally through a gavage tube and blood

(50 l) was drawn from the tail vein at 0, 60, 120 and 180 min after olive oil administration.

Triglycerides were measured using a enzymatic colorimetric assay kit (Roche Cat. No.

11877771216) based on the glycerol oxidase method.

Plasma cholesterol. Blood (50 l) was withdrawn from overnight-fasted mice (19:00-9:00).

Blood was mixed with 10% (v/v) TED, plasma separated by centrifugation at 4 °C and stored at

-80°C until assayed. Plasma was assayed for cholesterol using an enzymatic colorimetric assay

kit (Wako, Cat. No. 439-17501).

2.2.3 Histological analysis of atherosclerotic lesions

Lesion size in the aortic root was quantified as described above for the exendin-4 project. In

addition, percent lesion occupied in the descending and abdominal aorta (from the left

subclavian artery to the iliac branch) was quantified as described (Tangirala, Rubin, and Palinski

1995; Whitman 2004) with the following modifications: periadventitial adipose tissue was

excised using a dissecting microscope, stained with Nile red fluorescent stain for 1 hour and

destained in PBS for 5 min. The aorta was then cut open longitudinally to expose the intima,

pinned on waxed plates on a black background and imaged using a fluorescent microscope.

Percent lesion area analysis was conducted with ImageJ software by a thresholding method.

2.2.4 Immunohistochemical analysis of macrophage infiltration

In addition to determining lesion size by oil red O at the aortic sinus, the 15th

and 20th

aortic

sections (refer to atherosclerotic lesion size method above under exendin-4 project) were stained

with MAC-2 primary antibody and stained by diaminobenzidine (DAB). The sections were

quantified for percent occupancy by macrophages within lesions using ImageScope software

(Aperio).

The aortic arch cut from sinus to left subclavian artery, with about 1-mm sections of the three

branching arteries (the brachiocephalic trunk, left common carotid artery, and left subclavian

Page 52: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

41

artery), was cleaned, fixed in 10% formaldehyde overnight and embedded in paraffin.

Longitudinal sections were cut and sections that contained all three arteries stained with MAC-

2/DAB. ImageScope (Aperio) software was used to quantify the stained area in the arch.

2.2.5 Plasma adipokine measurements

Blood (100 l) was withdrawn from 6-hour fasted mice (7:00-13:00). Blood was mixed with

10% (v/v) TED, plasma separated by centrifugation at 4 °C and stored at -80°C until assayed.

The MilliplexTM

MAP Mouse Serum Adipokine Kit (Millipore, MADPK-71K) was used to

measure plasma levels of leptin, interleukin-6, resistin, TNF- and MCP-1.

2.3 Statistical analyses

Results are presented as mean ± standard error. Statistical analyses were done using Repeated

Measures Analysis of Variance (ANOVA) followed by Bonferroni post hoc test. A P value of

<0.05 was considered to be statistically significant. To determine whether future experiments

were worth conducting to increase sample size and achieve statistical significance, a sample size

calculation was approximated using GraphPad StatMate 2.00 software. The largest standard

deviation and a power of 80% were chosen to estimate the required sample size, N, per

treatment group. The effect size was estimated from the mean difference observed between

untreated controls and either exendin-4 or taspoglutide-treated mice.

Page 53: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

42

Chapter 3 Results

3.1 Exendin-4 project

Figure 1 shows a simplified timeline for the exendin-4 project and the specific time points at

which experiments were conducted. The actual exendin-4 project was split in two groups

staggered in time. Each staggered group consisted of all treatment groups with N of 4 mice per

treatment group such that after pooling the two staggered groups, each treatment group had a

total N of 8 mice.

3.1.1 Metabolic phenotype

Increased body weight has been shown to accelerate atherogenesis in Apoe-/-

mice fed a high-fat

diet with very low levels of cholesterol (0.003%) (King et al. 2010). In the current study, mice

were given the 60 kcal% high-fat diet with no cholesterol (0.003% by weight) for 22 weeks and

showed no significant change in a) body weight b) fat to body weight ratios as assessed by MRI

and c) epididymal white adipose tissue (WAT) weights normalized to body weight when treated

with either metformin (225 mg/kg/day supplied in food) or exendin-4 (10 nmol/kg/day, ip)

(Figure 2).

Apoe-/-

mice on the 60 kcal% diet exhibited glucose intolerance (Figure 3A). Surprisingly,

metformin treatment deteriorated oral glucose intolerance towards the end of the study, although

this difference was not statistically significant (P = 0.21). Insulin-to-glucose ratios were not

different among groups (Figure 3B). Hemoglobin A1c levels were not different among groups,

although the group treated with metformin showed lower levels (P = 0.16) (Figure 3C).

3.1.2 Atherosclerosis

No statistically significant differences in aortic sinus plaque areas were detected across the three

groups (Figure 4A and 4C). Interestingly, a non-significant (P = 0.15) increase in plaque lipid

content was observed in the aorta of mice treated with exendin-4 (Figure 4B and 4C).

Page 54: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

43

Tim

elin

e f

or

the

Exe

nd

in-4

Pro

ject

Figu

re 1

. Sim

plif

ied

tim

elin

e f

or

the

exe

nd

in-4

stu

dy

pro

ject

. Mal

e A

po

e-/-m

ice

we

re o

rder

ed f

rom

Jac

kso

n L

abo

rato

ries

an

d f

ed a

sta

nd

ard

ch

ow

die

t u

nti

l 8 w

eek

s o

ld. T

her

e w

ere

th

ree

trea

tmen

t gr

ou

ps

wit

h N

= 8

per

tre

atm

ent

gro

up

:1

)6

0%

hig

h-f

at d

iet

(HFD

) an

d P

BS

inje

ctio

ns

twic

e d

aily

2)

60

% H

FD w

ith

22

5 m

g/kg

/day

met

form

in in

th

e d

iet

and

PB

S in

ject

ion

s tw

ice

dai

ly (

this

is a

co

ntr

ol f

or

glu

cose

low

eri

ng)

3)

60

% H

FD w

ith

10

nm

ol/

kg/d

ay e

xen

din

-4 in

ject

ion

s tw

ice

dai

ly (

5 n

mo

l/kg

eac

h)

The

tim

elin

e is

no

t to

sca

le.

OG

TT, o

ral g

luco

se t

ole

ran

ce t

est

; BP

, blo

od

pre

ssu

re.

Acc

limat

ize

fo

r 1

We

ek

Salin

e, M

etfo

rmin

or

Exe

nd

in-4

Tre

atm

en

ts S

tart

ed

@ 8

We

eks

Old

All

Mic

e S

tart

ed o

n 6

0%

HFD

(N

O C

HO

LEST

ERO

L)

5-W

ee

k Tr

eat

me

nt

MR

IO

GT

T

Trea

tmen

ts F

ollo

wed

fo

r 2

2 W

eeks

Un

til 3

0-W

eek-

Old

10

-We

ek

Tre

atm

en

tM

RI

21

-We

ek

Tre

atm

en

tO

GT

T

6-W

ee

k Tr

eat

me

nt

Lip

id T

ole

ran

ce T

est

Mic

e f

rom

Jac

kso

nA

rriv

e @

6 W

ee

ks O

ld

9-W

ee

k Tr

eat

me

nt

OG

TT

Bas

al

Me

asu

res

8-W

ee

k O

ld

25

-We

ek

Old

11

-We

ek

Tre

atm

en

tB

P

Page 55: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

44

Figure 2. Body weight and body fat. Apoe-/- mice were fed a 60 kcal% high-fat diet (HFD) and treated with either 225 mg/kg/day metformin (Met) supplied in food or 10 nmol/kg/day exendin-4 (EX-4) (intraperitonealinjection given twice a day) for 22 weeks. (A) Body weights of mice measured at the end of the study. (B) Fat mass was assessed by whole-body magnetic resonance imaging (MRI) after 11 weeks of treatments and (C) by weight of epididymal white adipose tissue (WAT) normalized to body weight (BW) after conclusion of the study in male Apoe-/- mice on a 60 kcal% high-fat diet (HFD). Numbers in parentheses show sample size, N.HFD, high-fat diet; Met, metformin; EX-4, exendin-4.

HFD(8)

HFD+

Met(8)

HFD+

EX-4(8)

Body WeightsA

30

20

40

50

60

10

0

Bo

dy

We

igh

t (g

)

Epididymal WATWeight per BW

HFD(8)

HFD+

Met(8)

HFD+

EX-4(7)

C0.07

0.06

0.05

0.04

0.03

0.02

0.01

0.00

Epid

. W

AT

Wt/

BW

0

5

10

15

20

25

HFD(8)

HFD+

Met(8)

HFD+

EX-4(8)

Fat MassB

Fat

(%)

Figure 3. Glucose tolerance, insulin levels and HbA1c. Effect of treatment with metformin (225 mg/kg/day in food) or exendin-4 (10 nmol/kg/day, i.p.) on (A) oral glucose (1.5 mg/g glucose) tolerance test (OGTT), (B) insulin:glucose ratios at basal and 10 min after oral gavage of glucose, and (C) hemoglobin A1c levels compared to the HFD control group after 22 weeks. Statistical analyses were carried out by one-way ANOVA for each time point in the OGTT; AUC glucose (inset in A) and HbA1c were analyzed by one-way ANOVA; insulin:glucose ratios (B) were analyzed by two-way ANOVA. HFD, high-fat diet; Met, metformin; EX-4, exendin-4; OGTT, oral glucose tolerance test.

Oral Glucose Tolerance Test

0 20 40 60 80 100 120

HFD (8)

HFD + Met (8)

HFD + EX-4 (8)

A

AU

C (

mM

·min

)

0

5

10

15

20

25

30

35

Blo

od

Glu

cose

(m

M)

0 10 0 10 0 10

Time (min)

B

**

0

0.02

0.04

0.06

0.08

0.10

0.12

0.14

Insu

lin:G

luco

seR

atio

(m

g/m

ol) HFD

HFD + Met

HFD + EX-4

Insulin:Glucose Ratios DuringOral Glucose Tolerance Test

Hemoglobin A1cC

HFD(8)

HFD+

Met(8)

HFD+

EX-4(7)

0

1

2

6

5

4

3

Hb

A1

c (%

)

Page 56: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

45

Figure 4. Atherosclerotic plaque development after treatment with metformin or exendin-4 for 22 weeks on male Apoe-/-. (A) Plaque size was quantified in the aortic root and (B) intraplaque lipid was assessed by ORO staining for mice treated with either 225 mg/kg/day metformin or 10 nmol/kg/day exendin-4 for 22 weeks. (C) Representative cross-sections of aortic roots cut at the same level (reference point taken was the first sign of aortic valves protruding from the aortic wall while cutting from the top towards the aortic valves) and stained with ORO (magnification 5X). To achieve statistical significance for Figure 4A, an approximate sample size of 80 mice per group would be needed for a power of 80% as assessed from the effect size and variability of the data using GraphPad StatMate 2.00 software.

HFD Control HFD+EX-4HFD+Met

C

Aortic Root Plaque Size

HFD HFD+Met HFD+EX-4

A

0.00

0.45

0.40

0.35

0.30

0.25

0.20

0.15

0.10

0.05

Pla

qu

e S

ize

(m

m2 )

Aortic Root Lipid Content

HFD HFD+Met HFD+EX-4

B85

75

65

55

45

35

25

Intr

apla

qu

elip

id (

%)

Page 57: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

46

3.2 Taspoglutide project

Figure 5 shows a simplified timeline for the taspoglutide project and the specific time points at

which experiments were conducted. The actual taspoglutide project was split in three groups

staggered in time. Each staggered group consisted of all treatment groups with N of 5 mice per

treatment group such that after pooling the three staggered groups, each treatment group had a

total N of 15 mice.

3.2.1 Metabolic phenotype

Treatment with taspoglutide significantly decreased body weight by 2.42 g or 8.44% when

compared to its diabetic control (P <0.05). The decrease in body weight for taspoglutide-treated

mice may be due to reduced food intake compared to diabetic control (Figure 6B) (P <0.05).

There were no significant differences in percent fat mass between groups as assessed by whole-

body MRI after 10 weeks of treatment (Figure 6C) or by gravimetric measurement of

epididymal WAT at the end of the study (Figure 6D).

Taspoglutide-treated mice had improved blood glucose control: treatment of diabetic Apoe-/-

mice with taspoglutide significantly improved oral glucose tolerance (P <0.05) (Figure 7A),

increased basal levels of plasma insulin (P <0.05) (Figure 7B) and showed reduced hemoglobin

A1c levels (P = 0.19) (Figure 7C).

3.2.2 Lipid metabolism

Lipid metabolism plays a major role in the development and progression of atherosclerosis

(Weber and Noels 2011). To assess changes in post-prandial triglyceride concentrations in

plasma after a meal, a lipid tolerance test was carried out in overnight-fasted mice administered

olive oil. After six weeks of treatment, no differences were seen among diabetic groups for

triglyceride levels in plasma during the lipid tolerance test (Figure 8). While this result may be

surprising at first it is important to note that, in contrast to other studies, no triton WR1339 was

administered to inhibit lipase activity; the decrease in plasma triglycerides observed by Hsieh et

Page 58: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

47

al (Hsieh et al. 2010) after acute administration of exendin-4 in wildtype C57BL6 mice was

after triton administration to inhibit the action of lipase. In addition, Xiao et al (Xiao et al. 2012)

demonstrated that infusion of a liquid meal formula directly into the duodenum (through a

nasoduodenal tube) of human volunteers given exenatide intravenously did not change

triglyceride levels from those of control subjects during a pancreatic clamp. While not shown

here, there were no differences in gastric emptying among groups of mice in our study as

assessed by measuring blood acetaminophen levels administered during an oral glucose

tolerance test and measuring acetaminophen levels at 0 and 10 minutes after oral gavage.

Furthermore, no differences were observed after 11 weeks of treatment in plasma triglyceride

(Figure 9A) or cholesterol (Figure 9B) levels after a six-hour fast.

Page 59: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

48

4 W

eeks

45

% H

FD3

Wee

ks f

or

STZ

PB

O, M

et o

r TA

SPO

STA

RTE

D @

13

-Wee

k-O

ld f

or

12

Wee

ks

5-W

ee

k Tr

eat

me

nt

OG

TT

10

-We

ek

Tre

atm

en

tP

lasm

a fo

r Li

pid

sM

RI

11

-We

ek

Tre

atm

en

tP

lasm

a fo

r A

dip

oki

ne

sB

P

6-W

ee

k Tr

eat

me

nt

Lip

id T

ole

ran

ce T

est

Mic

e f

rom

Jac

kso

nA

rriv

e @

4 W

ee

ks O

ld

Tim

elin

e f

or

the

Tas

po

glu

tid

eP

roje

ct

Figu

re 5

. Sim

plif

ied

tim

elin

e f

or

the

tas

po

glu

tid

est

ud

y p

roje

ct. M

ale

Ap

oe-/

-m

ice

we

re o

rder

ed f

rom

Jac

kso

n L

abo

rato

ries

an

d a

ll m

ice

we

re f

ed a

45

kc

al%

hig

h-f

at d

iet

(HFD

) im

med

iate

ly u

po

n a

rriv

al. T

her

e w

ere

th

ree

trea

tmen

t gr

ou

ps

wit

h N

= 1

5 p

er t

reat

men

t gr

ou

p:

1)

45

% h

igh

-fat

die

t (H

FD),

dia

bet

ic (

D),

inje

cte

d w

ith

th

e p

lace

bo

(P

BO

) m

icro

tab

let

ever

y 4

we

eks

2)

45

% h

igh

-fat

die

t (H

FD),

dia

bet

ic (

D),

tre

ated

wit

h 4

00

mg/

kg/d

ay m

etfo

rmin

an

d in

ject

ed

wit

h t

he

pla

ceb

o (

PB

O)

mic

rota

ble

tev

ery

4 w

eek

s (t

his

m

etfo

rmin

gro

up

is a

co

ntr

ol f

or

glu

cose

low

eri

ng)

3)

45

% h

igh

-fat

die

t (H

FD),

dia

bet

ic (

D),

inje

cted

wit

h t

he

0.4

-mg

tasp

ogl

uti

de

(TA

SPO

) m

icro

tab

let

ever

y 4

wee

ksO

GTT

, ora

l glu

cose

to

lera

nce

te

st; B

P, b

loo

d p

ress

ure

; PB

O, p

lace

bo

; Met

, met

form

in; T

ASP

O, t

asp

ogl

uti

de

.

8-W

ee

k O

ld1

3-W

ee

k O

ld

Bas

als

Page 60: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

49

Figure 6. Body weight, food intake and fat mass in male diabetic Apoe-/- mice in the taspoglutide study project. Apoe-/- mice with streptozocin-induced diabetes were fed a 45 kcal% high-fat diet (HFD) and treated with either 400 mg/kg/day metformin (Met) supplied in water or taspoglutide microtablets(subcutaneous injection given every 4 weeks) for 12 weeks. (A) Body weights were measured at the end of the study. (B) Food intake was assessed after 12 weeks of treatment. (C) Body fat content was measured after 10 weeks of treatment by whole-body magnetic resonance imaging (MRI) and (D) epididymal white adipose tissue (WAT) weight normalized to body weight at the end of the study. *P<0.05, ***P<0.001. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Comparison of groups was made by one-way ANOVA using Graphpad Prism. Not all mice were used for food intake or fat mass assessment by MRI.

Body WeightsA

*

DHFDPBO(14)

DHFDMetPBO(15)

DHFD

TASPO(15)

35

15

10

5

0

30

25

20

Bo

dy

We

igh

t (g

)

B Food Intake

DHFDPBO(9)

DHFDMetPBO(10)

DHFD

TASPO(9)

*

0.25

0.20

0.15

0.10

0.05

0.00

Foo

d in

take

(g/

ho

ur)

D Epididymal WATWeight per BW

DHFDPBO(14)

DHFDMetPBO(15)

DHFD

TASPO(15)

0.020

0.015

0.010

0.005

0.000

Epid

. W

AT

Wt/

BW

% Fat Massper Body Weight

DHFDPBO(5)

DHFDMetPBO(5)

DHFD

TASPO(5)

C

15.0

12.5

10.0

7.5

5.0

2.5

0.0

% F

at M

ass

pe

r B

D

Page 61: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

50

Figure 7. Oral glucose tolerance test, insulin levels and HbA1c levels in the taspoglutide study. (A) Oral glucose (0.75 mg/g glucose) tolerance test after 5 weeks of treatment. The concentration of glucose was halved compared to the exendin-4 study to avoid readings above 33.3 mM, the upper limit for the glucometers used. (B) Plasma insulin to glucose ratios at basal and 10 min after oral glucose gavage after 5 weeks of treatment (C) Hemoglobin A1c levels at the conclusion of the study, after 12-week treatment. *P<0.05, **P<0.01, ***P<0.001. Plasma insulin:glucose ratios were analyzed by two-way ANOVA. All other graphs were analyzed by one-way ANOVA. To detect a statistically significant difference in Figure 7C, we would need approximately 60 mice per group for 80% statistical power. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide.

C Hemoglobin A1c

DHFDPBO(14)

DHFDMetPBO(15)

DHFD

TASPO(15)

Hb

A1

c (%

)

0.0

4.5

4.0

3.5

3.0

2.5

2.0

1.5

1.0

0.5

Plasma Insulin:Glucose RatiosDuring OGTT

B

*

D HFD PBO (14)

D HFD+Met PBO (15)

D HFD TASPO (15)

0.000

0.035

0.030

0.020

0.015

0.010

0.005

0.025

Insu

lin:

glu

cose

rat

io (

mg/

mo

l)

0 10 0 10 0 10Time (min)

Oral Glucose Tolerance TestA

D PBO (14)

D Met PBO (15)

D TASPO (15)

*

*** ***

0

30

25

20

15

10

5

*

0 20 40 60 80 100 120

Blo

od

Glu

cose

(m

M) AU

C

Time (min)

Figure 8. Plasma triglyceride levels during oral lipid tolerance test in diabetic male Apoe-/-

mice in the taspoglutide study. Oral olive oil was administered after overnight fasting (14-16 hours) in the absence of triton WR1339 (no lipase inhibition) and levels of triglycerides in plasma measured using colorimetric assay. Analysis of lipid tolerance graph was made by two-way ANOVA and AUC by one-way ANOVA. To detect a statistically significant difference, the approximate sample size per group would have to be 25 mice.

3

Oral Lipid Tolerance Test

D HFD PBO (5)

D HFD+Met PBO (5)

D HFD TASPO (5)

150

75

50

25

0

125

100

0 1 2

Time (hours)

Trig

lyce

rid

e (

mg/

dl)

AU

C

175

Page 62: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

51

Livers from the diabetic control group (P = 0.20) and the group treated with metformin (P

<0.05) were heavier compared to mice treated with taspoglutide (Figure 10A). In addition, livers

from both control groups appeared nodular, yellow and unhealthy by simple visual inspection

(Figure 10D). Surprisingly, livers from mice treated with taspoglutide had a healthy and normal

appearance with no gross enlargement (Figure 10D). Triglycerides were decreased in the livers

of diabetic animals treated with taspoglutide (Figure 10B, P <0.05). In addition treatment with

either metformin or taspoglutide significantly reduced hepatic cholesterol levels (Figure 10C).

Triglyceride and cholesterol levels in stools were not significantly different among the three

diabetic groups (Figure 11A, B) but were slightly increased in mice treated with taspoglutide

(Figure 11A, B) (P = 0.20).

Figure 9. Plasma triglyceride and cholesterol levels in diabetic male Apoe-/- mice in the taspoglutidestudy. Plasma levels of (A) triglycerides and (B) cholesterol were measured by colorimetric assay after a 6-hour fast (7:00-13:00) after 11 weeks of treatment. *P<0.05, **P<0.01, ***P<0.001. Statistical comparisons made by one-way ANOVA. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide.

B Plasma Cholesterol

DHFDPBO(8)

DHFDMetPBO(8)

DHFD

TASPO(8)

500

400

300

200

0

100

Ch

ole

ste

rol (

mg/

dl)

Plasma TriglycerideA

DHFDPBO(8)

DHFDMetPBO(8)

DHFD

TASPO(8)

55

40

35

30

25

20

15

10

5

0

50

45

Trig

lyce

rid

e (

mg/

dl)

Page 63: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

52

0.000

0.005

0.010

0.015

0.020

0.025

0.030

0.035

Liv

er

Tri

gly

ce

rid

e (

mg

/mg

)

DHFDPBO(5)

DHFDMetPBO(5)

DHFD

TASPO(5)

Intrahepatic TriglyceridesB

*

Figure 10. Liver weights and intrahepatic lipids in male diabetic Apoe-/- mice in the taspoglutidestudy. (A) Wet liver weights normalized to body weight after 12 weeks of treatment. (B) Lipids in liver tissue were extracted by methanol:chloroform (Bligh & Dyer method) and triglycerides and (C) cholesterol levels quantified using colorimetric assays. To detect a significant difference in Figure 10A between diabetic control and taspoglutide mice, 35 mice per treatment group would be needed. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA.

0.000

0.001

0.002

0.003

0.004

0.005

Liv

er

Ch

ole

ste

rol

(mg

/mg

)

DHFDPBO(5)

DHFDMetPBO(5)

DHFD

TASPO(5)

Intrahepatic CholesterolC

*** ***

0.00

0.01

0.02

0.03

0.04

0.05

0.06

0.07

0.08

0.09

Liv

er

We

igh

t p

er

BW

Liver Weight per Body WeightA

DHFDPBO(14)

DHFDMetPBO(15)

DHFD

TASPO(15)

* **

D HFD D HFD + Met D HFD TASPO

D

Page 64: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

53

3.2.3 Plasma adipokines

Plasma levels of IL-6 were significantly higher in the diabetic control group than the group

treated with taspoglutide (Figure 12A). Plasma leptin levels were not significantly different

among groups (Figure 12B). Plasma resistin levels were increased in taspoglutide-treated mice

(Figure 12C, P = 0.17) and were significantly higher than in the metformin-treated group (P

<0.05). Plasma levels of TNF- and MCP-1 were below the detectable levels by the assay

(sensitivity of TNF-, 4.39 pg/mL; MCP-1, 14.4 pg/mL).

Figure 11. Triglyceride and cholesterol concentrations in stools. (A) Triglyceride or (B) cholesterol excretion in stools were measured by colorimetric assays after methanol:chloroform extraction of lipids from stools after 12 weeks of treatment. To detect a statistically significant difference in Figure 11A and Figure 11B, approximately 18 and 20 mice per group, respectively, would be needed for 80% statistical power. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA

Triglyceride Levels in StoolsA

DHFDPBO(5)

DHFDMetPBO(4)

DHFD

TASPO(7)

P = 0.17

Sto

ol T

rigl

yce

rid

e (

mg/

mg)

10

7.5

5.0

2.5

0.0

Cholesterol Levels in StoolsB

DHFDPBO(5)

DHFDMetPBO(4)

DHFD

TASPO(6)St

oo

l Ch

ole

ste

rol (

mg/

mg)

1.31.21.11.00.90.80.70.60.50.40.30.20.10.0

Page 65: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

54

Figure 12. Plasma adipokines of male Apoe-/- mice. (A) Plasma IL-6 (B) leptin and (C) resistin levels were measured after 11 weeks of treatment after a 6-hour fast (7:00-13:00) using the Millipore MilliplexTM MAP Mouse Serum Adipokine Kit. TNF- and MCP-1 levels were also measured, but levels in all mice were below the sensitivity of the assay (sensitivity TNF- 4.39 pg/mL, MCP-1 14.4 pg/mL). *P<0.05, **P<0.01, ***P<0.001 by one-way ANOVA. To detect a statistically significant difference in Figure 12C, 35 mice per group would be needed for 80% statistical power. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA.

***

Plasma IL-6A

DHFDPBO(5)

DHFDMetPBO(4)

DHFD

TASPO(4)

Pla

sma

IL-6

(p

g/m

l)

90

80

70

60

50

40

30

20

10

0

Plasma LeptinB

DHFDPBO(8)

DHFDMetPBO(8)

DHFD

TASPO(8)

Pla

sma

Lep

tin

(pg/

ml)

1300120011001000900800700600500400300200100

0

Plasma ResistinC

DHFDPBO(8)

DHFDMetPBO(8)

DHFD

TASPO(8)

*

Pla

sma

Re

sist

in(p

g/m

l)

4500

4000

3500

3000

2500

2000

1500

1000

500

0

3.2.4 Atherosclerosis

Atherosclerotic plaque area was measured in the aortic root using ORO staining. Interestingly,

increased plaque area was detected in mice treated with taspoglutide at the aortic root (P = 0.18)

and this difference was statistically significant compared to metformin-treated controls (P <0.05,

Figure 13A and 13B). There were no differences in lipid content within the plaques of

taspoglutide-treated mice compared to the diabetic control group; however, the metformin-

Page 66: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

55

treated group exhibited significantly lower lipid levels within the plaques compared to diabetic

control mice (Figure 13C) (P <0.05).

Macrophage content of plaques at the aortic sinus was quantified using Mac-2 antibody. The

percent macrophage area within the plaques of the aortic sinus was not different among all

diabetic groups regardless of treatment (Figure 13D). Similarly, macrophage infiltration

assessed at the aortic arch was also not different among the study groups (Figure 14).

Atherosclerosis area in the descending aorta was quantified using Nile red staining. While

plaque area was slightly reduced in taspoglutide-treated mice, the decrease did not reach

statistical significance (P = 0.19) (Figure 15).

It was interesting to note that plaques in the aortic sinus were complex and in later stages

whereas plaques in the aortic arch and abdominal aorta were of very early stages, predominantly

fatty streaks. This may be because a diet very low in cholesterol was used and the aortic sinus is

the site that is most prone for lesion formation; other sites require even greater increases in

plasma cholesterol to accelerate lesion formation (Daugherty et al. 2009; Whitman 2004).

3.2.5 Blood Pressure

Systolic blood pressure was not different among groups when measured using a tail-cuff system

for the taspoglutide-treated groups or the exendin-4 treated groups (Figure 16).

Page 67: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

56

Figure 13. Characterization of plaques in the aortic root in male diabetic Apoe-/-mice in the taspoglutidestudy. (A) Plaque size was quantified in the aortic root. (B) Representative oil-red O histology images of aortic sinus cross-sections cut at the same level (reference point taken was the first sign of aortic valves protruding from the aortic wall while cutting from the top towards the aortic valves). Magnification 5X. (C)Intraplaque lipid was assessed with oil-red O staining for lipids. (D) Mac-2 antibody was used for staining for macrophages in plaques at the aortic sinus. *P<0.05, **P<0.01. . To detect a statistically significant difference in Figure 13A, approximately 55 mice per treatment group would be needed for a statistical power of 80%. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA.

D HFD PBO D HFD+Met PBO D HFD TASPO

B

Aortic Sinus Plaque Area

DHFDPBO(13)

DHFDMetPBO(14)

DHFD

TASPO(14)

A

*

Pla

qu

e A

rea

(mm

2 )

0.35

0.30

0.25

0.20

0.15

0.10

0.05

0.00

DHFDPBO(13)

DHFDMetPBO(15)

DHFD

TASPO(14)

Intraplaque LipidAt the Aortic Sinus

C

*

Intr

a-p

laq

ue

lip

id (

%)

60

50

40

30

20

10

0

D Mac-2 Staining in PlaquesAt the Aortic Sinus

DHFDPBO(5)

DHFDMetPBO(5)

DHFD

TASPO(4)

Mac

-2-s

tain

ed

are

a (%

)

35

30

25

20

15

10

5

0

Page 68: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

57

B

D HFD PBO D HFD+Met PBO D HFD TASPO

Figure 14. Macrophage area stained at the aortic arch. (A) Macrophages were quantified in the aortic arch using mac-2 antibody and positive pixel analysis to determine stained area. (B) Representative longitudinal sections of the aortic arch stained with mac-2 antibody. To detect a statistically significant difference, an approximate sample size of 100 mice per group would be needed for a power of 80%. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA.

DHFDPBO(13)

DHFDMetPBO(15)

DHFD

TASPO(13)

Aortic Arch Macrophage AreaA

Mac

-2 a

rea

(mm

2 )

0.130.120.110.100.090.080.070.060.050.040.030.020.010.00

Page 69: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

58

D HFD PBO D HFD+Met PBO D HFD TASPO

B

Figure 15. Plaque area occupied in the descending aorta of diabetic male Apoe-/- mice in the taspoglutidestudy. (A) The area of early atherosclerotic plaques in the descending aorta was determined by the en facemethod of aortas stained with Nile red. The area was quantified by thresholding and expressed as the percent total area occupied within total intimal area. To detect a statistically significant difference, 60 mice per treatment group would be needed for a power of 80%. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA.

% Atherosclerotic Surface AreaDescending Aorta

A

DHFDPBO(13)

DHFDMetPBO(12)

DHFD

TASPO(13)

Pla

qu

e a

rea

(%)

5

4

3

1

0

2

Page 70: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

59

0

25

50

75

100

125

Syst

olic

BP

(m

mH

g)

Systolic Blood PressureTaspoglutide Study

DHFDPBO(14)

DHFDMetPBO(15)

DHFD

TASPO(15)

Figure 16. Systolic blood pressure of male Apoe-/- mice in both studies. Systolic blood pressure was measured by tail-cuff measurements after mice were trained. (A) Systolic blood pressure of glucose-intolerant male Apoe-/- mice after 11 weeks of treatment in the exendin-4 study. (B) Systolic blood pressure of male diabetic Apoe-/- mice after 10 weeks of treatment in the taspoglutide study. D, diabetic; HFD, high-fat diet; PBO, placebo; Met, metformin; TASPO, taspoglutide. Statistical comparisons were made by one-way ANOVA.

0

25

50

75

100

150

Syst

olic

BP

(m

mH

g)

125

Systolic Blood PressureExendin-4 Study

HFD(8)

HFD+

Met(8)

HFD+

EX-4(8)

Page 71: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

60

Chapter 4 Discussion

In order to identify potential roles of GLP-1R activation on atherogenesis, it was important to

elucidate the effect of GLP-1R activation on adipocyte mass, insulin resistance,

hyperinsulinemia, plasma glucose levels, triglyceride and cholesterol levels in the Apoe

knockout mouse model as these impact the development of atherosclerosis (Weber and Noels

2011; Molecular Mechanisms of Atherosclerosis 2005; Rask-Madsen et al. 2012; Rask-Madsen

et al. 2010). Because previous studies implicate the GLP-1R in having a role in the reduction of

body weight, increase in insulin secretion, regulation of blood glucose homeostasis, and

reduction of triglyceride and cholesterol levels (section 1.4), it was hypothesized that GLP-1R

activation would lead to decreased atherosclerotic lesion area. Our results, however, revealed an

overall neutral effect of either exendin-4 or taspoglutide treatment on atherogenesis when lesion

size and macrophage infiltration was assessed at the aortic sinus, aortic arch and the abdominal

aorta.

4.1 Effect of GLP-1R activation on body weight, adipocyte mass, adipokines and atherogenesis

Reductions in body weight in T2DM patients treated with exenatide or liraglutide is well-

known; a recent analysis of 39 randomized controlled trials that assessed different treatments for

T2DM found GLP-1 analogues significantly decreased body weight with a mean change of -

1.66 kg (Liu et al. 2012). GLP-1 agonists are known to induce satiety through their actions in

the brain; intracerebroventricular administration of GLP-1 dose-dependently inhibits food intake

in fasted rats (Alhadeff, Rupprecht, and Hayes 2012; Gutzwiller et al. 1999; Turton et al. 1996).

We showed that long-term taspoglutide treatment for 12 weeks in Apoe-/-

mice fed a high-fat

diet significantly reduced body weight. However, long-term exendin-4 treatment for 22 weeks

did not significantly alter body weight; a dose of exendin-4 was chosen (10 nmol/kg/day) that

would not induce significant changes in body weight and this dose was assessed in a separate

pilot study of 6-week-old C57BL6 wildtype mice given exendin-4 for 4 weeks at various doses

to optimize the effect of blood glucose lowering while maintaining a neutral effect on body

weight. The reduction in body weight in taspoglutide-treated mice did not correlate with a

Page 72: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

61

reduction in fat mass normalized to body weight as assessed by either whole-body magnetic

resonance imaging (MRI) or epididymal white adipose tissue weights normalized to body

weight. Taspoglutide-treated mice’s caloric intake was significantly reduced compared to

control and may explain the reduction in body weight observed. Thus, while caloric intake was

reduced with GLP-1R activation, body fat ratio did not change. This result was probably due to

a balancing combination of increased insulin levels in taspoglutide-treated mice compared to

control (which increased the rate of adipogenesis (Guller et al. 1988)), but decreased food intake

in taspoglutide-treated mice, which limited the extent of overall body weight gain and

adipogenesis.

As introduced in section 1.3.4, obesity is associated with a predisposition to developing

atherosclerosis and this is thought to be due to an establishment of a pro-inflammatory milieu

involving macrophage infiltration into adipocytes and the secretion of adipocytokines and

cytokines (Tilg and Moschen 2006; Wellen and Hotamisligil 2005).

In the taspoglutide study, diabetic control animals had significantly elevated plasma IL-6 levels

compared to animals treated with either metformin or taspoglutide. Chronic elevations of

plasma IL-6 are thought to be pro-inflammatory due to induction of hepatic insulin resistance

and its association with systemic CRP (Maury and Brichard 2010), which has been shown to

enhance atherosclerosis in Apoe-/-

mice overexpressing human CRP (Paffen and DeMaat 2006).

Thus, the elevations in IL-6 seen in diabetic control animals may induce increased

atherogenesis. On the other hand, IL-6 has also been shown to have anti-inflammatory effects

(see section 1.3.4.3), and thus, the exact significance of elevated IL-6 levels and its net effect on

atherogenesis is unclear.

The precise role of leptin in atherosclerosis is not known. However, it is postulated that

hyperleptinemia is atherogenic (see section 1.3.4.1) (Ritchie et al. 2004). Plasma leptin levels

were unchanged by either metformin or taspoglutide treatments. Given that leptin is secreted by

adipocytes in proportion to their mass (Maury and Brichard 2010), this result is not surprising.

Interestingly, resistin levels in taspoglutide-treated mice were slightly elevated compared to

control, but this difference was not statistically significant (P = 0.17). Resistin is thought to be

pro-atherogenic (see section 1.3.4.2), but the small increase seen in taspoglutide-treated mice is

unlikely to have a significant role (Tedgui and Mallat 2006; Tilg and Moschen 2006).

Page 73: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

62

Thus, the net effect of taspoglutide on the reduction of body weight and IL-6 levels compared to

diabetic controls predicts a reduction in atherogenesis. However, the reduction in body weight

was only an 8.44% change from controls and did not result in a significant change in fat mass to

body weight ratio. Furthermore, the pro-inflammatory effects of chronically elevated IL-6 is a

matter of controversy (see section 1.3.4.3). Therefore, the reduction in body weight and IL-6

levels may have contributed to the neutral effect we observe on atherosclerosis development

compared to control mice.

4.2 Effect of GLP-1R activation on glucose homeostasis and atherogenesis

Activating the GLP-1R aids in the maintenance of glucose homeostasis, which is mediated not

only by stimulation of increased insulin release and inhibition of glucagon release, but also

through indirect mechanisms that stimulate glucose uptake by peripheral tissues (reviewed in

(Baggio and Drucker 2007)). Insulin resistance is often accompanied by hyperinsulinemia and

T2DM is characterized by hyperglycemia (Molecular Mechanisms of Atherosclerosis 2005).

The relative contributions of insulin resistance, hyperinsulinemia and hyperglycemia to

atherosclerosis development were discussed in section 1.4.1.4.

In the exendin-4 study, 10 nmol/kg/day of extendin-4 administered as two intraperitoneal

injections per day did not improve oral glucose tolerance beyond that of control. Interestingly,

metformin showed a trend (P = 0.21) for worsening glucose tolerance. Insulin-to-glucose ratios

and HbA1c were similarly unaffected in the exendin-4 study. A likely reason for this is that the

dose of exendin-4 administered was not sufficient to improve glucose homeostasis. The chosen

dose of exendin-4 was arrived at with 6-week-old mice in the absence of glucose intolerance

induced by a high-fat diet as in the exendin-4 study; it is likely that in the setting of diet-induced

glucose intolerance, the dose needed had to be higher. However, we opted to keep the dose

constant in order to avoid the confounding effects of weight loss in atherosclerosis. Mice in the

exendin-4 study were glucose intolerant and hyperinsulinemic, which are two important

characteristics of insulin resistance. Because the exendin-4 dose used did not improve glucose

homeostasis, this allowed us to eliminate the contribution of reduction in hyperglycemia that

would impact atherogenesis. However, independent of changes in glucose, the dose of exendin-

Page 74: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

63

4 used (10 nmol/kg/day) did not change atherosclerosis lesion size or intraplaque lipid content in

the aortic sinus. The reason for this observation may also have been due to higher dose

requirements as these mice age; Arakawa et al. (Arakawa et al. 2010) used an infusion rate of 24

nmol/kg/day of exendin-4 for their high-dose group in order to observe a significant difference

in plaque size at the aortic sinus of relatively younger (12-week-old) Apoe-/-

mice. Another

difference in our experimental setup from that of Arakawa et al. is that we intermittently

injected the mice with two i.p. injections per day, whereas Arakawa et al. continuously infused

exendin-4 through an osmotic pump, thus activating the GLP-1R continuously rather than

intermittently. Assuming no desensitization of the GLP-1R, it is possible that continuously

activating the receptor may be more effective in attenuating plaque development. For this

reason, we opted to use taspoglutide, a GLP-1R agonist formulated as microtablets with a much

longer half-life that is administered every 4 weeks in mice.

Taspoglutide improved oral glucose tolerance, increased insulin secretion and improved HbA1c

(P = 0.19). Lower insulin levels in control diabetic mice were associated with more

hyperglycemia. Despite improved glucose homeostasis, taspoglutide did not change

atherosclerosis lesion size in the aortic sinus or the descending aorta, or macrophage infiltration

in plaques at the aortic sinus or in the aortic arch. Improvements in glucose levels in

taspoglutide-treated mice was expected to decrease macrophage infiltration in early lesions, as

shown in the study by Renard et al, where they employed Ldlr-/-

mice expressing a viral

glycoprotein (GP) under the control of the rat insulin promoter such that, when infected with the

lymphocytic choriomeningitis virus, they develop type 1 diabetes due to selective destruction of

cells by T cells. These Ldlr-/-

:GP mice had greater macrophage infiltration in early lesions of

the brachiocephalic artery (BCA) compared to non-diabetic controls independent of changes in

the lipid profile (mice were induced with diabetes at 7-10 weeks of age and given a cholesterol-

free, normal chow diet for 12 weeks) (Renard et al. 2004). While there was a slight decrease in

infiltration of macrophages in the early lesions of the aortic arch of metformin and taspoglutide-

treated mice compared to control mice, the effect was not statistically significant. It is possible

that the lack of effect in macrophage infiltration may be due to changes in the dynamic of

infiltrating cells as the atherosclerotic lesions examined in the aortic arch may have progressed

past the initiating stages of atherogenesis, and it is possible that activation of the GLP-1R past

the early fatty streaks may have a different role as more advanced lesions of the aortic sinus

reveal a relative increase in lesion size compared to diabetic and metformin-treated controls.

Page 75: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

64

Furthermore, while insulin sensitivity was not assessed, the decrease in glycemia from

taspoglutide treatment would be expected to improve insulin sensitivity according to the glucose

toxicity theory, where peripheral cells adapt to chronic increases in glucose by increasing insulin

resistance to decrease the rate of glucose entry (Yki-Jarvinen 1990, 1997). The improvement in

insulin sensitivity in taspoglutide-treated mice, in turn, was expected to halt the accelerated

progression of atherosclerosis due to diabetes, as studies in EIRAKO mice showed increased

lesion area with decreased insulin signaling (discussed in section 1.3.2.2) (Rask-Madsen et al.

2010). Rather, only a small (not statistically significant) decrease was recorded in the early

lesions of the descending aorta of taspoglutide-treated mice. This result may be due to a

relatively small effect in the increase in insulin secretion in the taspoglutide group compared to

controls (while basal insulin:glucose ratios were statistically significant, post-prandial levels did

not reach statistical significance).

4.3 Effect of GLP-1R activation on lipoprotein metabolism

One major characteristic of diabetes (type 2) is increased hepatic VLDL secretion that results in

increased plasma VLDL triglyceride and APOB secretion (Adiels et al. 2005). Mechanisms

proposed include increased free fatty acid flux into the liver due to peripheral insulin resistance

that increase availability of lipid for VLDL synthesis and secretion (reviewed in (Lewis 1997)).

Furthermore, insulin has been shown to suppress VLDL APOB production in healthy human

volunteers while insulin-resistant subjects are resistant to the suppressive effect of insulin on

VLDL APOB production (Lewis and Steiner 1996). Thus, it may seem that decreased insulin

signaling in the periphery and liver could both act to increase VLDL secretion.

Activation of the GLP-1R has been shown to decrease postprandial lipid levels (section 1.4.1.1).

In the taspoglutide study, however, treatment with taspoglutide for six weeks did not change

postprandial lipid levels during an oral olive oil gavage on overnight-fasted mice. Experiments

to date have shown an inhibitory effect of GLP-1R activation on intestinal APOB lipoprotein

production (Hsieh et al. 2010; Qin et al. 2005). Thus, while taspoglutide treatment might have

decreased the rate of intestinal lipoprotein production, it is not known whether hepatic and/or

peripheral tissues had a decreased rate of uptake of lipoproteins. The decreased levels of

Page 76: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

65

triglycerides and cholesterol in the liver suggest this may in fact be the case. Alternatively,

taspoglutide may have reduced hepatic lipid synthesis or increased lipid oxidation.

Taspoglutide treatment also did not change triglyceride or total cholesterol levels after 11 weeks

of treatment after a six-hour fast. This result is in agreement with similar reports where Apoe-/-

mice were treated with exendin-4 (Arakawa et al. 2010), GLP-1(7-36)NH2 (Nagashima et al.

2011), or sitagliptin (Vittone et al. 2012).

Interestingly, while plasma triglyceride and cholesterol levels remained unchanged, intrahepatic

levels differed greatly in the taspoglutide group compared to diabetic controls; intrahepatic

triglycerides showed a mean decrease of about 40%, and intrahepatic cholesterol decreased

about 39%. In addition, liver weight normalized to body weight tended to be lower in

taspoglutide-treated mice (about 12% smaller). Measurement of triglycerides and cholesterol in

stools revealed that some of the lipids were being excreted; stools of taspoglutide-treated mice

had a mean triglyceride concentration about 67% higher, and a mean cholesterol concentration

about 45% higher, although these differences did not reach statistical significance (P = 0.17 for

triglyceride and P = 0.20 for cholesterol). Thus, intrahepatic lipid concentrations may be partly

explained by enhanced excretion in the stools.

To compare our current observations with those in the literature, one report (Burmeister et al.

2012) showed that in 18-week-old high-fat-fed wildtype mice (60 kcal/g for 12 weeks), an acute

intracerebroventricular infusion of GLP-1(7-36)NH2 (0.01 g/min) during a hyperinsulinemic

euglycemic clamp led to a marked decrease in intrahepatic triglycerides. In addition, chronic

activation of the GLP-1R also reduces hepatic fat. One study showed 4-5-week old wildtype

C57BL6 mice fed a 45 kcal% high-fat diet rich in hydrogenated vegetable oil for 8 weeks then

treated with liraglutide (200 g/kg/day, i.p.) for 4 weeks exhibited reduced liver weight, serum

triglycerides and cholesterol, and decreased hepatic steatosis (as examined by ORO staining).

These mice showed reduced levels of mRNA transcripts for genes associated with lipid

synthesis (fatty acid synthase, acetyl CoA carboxylase alpha and PPAR-) and increased levels

of mRNA transcripts of peroxisomal fatty acid -oxidation (acyl-CoA oxidase-2 (Acox2))

(Mells et al. 2012).

The increase in triglyceride concentration in stools in taspoglutide-treated mice could be due to

decreased absorption of triglycerides; infusion of GLP-1(7-36)NH2 (1 pmol/kg/min

Page 77: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

66

intravenously) inhibited human gastric lipase secretion in healthy human volunteers, which may

prevent the hydrolysis of triglycerides for absorption by enterocytes (Wojdemann et al. 1998).

Intrahepatic cholesterol may also have been reduced from higher excretion rates in bile and/or

decreased reabsorption from the gut as concentrations observed in stool tended to be higher than

in diabetic control animals.

The increase in liver weights and unhealthy appearance of livers in diabetic control mice was a

notable finding. It is important to note that compared to wildtype C57BL6 mice, Apoe-/-

mice

accumulate triglycerides in liver to a greater extent; one report showed male C57BL6 wildtype

mice fed a regular chow diet (6.2% fat and 0.01% cholesterol) until 3-4 months of age

accumulated 20.6 nmol/mg of triglycerides in liver, while Apoe-/-

mice accumulated 64.1

nmol/mg (P < 0.05) (Mensenkamp et al. 1999). Thus, the combination of increased intrahepatic

triglycerides and increased glucose flux due to hyperglycemia in diabetic mice in the

taspoglutide study may have led to the increase in liver weight, triglycerides and cholesterol,

which are important features for the development of NAFLD. Since taspoglutide-treated mice

had decreased intrahepatic triglycerides and cholesterol, there may have been a smaller chance

for development of NAFLD. Thus, the decrease in intrahepatic lipids and improvement in

glucose homeostasis from taspoglutide treatment may be the reason for the observed differences

in the livers’ physical texture, colour appearance and size between groups.

4.4 Effect of GLP-1R activation on atheroma development

As discussed in section 1.4.2.4, previous studies (Arakawa et al. 2010; Nagashima et al. 2011)

demonstrated that activation of the GLP-1R resulted in decreased lesion size in non-diabetic

mice. The potential mechanisms leading to the smaller lesions in these reports was a decrease in

the mRNA expression of adhesion molecules expressed on the endothelium such as ICAM-1

and VCAM-1; reduced CD11b receptor on monocytes for binding to ICAM-1; reduction in

LPS-induced MCP-1 and TNF-; and reduced cholesteryl ester accumulation due to reduced

CD36 and ACAT-1 in macrophages (Arakawa et al. 2010; Nagashima et al. 2011).

However, in the presence of diet-induced glucose intolerance as in our exendin-4 study,

exendin-4 treatment did not influence the size of lesions in the aortic root compared to control.

In addition, in the presence of streptozotocin-induced diabetes as in our taspoglutide study, there

Page 78: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

67

was no significant change in the size of the lesions in the aortic root or the percent area occupied

by lesions in the abdominal aorta. Plaques in the aortic sinus were complex and in later stages

whereas plaques in the aortic arch and abdominal aorta were of very early stages, predominantly

fatty streaks.

Examination of macrophage content within advanced plaques of the aortic sinus or infiltrated

macrophage area within the intima and/or fatty streaks of the aortic arch also did not reveal

significant differences among groups.

Our studies thus reveal that in the setting of diet-induced glucose intolerance or streptozotocin-

induced diabetes, chronic activation of the GLP-1R does not have an effect on atherogenesis. In

fact, there was an increase in plaque area in the aortic sinus and a slight decrease in the

abdominal aorta in taspoglutide-treated mice. Normally, an increase in plaque size in the aortic

sinus corresponds to an increase in plaque size in the abdominal aorta (Tangirala, Rubin, and

Palinski 1995). However, in our taspoglutide study, there is a slight reduction in lesion size

observed for the early stages of atherosclerosis in the abdominal aorta (P = 0.19) and a slight

enlargement in size in more advanced lesions in the aortic sinus (P = 0.18). Furthermore, a slight

trend for a decrease in macrophage area in early lesions of the aortic arch was observed while no

change in the percentage occupied by macrophages is observed at the aortic sinus. The small

decrease in macrophage area in early lesions of the aortic arch in mice treated with either

metformin or taspoglutide may be due to improved glycemia, as discussed above in section 4.2.

Previous studies (Arakawa et al. 2010; Nagashima et al. 2011) demonstrated that activation of

the GLP-1R resulted in decreased lesion size in the aortic root and abdominal aorta in non-

diabetic mice. These studies activated the GLP-1R with either exendin-4 for 4 weeks in young

(8-week-old) mice (Arakawa et al. 2010) or with GLP-1(7-36)NH2 in slightly older (17-week-

old) mice (Nagashima et al. 2011). In the report by Nagashima et al. (Nagashima et al. 2011),

mice were given a high-fat, high-cholesterol diet for 4 weeks starting at 17 weeks of age. Mice

infused with GLP-1(7-36)NH2 for 4 weeks concurrently with the high-fat, high-cholesterol diet

(until 21 weeks old) had no increase in plaque area compared to mice 17 weeks of age (before

the introduction of the high-fat, high-cholesterol diet) when quantified en face from the aortic

arch to the iliac bifurcation; in fact, lesion area was slightly lower in GLP-1-treated mice

compared to 17-week-old controls. In contrast, lesions at the aortic root still increased with 4-

Page 79: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

68

week GLP-1 infusion compared to 17-week-old controls, although not as much as untreated 21-

week-old controls given the high-fat, high-cholesterol diet. They also saw only small (but

statistically significant) reductions in macrophage infiltration after GLP-1 treatment in the aortic

root. Similarly, Arakawa et al. (Arakawa et al. 2010) observed marked reductions in monocyte

adhesion at branching points of the thoracic aorta at the endothelium surface in the very early

stages of atherosclerosis after 4 weeks of exendin-4 treatment, while only small reductions in

plaque area could be observed in the aortic sinus after 4 weeks of exendin-4 treatment. The

decreasing trends in macrophage infiltration in the aortic arch, as well as early stage lesion area

in the abdominal aorta in our studies agree with the reports by Arakawa et al. and Nagashima et

al. However, in our studies, activating the GLP-1R for 12 weeks, rather than just 4 weeks,

resulted in increased lesion size in the aortic sinus, while the other two papers (Arakawa et al.

2010; Nagashima et al. 2011) reported a statistically significant reduction in lesion size after the

shorter 4-week treatment. Thus, it is likely that activating the GLP-1R in diabetic mice, rather

than non-diabetic mice, has different consequences for early lesions, such as in the abdominal

aorta, and different consequences in more advanced lesions, such as in the aortic sinus. Indeed,

as discussed above, the study by Renard et al found diabetic mice had accelerated formation of

early lesions by inducing greater macrophage infiltration in the BCA independent of changes in

lipoprotein profile (Renard et al. 2004).

The development of atherosclerosis involves many cell types and their involvement is very

specific according to the stage and complexity of the lesion (reviewed in (Hansson and

Hermansson 2011; Weber and Noels 2011)). One possible explanation for the reduction of

atherosclerosis development in the early stages would be the activation of the GLP-1R in

regulatory T cells (Tregs). Our lab has shown exendin-4 treatment (2 g twice daily for 26

weeks) in nonobese diabetic (NOD) mice, a model of type 1 diabetes that spontaneously

develop diabetes by autoimmune destruction of pancreatic beta cells, delays the onset of type 1

diabetes (Hadjiyanni et al. 2008). Furthermore, purified CD4+CD25+ ex vivo lymphocytes (at

least 75% of which are Tregs) from male C57BL6 mice express the Glp1r receptor mRNA

(Hadjiyanni et al. 2008; Hadjiyanni et al. 2010). It has also been shown that Glp1r-/-

lymph-

node-derived lymphocytes are hyperproliferative in response to mitogenic stimulation such as

concanavalin A or phorbol myristate acetate plus ionomycin. And, a significant reduction of

CD4+CD25+FoxP3+ Treg cells is observed in lymph nodes from Glp1r-/-

mice (Hadjiyanni et

al. 2010). Thus, GLP-1R signaling may regulate the proliferation and maintenance of regulatory

Page 80: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

69

T cells. Experiments in Apoe-/-

mice have also revealed an atheroprotective role of Tregs. The

first demonstration consisted of transfer experiments where in vitro-expanded Tregs from

BALB/c mice were injected intraperitoneally (106 cells per mouse) to female Apoe

-/- mice; mice

that received the Treg clones had markedly reduced atherosclerosis and macrophage infiltration

independent of plasma cholesterol (Mallat et al. 2003). Other experiments have also shown an

atheroprotective role for Tregs. For example, an experiment that disrupted the CXCR3

chemokine receptor in Apoe-/-

mice (CXCR3-/-

Apoe-/-

) (CXCR3 is a chemokine receptor of the

CXC family expressed in T lymphocytes) increased the number of Tregs and delayed early

atherosclerosis (lesion size at the aortic sinus was unaffected) by attenuating recruitment of T

helper 1 (TH1) cells (Veillard et al. 2005). One limitation of our studies is that we did not

examine T-cell populations within plaques, however it is possible that treatment with exendin-4

and/or taspoglutide may have attenuated early atherosclerosis lesion formation by activation of

the GLP-1R on Tregs. This may explain our trend for decreased atherosclerosis in the

abdominal aorta of taspoglutide-treated mice, but as the lesion progressed into later stages in the

aortic sinus, the atheroprotective role of Tregs was no longer apparent (their role in later stages

is currently not clear).

For later stages of atherosclerosis, such as in the aortic sinus, it is possible that GLP-1R

activation by exendin-4 and/or taspoglutide may have led, directly and/or indirectly, to the

secretion of extracellular matrix material by macrophages and/or smooth muscle cells that led to

the slight increase in size. Activation of the GLP-1R may have also stimulated the proliferation

and/or migration of smooth muscle cells to stabilize the plaque, which may be an important

finding despite the observed trend for an increase in lesion size because stabilization of plaques

by smooth muscle cells may prevent thrombosis and/or plaque rupture, which is clinically

important to reduce the incidence of cardiovascular events. One report clearly illustrates this

point: Apoe-/-

mice fed a high-fat (40 kcal%), high-cholesterol (1.25%) diet and treated with

sitagliptin (576 mg/kg) for 12 weeks did not change lesion size in the aortic root or the rest of

the aorta, but significant reductions in macrophage infiltration, metalloproteinase-9 and a two-

fold increase in collagen were observed, which indicate that, while plaque size was unaffected,

the plaques themselves have a more stable composition (Vittone et al. 2012). While we did not

observe a reduction in macrophage infiltration, it is possible that the increase in plaque size in

the aortic sinus may be from an increase in secreted collagen to stabilize the lesions.

Page 81: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

70

4.5 Expression of the GLP-1R in liver and atheroma

A possible explanation to the conflicting findings in atherogenesis and macrophage area from

previous reports (Arakawa et al. 2010; Nagashima et al. 2011) is that the GLP-1R may not be

expressed in monocytes and/or macrophages, as reported in non-diabetic Apoe-/-

mice.

The expression of the GLP-1R in different tissues has been difficult to reconcile due to

difficulties in generating antibodies specific for the receptor. Antibodies for GPCRs are usually

raised against a fragment(s) of synthetic peptide antigen that corresponds to the membrane

protein. The epitope recognized by the antibody may not be specific and may in fact, yield

several bands during Western blot analysis. The concern of antibody specificity for GPCRs is

not new, and concerns for faulty specificity for membrane receptors including adrenergic,

muscarinic and dopaminergic receptors, have been expressed before (Kirkpatrick 2009). One

method to identify and/or verify expression of the receptor is by examining mRNA expression

using reverse-transcribed cDNA followed by PCR using primers for the full length Glp1r.

Identification of the full length expression is important as primers directed at amplifying partial

fragments may yield faulty results (there may be leaky transcription in some tissues, but the

partial mRNA may never be translated).

Thus, it is possible that the anti-GLP-1R antibody from Abcam used by both studies (Arakawa

et al. 2010; Nagashima et al. 2011) may not be specific for the receptor and the observed

decrease in monocyte adhesion and/or plaque may be rather an indirect result of GLP-1R

activation in tissues other than monocytes and/or macrophages. Furthermore, the reduction in

cholesteryl esters recorded by Nagashima et al. (Nagashima et al. 2011) in peritoneal

macrophages from non-treated Apoe-/-

mice may have been an effect of the truncated peptide,

GLP-1(9-36)NH2 through a GLP-1R-independent mechanism possibly by acting through

another receptor that has yet to be identified (Ban et al. 2008).

Page 82: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

71

Chapter 5 Conclusions

The present study demonstrated that exendin-4 treatment twice daily (10 nmol/kg/day, i.p.) in

Apoe-/-

mice fed a 60 kcal% high-fat diet for 22 weeks did not reduce body weight, adiposity,

glucose tolerance nor insulin to glucose ratios, and did not reduce plaque size in the aortic sinus.

In the taspoglutide study, taspoglutide:

- Significantly reduced body weight (P <0.05)

- Significantly reduced food intake (P <0.05)

- Had no effect on adiposity

- Improved glucose tolerance beyond that of diabetic control animals (P <0.05)

- Significantly increased basal insulin to glucose ratio (P <0.05)

- Tended to improve hemoglobin A1c (P = 0.19)

- Did not change plasma triglyceride or cholesterol levels compared to diabetic controls

- Tended to decrease liver weight per body weight (P = 0.17)

- Significantly reduced intrahepatic TG (P <0.05) and cholesterol (P < 0.001)

- Tended to increase triglyceride (P = 0.17) and cholesterol levels in stools (P = 0.20)

- Significantly reduced plasma IL-6 levels

Unexpectedly, taspoglutide did not change overall plaque size in STZ-induced diabetic mice

compared to diabetic controls in the aortic sinus (P = 0.18) or the descending aorta (P = 0.19).

Furthermore, there was no change in macrophage-infiltrated area within plaques of the aortic

sinus or early lesions in the aortic arch.

Page 83: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

72

Chapter 6 Future Directions

6.1 Atherosclerosis

One of the important questions that arise from our current studies is whether GLP-1R activation

changes in any way the composition and/or secretion of metalloproteinases and TIMPs (tissue

inhibitor of metalloproteinases) within the plaque. This is an important question from a clinical

standpoint because (assuming results in mice translate to humans) if plaque stability is

enhanced, this may translate into reduced cardiovascular events from treatment with GLP-1R

agonists. Conversely, if plaques become more unstable from GLP-1R activation, this may lead

to increased plaque rupture and/or thrombosis which translates into increased cardiovascular

events. Thus, in order to assess plaque stability in these mice, stains for collagen, SMCs.

metalloproteinases and TIMPs may aid in answering these questions. Collagen stains would

help determine the thickness of fibrous caps (the thicker the cap, the more stable the plaque) and

an increase in the number of smooth muscle cells may also give an indication of increased

stability. Increased metalloproteinases destabilize the plaque, while increased TIMPs inhibit

metalloproteinases and stabilize the plaque.

Our current studies, as well as studies by others (Arakawa et al. 2010; Matsubara et al. 2012;

Nagashima et al. 2011; Shah et al. 2011; Ta et al. 2011; Terasaki et al. 2012; Vittone et al.

2012), also suggest a small effect in the initiating events of atherosclerosis from GLP-1R

activation. While different pathways in macrophages have been suggested, T cells are also

involved in the initiating events of atherosclerosis (Weber and Noels 2011). Since T cells

express the GLP-1R (Hadjiyanni et al. 2010), it may be important to understand the specific

pathways engaged by activation of the GLP-1R in T cells, especially in the post-prandial state

(when GLP-1 levels rise). First, one must assess whether GLP-1R activation leads to an altered

number of T cells and/or lymphocyte infiltration into the intima. These experiments would be

conducted en face using immunofluorescence methods to stain monocytes, T cells, dendritic

cells and macrophages to assess differences in the ratios of different cells infiltrating the intima.

These experiments would show how GLP-1R activation (either acutely or chronically) in mice

(non-diabetic or diabetic) influences the infiltration of different immune cells, including T cells.

Page 84: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

73

6.2 Fatty liver and lipoprotein metabolism

The surprising finding of livers of healthy appearance in mice treated with taspoglutide

compared to the enlarged, granulated and yellow livers of control mice may be worth exploring

further. The results in our study reveal a possible role of the GLP-1R in lipid metabolism as

triglyceride and cholesterol levels in the liver were markedly reduced, while there was a trend

for increased levels in stools (Figures 10 and 11). It may be worth exploring key markers of ER

stress (protein and/or RNA) and inflammation during the course of treatment of mice and

identify the cause of the stress (is it due to the accumulation of triglycerides and/or cholesterol

in the liver?). Thus, an experiment with Apoe-/-

mice on a low-fat diet and mice on a high-fat

diet, both with streptozotocin-induced diabetes, should be first compared at different time points

for liver morphology, intrahepatic triglyceride and cholesterol levels, and ER stress markers

(there is no such data currently in the literature). Then, the effects of activating the GLP-1R in

the liver in low-fat diabetic and high-fat diabetic mice should be contrasted to identify which

signaling pathways may be involved in the liver.

While the acute and chronic effects of GLP-1R activation in fasting and post-prandial

lipoprotein metabolism, triglyceride absorption, triglyceride secretion, and cholesterol

metabolism have been examined in non-diabetic mice and hamsters (see section 1.4.1.1), data is

lacking as to the effect of GLP-1R activation in diabetic mice. Intestinal triglyceride and

cholesterol absorption may be examined by Western blotting of APOB and by colorimetric

methods for triglyceride and cholesterol concentration in plasma during fasting and after an oral

lipid load in diabetic Apoe-/-

mice given tyloxapol to inhibit LPL and the breakdown of

triglycerides. Plasma from these experiments may also be pooled and separated on a gel

filtration column to identify triglyceride and cholesterol concentrations in VLDL, IDL/LDL and

HDL. Triglyceride and cholesterol levels in stools (by colorimetric method) give information

about triglyceride absorption and cholesterol reabsorption from the bile. Lastly, fasting and post-

prandial levels of triglycerides and cholesterol in lipoprotein fractions (VLDL, IDL/LDL and

HDL) during the course of the study (by fractionation through a gel filtration column) may be of

importance to explain the overall cycle and/or liver function.

It is very likely that the reductions in triglycerides and/or cholesterol in the liver in taspoglutide-

treated mice in our studies are mediated indirectly by the brain, or other organs, as no GLP-1R

is expressed in the liver. Thus, further studies may have to examine the effects of

Page 85: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

74

intracerobrovascular (icv) injections of a GLP-1R agonist to record acute effects in triglyceride

and cholesterol accumulation and/or changes in signaling and/or expression of genes and

proteins involved in lipoprotein and cholesterol metabolism in the liver. For example, levels of

APOB protein and mRNA transcript levels may be quantified to further examine any role of

GLP-1R activation in the regulation of APOB.

To dissect peripheral and central actions of GLP-1R activation, icv administration of the GLP-

1R antagonist, exendin(9-39) with concurrent i.p. administration of a GLP-1R agonist then

subsequent analysis of livers for triglyceride and cholesterol accumulation, plus genes and

proteins involved in lipoprotein and cholesterol metabolism may give information as to whether

the induced changes in the liver are mediated by the brain.

Upon conclusion of these studies, a much better understanding of the physiological role of the

GLP-1R in the immune system and lipoprotein metabolism could be drawn. This information

could also be applied in clinical practice for treating diabetic patients and/or, perhaps, patients

with non-alcoholic fatty liver disease.

Page 86: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

75

References

Adiels, M., J. Boren, M. J. Caslake, P. Stewart, A. Soro, J. Westerbacka, B. Wennberg, S. O.

Olofsson, C. Packard, and M. R. Taskinen. 2005. "Overproduction of VLDL1 driven by

hyperglycemia is a dominant feature of diabetic dyslipidemia." Arteriosclerosis,

thrombosis, and vascular biology no. 25 (8):1697-703. doi:

10.1161/01.ATV.0000172689.53992.25.

Alhadeff, Amber, Laura Rupprecht, and Matthew Hayes. 2012. "GLP-1 Neurons in the Nucleus

of the Solitary Tract Project Directly to the Ventral Tegmental Area and Nucleus

Accumbens to Control for Food Intake." Endocrinology no. 153 (2):647-705. doi:

10.1210/en.2011-1443.

Arakawa, M., T. Mita, K. Azuma, C. Ebato, H. Goto, T. Nomiyama, Y. Fujitani, T. Hirose, R.

Kawamori, and H. Watada. 2010. "Inhibition of monocyte adhesion to endothelial cells

and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist,

exendin-4." Diabetes no. 59 (4):1030-7. doi: 10.2337/db09-1694.

Baggio, L. L., and D. J. Drucker. 2007. "Biology of incretins: GLP-1 and GIP."

Gastroenterology no. 132 (6):2131-57. doi: 10.1053/j.gastro.2007.03.054.

Balks, H., J. Holst, A. von zur Mühlen, and G. Brabant. 1997. "Rapid oscillations in plasma

glucagon-like peptide-1 (GLP-1) in humans: cholinergic control of GLP-1 secretion via

muscarinic receptors." The Journal of clinical endocrinology and metabolism no. 82

(3):786-876.

Ban, K., S. Hui, D. J. Drucker, and M. Husain. 2009. "Cardiovascular consequences of drugs

used for the treatment of diabetes: potential promise of incretin-based therapies."

Journal of the American Society of Hypertension : JASH no. 3 (4):245-59. doi:

10.1016/j.jash.2009.04.001.

Ban, Kiwon, M. Noyan-Ashraf, Judith Hoefer, Steffen-Sebastian Bolz, Daniel Drucker, and

Mansoor Husain. 2008. "Cardioprotective and vasodilatory actions of glucagon-like

peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent

and -independent pathways." Circulation no. 117 (8867337f-9c43-68db-3ba6-

941232d2c29e):2340-2390. doi: 10.1161/circulationaha.107.739938.

Barton, B. E. 1996. "The biological effects of interleukin 6." Medicinal research reviews no. 16

(1):87-109. doi: 10.1002/(SICI)1098-1128(199601)16:1<87::AID-MED3>3.0.CO;2-Q.

Basu, Ananda, Nisha Charkoudian, William Schrage, Robert Rizza, Rita Basu, and Michael

Joyner. 2007. "Beneficial effects of GLP-1 on endothelial function in humans:

dampening by glyburide but not by glimepiride." American journal of physiology.

Endocrinology and metabolism no. 293 (75061ebf-891f-9473-fbd8-d0fff01d0c36):95.

doi: 10.1152/ajpendo.00373.2007.

Basu, S. K., M. S. Brown, Y. K. Ho, R. J. Havel, and J. L. Goldstein. 1981. "Mouse

macrophages synthesize and secrete a protein resembling apolipoprotein E."

Proceedings of the National Academy of Sciences of the United States of America no. 78

(12):7545-9.

Basu, S. K., Y. K. Ho, M. S. Brown, D. W. Bilheimer, R. G. Anderson, and J. L. Goldstein.

1982. "Biochemical and genetic studies of the apoprotein E secreted by mouse

macrophages and human monocytes." The Journal of biological chemistry no. 257

(16):9788-95.

Best, J. H., B. J. Hoogwerf, W. H. Herman, E. M. Pelletier, D. B. Smith, M. Wenten, and M. A.

Hussein. 2011. "Risk of cardiovascular disease events in patients with type 2 diabetes

prescribed the glucagon-like peptide 1 (GLP-1) receptor agonist exenatide twice daily or

Page 87: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

76

other glucose-lowering therapies: a retrospective analysis of the LifeLink database."

Diabetes care no. 34 (1):90-5. doi: 10.2337/dc10-1393.

Bobryshev, Y. V. 2000. "Dendritic cells and their involvement in atherosclerosis." Current

opinion in lipidology no. 11 (5):511-7.

Bodary, P. F., S. Gu, Y. Shen, A. H. Hasty, J. M. Buckler, and D. T. Eitzman. 2005.

"Recombinant leptin promotes atherosclerosis and thrombosis in apolipoprotein E-

deficient mice." Arteriosclerosis, thrombosis, and vascular biology no. 25 (8):e119-22.

doi: 10.1161/01.ATV.0000173306.47722.ec.

Bornfeldt, K. E., and I. Tabas. 2011. "Insulin resistance, hyperglycemia, and atherosclerosis."

Cell metabolism no. 14 (5):575-85. doi: 10.1016/j.cmet.2011.07.015.

Bouloumie, A., T. Marumo, M. Lafontan, and R. Busse. 1999. "Leptin induces oxidative stress

in human endothelial cells." FASEB journal : official publication of the Federation of

American Societies for Experimental Biology no. 13 (10):1231-8.

Brownlee, M. 2001. "Biochemistry and molecular cell biology of diabetic complications."

Nature no. 414 (6865):813-20. doi: 10.1038/414813a.

Brubaker, P. L. 2006. "The glucagon-like peptides: pleiotropic regulators of nutrient

homeostasis." Annals of the New York Academy of Sciences no. 1070:10-26. doi:

10.1196/annals.1317.006.

Brufau, Gemma, Albert Groen, and Folkert Kuipers. 2011. "Reverse cholesterol transport

revisited: contribution of biliary versus intestinal cholesterol excretion." Arteriosclerosis,

thrombosis, and vascular biology no. 31 (c91cc132-3489-715a-c24f-

a75f25e783d5):1726-1759. doi: 10.1161/atvbaha.108.181206.

Bullock, B., R. Heller, and J. Habener. 1996. "Tissue distribution of messenger ribonucleic acid

encoding the rat glucagon-like peptide-1 receptor." Endocrinology no. 137 (a54c2538-

d295-5a90-55d9-9168f9f37958):2968-3046.

Bunck, Mathijs, Michaela Diamant, Bjorn Eliasson, Anja Cornér, Rimma Shaginian, Robert

Heine, Marja-Riitta Taskinen, Hannele Yki-Järvinen, and Ulf Smith. 2010. "Exenatide

affects circulating cardiovascular risk biomarkers independently of changes in body

composition." Diabetes care no. 33 (5f913fb5-98c5-e3d8-6559-bfe78af81d27):1734-

1741. doi: 10.2337/dc09-2361.

Burmeister, Melissa, Tura Ferre, Jennifer Ayala, Emily King, Rochelle Holt, and Julio Ayala.

2012. "Acute activation of central GLP-1 receptors enhances hepatic insulin action and

insulin secretion in high-fat-fed, insulin resistant mice." American journal of physiology.

Endocrinology and metabolism no. 302 (84724815-7a00-201e-9806-21a4b92f7d8b):43.

doi: 10.1152/ajpendo.00409.2011.

Ceriello, A., L. Quagliaro, M. D'Amico, C. Di Filippo, R. Marfella, F. Nappo, L. Berrino, F.

Rossi, and D. Giugliano. 2002. "Acute hyperglycemia induces nitrotyrosine formation

and apoptosis in perfused heart from rat." Diabetes no. 51 (4):1076-82.

Ceriello, Antonio, Katherine Esposito, Roberto Testa, Anna Bonfigli, Maurizio Marra, and

Dario Giugliano. 2011. "The possible protective role of glucagon-like peptide 1 on

endothelium during the meal and evidence for an "endothelial resistance" to glucagon-

like peptide 1 in diabetes." Diabetes care no. 34 (22f9419b-4c29-c6ad-0d59-

d1e2f18058aa):697-1399. doi: 10.2337/dc10-1949.

Chan, J. L., K. Heist, A. M. DePaoli, J. D. Veldhuis, and C. S. Mantzoros. 2003. "The role of

falling leptin levels in the neuroendocrine and metabolic adaptation to short-term

starvation in healthy men." The Journal of clinical investigation no. 111 (9):1409-21.

doi: 10.1172/JCI17490.

Page 88: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

77

Chaudhuri, A., H. Ghanim, M. Vora, C. L. Sia, K. Korzeniewski, S. Dhindsa, A. Makdissi, and

P. Dandona. 2012. "Exenatide exerts a potent antiinflammatory effect." The Journal of

clinical endocrinology and metabolism no. 97 (1):198-207. doi: 10.1210/jc.2011-1508.

Christian, Rask-Madsen, and L. King George. 2007. "Mechanisms of Disease: endothelial

dysfunction in insulin resistance and diabetes." Nature clinical practice. Endocrinology

& metabolism no. 3 (2701c722-2fc0-5fe0-e469-c10a3df1cc9b):46-102. doi:

10.1038/ncpendmet0366.

Chrysant, S. G. 2010. "The role of Angiotensin receptor blocker and calcium channel blocker

combination therapy in treating hypertension: focus on recent studies." American journal

of cardiovascular drugs : drugs, devices, and other interventions no. 10 (5):315-20. doi:

10.2165/11538850-000000000-00000.

Cohen, Mark, Sandra Ellis, Carel Le Roux, Rachel Batterham, Adrian Park, Michael Patterson,

Gary Frost, Mohammad Ghatei, and Stephen Bloom. 2003. "Oxyntomodulin suppresses

appetite and reduces food intake in humans." The Journal of clinical endocrinology and

metabolism no. 88 (10):4696-5397.

Collins, T., and M. I. Cybulsky. 2001. "NF-kappaB: pivotal mediator or innocent bystander in

atherogenesis?" The Journal of clinical investigation no. 107 (3):255-64. doi:

10.1172/JCI10373.

Control, Group, F. Turnbull, C. Abraira, R. Anderson, R. Byington, J. Chalmers, W.

Duckworth, G. Evans, H. Gerstein, R. Holman, T. Moritz, B. Neal, T. Ninomiya, A.

Patel, S. Paul, F. Travert, and M. Woodward. 2009. "Intensive glucose control and

macrovascular outcomes in type 2 diabetes." Diabetologia no. 52 (14d640b7-6612-de46-

1e52-21a4b85659f1):2288-2386. doi: 10.1007/s00125-009-1470-0.

Copley, K., K. McCowen, R. Hiles, L. L. Nielsen, A. Young, and D. G. Parkes. 2006.

"Investigation of exenatide elimination and its in vivo and in vitro degradation." Current

drug metabolism no. 7 (4):367-74.

Courrèges, J. P., T. Vilsbøll, M. Zdravkovic, T. Le-Thi, T. Krarup, O. Schmitz, R. Verhoeven, I.

Bugáñová, and S. Madsbad. 2008. "Beneficial effects of once-daily liraglutide, a human

glucagon-like peptide-1 analogue, on cardiovascular risk biomarkers in patients with

Type 2 diabetes." Diabetic medicine : a journal of the British Diabetic Association no.

25 (2b15f024-46a6-2221-431c-c095a0c008ec):1129-1160. doi: 10.1111/j.1464-

5491.2008.02484.x.

Cybulsky, M. I., and J. Jongstra-Bilen. 2010. "Resident intimal dendritic cells and the initiation

of atherosclerosis." Current opinion in lipidology no. 21 (5):397-403. doi:

10.1097/MOL.0b013e32833ded96.

Dakin, Catherine, Caroline Small, Rachel Batterham, Nicola Neary, Mark Cohen, Michael

Patterson, Mohammad Ghatei, and Stephen Bloom. 2004. "Peripheral oxyntomodulin

reduces food intake and body weight gain in rats." Endocrinology no. 145 (6):2687-

2782. doi: 10.1210/en.2003-1338.

Daugherty, A., H. Lu, D. A. Howatt, and D. L. Rateri. 2009. "Modes of defining atherosclerosis

in mouse models: relative merits and evolving standards." Methods in molecular biology

no. 573:1-15. doi: 10.1007/978-1-60761-247-6_1.

Daugherty, A., and S. C. Whitman. 2003. "Quantification of atherosclerosis in mice." Methods

in molecular biology no. 209:293-309.

Deacon, C., M. Nauck, M. Toft-Nielsen, L. Pridal, B. Willms, and J. Holst. 1995. "Both

subcutaneously and intravenously administered glucagon-like peptide I are rapidly

degraded from the NH2-terminus in type II diabetic patients and in healthy subjects."

Diabetes no. 44 (1f50dde3-611d-30b2-02bd-579a1eb6dd21):1126-1157.

Page 89: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

78

Dhanvantari, S., N. Seidah, and P. Brubaker. 1996. "Role of prohormone convertases in the

tissue-specific processing of proglucagon." Molecular endocrinology (Baltimore, Md.)

no. 10 (6ec701d5-701b-d2db-db16-526cea0ea9fc):342-397.

Diakogiannaki, Eleftheria, Fiona Gribble, and Frank Reimann. 2011. "Nutrient detection by

incretin hormone secreting cells." Physiology & behavior. doi:

10.1016/j.physbeh.2011.12.001.

Dominiczak, Marek, and Muriel Caslake. 2011. "Apolipoproteins: metabolic role and clinical

biochemistry applications." Annals of Clinical Biochemistry no. 48 (2817241c-febe-

e666-857b-9cd9a5e175e1):498-1013. doi: 10.1258/acb.2011.011111.

Dong, J. Z., Y. Shen, J. Zhang, N. Tsomaia, D. F. Mierke, and J. E. Taylor. 2011. "Discovery

and characterization of taspoglutide, a novel analogue of human glucagon-like peptide-1,

engineered for sustained therapeutic activity in type 2 diabetes." Diabetes, obesity &

metabolism no. 13 (1):19-25. doi: 10.1111/j.1463-1326.2010.01313.x.

Doran, Amanda, Nahum Meller, and Coleen McNamara. 2008. "Role of smooth muscle cells in

the initiation and early progression of atherosclerosis." Arteriosclerosis, thrombosis, and

vascular biology no. 28 (e91e1821-f07e-23c4-b60c-d3cc60876f43):812-821. doi:

10.1161/atvbaha.107.159327.

Drucker, D., J. Philippe, S. Mojsov, W. Chick, and J. Habener. 1987. "Glucagon-like peptide I

stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell

line." Proceedings of the National Academy of Sciences of the United States of America

no. 84 (10):3434-3442.

Duckworth, W., C. Abraira, T. Moritz, D. Reda, N. Emanuele, P. D. Reaven, F. J. Zieve, J.

Marks, S. N. Davis, R. Hayward, S. R. Warren, S. Goldman, M. McCarren, M. E. Vitek,

W. G. Henderson, and G. D. Huang. 2009. "Glucose control and vascular complications

in veterans with type 2 diabetes." The New England journal of medicine no. 360 (2):129-

39. doi: 10.1056/NEJMoa0808431.

Eissele, R., R. Göke, S. Willemer, H. Harthus, H. Vermeer, R. Arnold, and B. Göke. 1992.

"Glucagon-like peptide-1 cells in the gastrointestinal tract and pancreas of rat, pig and

man." European journal of clinical investigation no. 22 (4):283-374.

Elhage, R., S. Clamens, S. Besnard, Z. Mallat, A. Tedgui, J. Arnal, A. Maret, and F. Bayard.

2001. "Involvement of interleukin-6 in atherosclerosis but not in the prevention of fatty

streak formation by 17beta-estradiol in apolipoprotein E-deficient mice." Atherosclerosis

no. 156 (2):315-20.

Erdogdu, O., D. Nathanson, A. Sjoholm, T. Nystrom, and Q. Zhang. 2010. "Exendin-4

stimulates proliferation of human coronary artery endothelial cells through eNOS-,

PKA- and PI3K/Akt-dependent pathways and requires GLP-1 receptor." Molecular and

cellular endocrinology no. 325 (1-2):26-35. doi: 10.1016/j.mce.2010.04.022.

Farmer, J. A. 2011. "Effects of combination lipid therapy in diabetes mellitus." Current

atherosclerosis reports no. 13 (2):104-6. doi: 10.1007/s11883-011-0162-7.

Farooqi, I. S., G. Matarese, G. M. Lord, J. M. Keogh, E. Lawrence, C. Agwu, V. Sanna, S. A.

Jebb, F. Perna, S. Fontana, R. I. Lechler, A. M. DePaoli, and S. O'Rahilly. 2002.

"Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and

neuroendocrine/metabolic dysfunction of human congenital leptin deficiency." The

Journal of clinical investigation no. 110 (8):1093-103. doi: 10.1172/JCI15693.

Farr, Sarah, and Khosrow Adeli. 2012. "Incretin-based therapies for treatment of postprandial

dyslipidemia in insulin-resistant states." Current opinion in lipidology no. 23 (1):56-117.

doi: 10.1097/MOL.0b013e32834d68f0.

Page 90: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

79

Fazio, S., V. R. Babaev, A. B. Murray, A. H. Hasty, K. J. Carter, L. A. Gleaves, J. B. Atkinson,

and M. F. Linton. 1997. "Increased atherosclerosis in mice reconstituted with

apolipoprotein E null macrophages." Proceedings of the National Academy of Sciences

of the United States of America no. 94 (9):4647-52.

Flock, Grace, Laurie Baggio, Christine Longuet, and Daniel Drucker. 2007. "Incretin receptors

for glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide are

essential for the sustained metabolic actions of vildagliptin in mice." Diabetes no. 56

(3962a46e-f128-bd96-9979-bb465daf3f8a):3006-3019. doi: 10.2337/db07-0697.

Gallwitz, B., J. Guzman, F. Dotta, B. Guerci, R. Simo, B. R. Basson, A. Festa, J. Kiljanski, H.

Sapin, M. Trautmann, and G. Schernthaner. 2012. "Exenatide twice daily versus

glimepiride for prevention of glycaemic deterioration in patients with type 2 diabetes

with metformin failure (EUREXA): an open-label, randomised controlled trial." Lancet

no. 379 (9833):2270-8. doi: 10.1016/S0140-6736(12)60479-6.

Gaspari, Tracey, HongBin Liu, Iresha Welungoda, Yunshan Hu, Robert Widdop, Lotte

Knudsen, Richard Simpson, and Anthony Dear. 2011. "A GLP-1 receptor agonist

liraglutide inhibits endothelial cell dysfunction and vascular adhesion molecule

expression in an ApoE-/- mouse model." Diabetes & vascular disease research : official

journal of the International Society of Diabetes and Vascular Disease no. 8 (de4899c9-

9702-f090-ec4f-b5d7e62b09d3):117-141. doi: 10.1177/1479164111404257.

Gedulin, B. R., S. E. Nikoulina, P. A. Smith, G. Gedulin, L. L. Nielsen, A. D. Baron, D. G.

Parkes, and A. A. Young. 2005. "Exenatide (exendin-4) improves insulin sensitivity and

{beta}-cell mass in insulin-resistant obese fa/fa Zucker rats independent of glycemia and

body weight." Endocrinology no. 146 (4):2069-76. doi: 10.1210/en.2004-1349.

Gerstein, H. C., M. E. Miller, R. P. Byington, D. C. Goff, Jr., J. T. Bigger, J. B. Buse, W. C.

Cushman, S. Genuth, F. Ismail-Beigi, R. H. Grimm, Jr., J. L. Probstfield, D. G. Simons-

Morton, and W. T. Friedewald. 2008. "Effects of intensive glucose lowering in type 2

diabetes." The New England journal of medicine no. 358 (24):2545-59. doi:

10.1056/NEJMoa0802743.

Ghiglione, M., L. Uttenthal, S. George, and S. Bloom. 1984. "How glucagon-like is glucagon-

like peptide-1?" Diabetologia no. 27 (d6ceb933-a43b-4b84-1ddf-8c47aaad0def):599-

1199.

Göke, R., and J. Conlon. 1988. "Receptors for glucagon-like peptide-1(7-36) amide on rat

insulinoma-derived cells." The Journal of endocrinology no. 116 (2b9dabc3-3008-5de0-

5c88-7dda0bb81e06):357-419.

Golpon, H., A. Puechner, T. Welte, P. Wichert, and C. Feddersen. 2001. "Vasorelaxant effect of

glucagon-like peptide-(7-36)amide and amylin on the pulmonary circulation of the rat."

Regulatory peptides no. 102 (ed281f50-693b-460f-33ba-d0f1877eec70):81-87.

Gorboulev, Valentin, Annette Schürmann, Volker Vallon, Helmut Kipp, Alexander Jaschke,

Dirk Klessen, Alexandra Friedrich, Stephan Scherneck, Timo Rieg, Robyn Cunard,

Maike Veyhl-Wichmann, Aruna Srinivasan, Daniela Balen, Davorka Breljak, Rexhep

Rexhepaj, Helen Parker, Fiona Gribble, Frank Reimann, Florian Lang, Stefan Wiese,

Ivan Sabolic, Michael Sendtner, and Hermann Koepsell. 2012. "Na(+)-D-glucose

cotransporter SGLT1 is pivotal for intestinal glucose absorption and glucose-dependent

incretin secretion." Diabetes no. 61 (1):187-283. doi: 10.2337/db11-1029.

Goto, Hiromasa, Takashi Nomiyama, Tomoya Mita, Eisuke Yasunari, Kosuke Azuma, Koji

Komiya, Masayuki Arakawa, Wen Jin, Akio Kanazawa, Ryuzo Kawamori, Yoshio

Fujitani, Takahisa Hirose, and Hirotaka Watada. 2011. "Exendin-4, a glucagon-like

peptide-1 receptor agonist, reduces intimal thickening after vascular injury."

Page 91: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

80

Biochemical and biophysical research communications no. 405 (d4886d6e-0aeb-4baf-

30ae-d3c1c76ca187):79-163. doi: 10.1016/j.bbrc.2010.12.131.

Guller, S., R. E. Corin, D. C. Mynarcik, B. M. London, and M. Sonenberg. 1988. "Role of

insulin in growth hormone-stimulated 3T3 cell adipogenesis." Endocrinology no. 122

(5):2084-9.

Gutniak, M., B. Linde, J. Holst, and S. Efendić. 1994. "Subcutaneous injection of the incretin

hormone glucagon-like peptide 1 abolishes postprandial glycemia in NIDDM." Diabetes

care no. 17 (9):1039-1083.

Gutzwiller, J., B. Göke, J. Drewe, P. Hildebrand, S. Ketterer, D. Handschin, R. Winterhalder, D.

Conen, and C. Beglinger. 1999. "Glucagon-like peptide-1: a potent regulator of food

intake in humans." Gut no. 44 (1):81-87.

Guyton, J. R. 2010. "Combination regimens with statin, niacin, and intestinally active LDL-

lowering drugs: alternatives to high-dose statin therapy?" Current opinion in lipidology

no. 21 (4):372-7. doi: 10.1097/MOL.0b013e32833c1f16.

Hadjiyanni, I., L. L. Baggio, P. Poussier, and D. J. Drucker. 2008. "Exendin-4 modulates

diabetes onset in nonobese diabetic mice." Endocrinology no. 149 (3):1338-49. doi:

10.1210/en.2007-1137.

Hadjiyanni, I., K. A. Siminovitch, J. S. Danska, and D. J. Drucker. 2010. "Glucagon-like

peptide-1 receptor signalling selectively regulates murine lymphocyte proliferation and

maintenance of peripheral regulatory T cells." Diabetologia no. 53 (4):730-40.

Hamilton, S. J., G. T. Chew, and G. F. Watts. 2007. "Therapeutic regulation of endothelial

dysfunction in type 2 diabetes mellitus." Diabetes & vascular disease research : official

journal of the International Society of Diabetes and Vascular Disease no. 4 (2):89-102.

doi: 10.3132/dvdr.2007.026.

Hansen, Katrine, Mette Rosenkilde, Filip Knop, Niels Wellner, Thi Diep, Jens Rehfeld, Ulrik

Andersen, Jens Holst, and Harald Hansen. 2011. "2-Oleoyl glycerol is a GPR119 agonist

and signals GLP-1 release in humans." The Journal of clinical endocrinology and

metabolism no. 96 (9):17. doi: 10.1210/jc.2011-0647.

Hansen, L., C. Deacon, C. Orskov, and J. Holst. 1999. "Glucagon-like peptide-1-(7-36)amide is

transformed to glucagon-like peptide-1-(9-36)amide by dipeptidyl peptidase IV in the

capillaries supplying the L cells of the porcine intestine." Endocrinology no. 140

(cf8f2218-ed35-6f8c-5cd2-57a8f0435370):5356-5419.

Hansson, Göran, and Andreas Hermansson. 2011. "The immune system in atherosclerosis."

Nature immunology no. 12 (51e4664e-0754-e304-1e3e-21a4b83cac34):204-216. doi:

10.1038/ni.2001.

Harris, T. B., L. Ferrucci, R. P. Tracy, M. C. Corti, S. Wacholder, W. H. Ettinger, Jr., H.

Heimovitz, H. J. Cohen, and R. Wallace. 1999. "Associations of elevated interleukin-6

and C-reactive protein levels with mortality in the elderly." The American journal of

medicine no. 106 (5):506-12.

Hattori, Y., T. Jojima, A. Tomizawa, H. Satoh, S. Hattori, K. Kasai, and T. Hayashi. 2010. "A

glucagon-like peptide-1 (GLP-1) analogue, liraglutide, upregulates nitric oxide

production and exerts anti-inflammatory action in endothelial cells." Diabetologia no. 53

(e2e2bb73-111a-8361-638c-c79309ff90a0):2256-2319. doi: 10.1007/s00125-010-1831-

8.

Heller, S. R. 2009. "A summary of the ADVANCE Trial." Diabetes care no. 32 Suppl 2:S357-

61. doi: 10.2337/dc09-S339.

Page 92: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

81

Herrmann, C., R. Göke, G. Richter, H. Fehmann, R. Arnold, and B. Göke. 1995. "Glucagon-like

peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in response

to nutrients." Digestion no. 56 (2):117-143.

Hirata, Kunio, Shinji Kume, Shin-ichi Araki, Masayoshi Sakaguchi, Masami Chin-Kanasaki,

Keiji Isshiki, Toshiro Sugimoto, Akira Nishiyama, Daisuke Koya, Masakazu Haneda,

Atsunori Kashiwagi, and Takashi Uzu. 2009. "Exendin-4 has an anti-hypertensive effect

in salt-sensitive mice model." Biochemical and biophysical research communications

no. 380 (1):44-53. doi: 10.1016/j.bbrc.2009.01.003.

Horton, E., A. Cohen, H. Gibson, B. Lamparello, S. Herzlinger, and L. McFarland. 2009.

"Effects of exenatide vs insulin glargine on cardiovascular risk factors in subjects with

type 2 diabetes." Diabetologia no. 52 (401837e1-783f-46cf-085e-c07a2b23340b).

Hotta, K., T. Funahashi, Y. Arita, M. Takahashi, M. Matsuda, Y. Okamoto, H. Iwahashi, H.

Kuriyama, N. Ouchi, K. Maeda, M. Nishida, S. Kihara, N. Sakai, T. Nakajima, K.

Hasegawa, M. Muraguchi, Y. Ohmoto, T. Nakamura, S. Yamashita, T. Hanafusa, and Y.

Matsuzawa. 2000. "Plasma concentrations of a novel, adipose-specific protein,

adiponectin, in type 2 diabetic patients." Arteriosclerosis, thrombosis, and vascular

biology no. 20 (6):1595-9.

Hsieh, J., C. Longuet, C. Baker, B. Qin, L. Federico, D. Drucker, and K. Adeli. 2010. "The

glucagon-like peptide 1 receptor is essential for postprandial lipoprotein synthesis and

secretion in hamsters and mice." Diabetologia no. 53 (3):552-613. doi: 10.1007/s00125-

009-1611-5.

Huber, S. A., P. Sakkinen, D. Conze, N. Hardin, and R. Tracy. 1999. "Interleukin-6 exacerbates

early atherosclerosis in mice." Arteriosclerosis, thrombosis, and vascular biology no. 19

(10):2364-7.

Huff, M. W. 2003. "Dietary cholesterol, cholesterol absorption, postprandial lipemia and

atherosclerosis." The Canadian journal of clinical pharmacology = Journal canadien de

pharmacologie clinique no. 10 Suppl A:26A-32A.

Iakoubov, Roman, Ausma Ahmed, Lina Lauffer, Richard Bazinet, and Patricia Brubaker. 2011.

"Essential role for protein kinase Cζ in oleic acid-induced glucagon-like peptide-1

secretion in vivo in the rat." Endocrinology no. 152 (4):1244-1296. doi:

10.1210/en.2010-1352.

Ishibashi, Yuji, Takanori Matsui, Masayoshi Takeuchi, and Sho-Ichi Yamagishi. 2010.

"Glucagon-like peptide-1 (GLP-1) inhibits advanced glycation end product (AGE)-

induced up-regulation of VCAM-1 mRNA levels in endothelial cells by suppressing

AGE receptor (RAGE) expression." Biochemical and biophysical research

communications no. 391 (83ef0f45-4add-116e-4010-21a4b83bb998):1405-1413. doi:

10.1016/j.bbrc.2009.12.075.

Jarrett, R. J. 1981. "Atherosclerosis, diabetes and obesity." Proc Nutr Soc no. 40 (2):209-12.

doi: S0029665181000347 [pii].

Jawien, J., P. Nastalek, and R. Korbut. 2004. "Mouse models of experimental atherosclerosis."

Journal of physiology and pharmacology : an official journal of the Polish Physiological

Society no. 55 (3):503-17.

Kauth, T., and J. Metz. 1987. "Immunohistochemical localization of glucagon-like peptide 1.

Use of poly- and monoclonal antibodies." Histochemistry no. 86 (5):509-524.

Kieffer, T., and J. Habener. 1999. "The glucagon-like peptides." Endocrine reviews no. 20

(6):876-1789.

Kieffer, T., C. McIntosh, and R. Pederson. 1995. "Degradation of glucose-dependent

insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by

Page 93: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

82

dipeptidyl peptidase IV." Endocrinology no. 136 (a741879a-686e-9681-2266-

57a26fd738ed):3585-3681.

Kim Chung, Le Thi, Toshio Hosaka, Masaki Yoshida, Nagakatsu Harada, Hiroshi Sakaue,

Tohru Sakai, and Yutaka Nakaya. 2009. "Exendin-4, a GLP-1 receptor agonist, directly

induces adiponectin expression through protein kinase A pathway and prevents

inflammatory adipokine expression." Biochemical and biophysical research

communications no. 390 (cba95f03-2238-8fb4-1bb3-bfe78af7b9ae):613-621. doi:

10.1016/j.bbrc.2009.10.015.

Kim, Ja-Young, Esther van de Wall, Mathieu Laplante, Anthony Azzara, Maria Trujillo,

Susanna Hofmann, Todd Schraw, Jorge Durand, Hua Li, Guangyu Li, Linda Jelicks,

Mark Mehler, David Hui, Yves Deshaies, Gerald Shulman, Gary Schwartz, and Philipp

Scherer. 2007. "Obesity-associated improvements in metabolic profile through

expansion of adipose tissue." The Journal of clinical investigation no. 117 (59da6ed9-

e6c9-1d2a-e5c6-c05e6f0e887c):2621-2658. doi: 10.1172/jci31021.

King, V. L., N. W. Hatch, H. W. Chan, M. C. de Beer, F. C. de Beer, and L. R. Tannock. 2010.

"A murine model of obesity with accelerated atherosclerosis." Obesity no. 18 (1):35-41.

doi: 10.1038/oby.2009.176.

Kirkpatrick, P. 2009. "Specificity concerns with antibodies for receptor mapping." Nature

reviews. Drug discovery no. 8 (4):278. doi: 10.1038/nrd2854.

Kodera, R., K. Shikata, H. U. Kataoka, T. Takatsuka, S. Miyamoto, M. Sasaki, N. Kajitani, S.

Nishishita, K. Sarai, D. Hirota, C. Sato, D. Ogawa, and H. Makino. 2011. "Glucagon-

like peptide-1 receptor agonist ameliorates renal injury through its anti-inflammatory

action without lowering blood glucose level in a rat model of type 1 diabetes."

Diabetologia no. 54 (4):965-78. doi: 10.1007/s00125-010-2028-x.

Koenen, Rory, and Christian Weber. 2010. "Therapeutic targeting of chemokine interactions in

atherosclerosis." Nature reviews. Drug discovery no. 9 (42d07fe2-ad29-4836-4dd2-

9bf93db093c4):141-194. doi: 10.1038/nrd3048.

Koh, K. K., S. H. Han, P. C. Oh, E. K. Shin, and M. J. Quon. 2010. "Combination therapy for

treatment or prevention of atherosclerosis: focus on the lipid-RAAS interaction."

Atherosclerosis no. 209 (2):307-13. doi: 10.1016/j.atherosclerosis.2009.09.007.

Kolterman, O. G., D. D. Kim, L. Shen, J. A. Ruggles, L. L. Nielsen, M. S. Fineman, and A. D.

Baron. 2005. "Pharmacokinetics, pharmacodynamics, and safety of exenatide in patients

with type 2 diabetes mellitus." American journal of health-system pharmacy : AJHP :

official journal of the American Society of Health-System Pharmacists no. 62 (2):173-81.

Koska, J., E. A. Schwartz, M. P. Mullin, D. C. Schwenke, and P. D. Reaven. 2010.

"Improvement of Postprandial Endothelial Function After a Single Dose of Exenatide in

Individuals With Impaired Glucose Tolerance and Recent-Onset Type 2 Diabetes."

Diabetes care no. 33 (981f8e80-9888-be31-8890-bc1f6d8e49d1). doi: 10.2337/dc09-

1961.

Kreymann, B., G. Williams, M. Ghatei, and S. Bloom. 1987. "Glucagon-like peptide-1 7-36: a

physiological incretin in man." Lancet no. 2 (8571):1300-1304.

Kwon, Gina, Jamie Schroeder, Marcelo Amar, Alan Remaley, and Robert Balaban. 2008.

"Contribution of macromolecular structure to the retention of low-density lipoprotein at

arterial branch points." Circulation no. 117 (4d93435c-905c-e9cb-160b-

9bca77be39e7):2919-2946. doi: 10.1161/circulationaha.107.754614.

Kypreos, K. E., and V. I. Zannis. 2007. "Pathway of biogenesis of apolipoprotein E-containing

HDL in vivo with the participation of ABCA1 and LCAT." The Biochemical journal no.

403 (2):359-67. doi: 10.1042/BJ20061048.

Page 94: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

83

Lamont, B. J., Y. Li, E. Kwan, T. J. Brown, H. Gaisano, and D. J. Drucker. 2012. "Pancreatic

GLP-1 receptor activation is sufficient for incretin control of glucose metabolism in

mice." The Journal of clinical investigation no. 122 (1):388-402. doi: 10.1172/JCI42497.

Lewis, G. F. 1997. "Fatty acid regulation of very low density lipoprotein production." Current

opinion in lipidology no. 8 (3):146-53.

Lewis, G. F., and G. Steiner. 1996. "Acute effects of insulin in the control of VLDL production

in humans. Implications for the insulin-resistant state." Diabetes care no. 19 (4):390-3.

Li, L., G. Yang, Q. Li, X. Tan, H. Liu, Y. Tang, and G. Boden. 2008. "Exenatide prevents fat-

induced insulin resistance and raises adiponectin expression and plasma levels."

Diabetes, obesity & metabolism no. 10 (92ec143b-7023-1e87-e6e2-bfe78af7604f):921-

951. doi: 10.1111/j.1463-1326.2007.00832.x.

Lindmark, E., E. Diderholm, L. Wallentin, and A. Siegbahn. 2001. "Relationship between

interleukin 6 and mortality in patients with unstable coronary artery disease: effects of an

early invasive or noninvasive strategy." JAMA : the journal of the American Medical

Association no. 286 (17):2107-13.

Liu, Hongbin, Anthony Dear, Lotte Knudsen, and Richard Simpson. 2009. "A long-acting

glucagon-like peptide-1 analogue attenuates induction of plasminogen activator inhibitor

type-1 and vascular adhesion molecules." The Journal of endocrinology no. 201

(320b16c9-b4e0-83ac-939a-21a4b92f8f79):59-125. doi: 10.1677/joe-08-0468.

Liu, S. C., Y. K. Tu, M. N. Chien, and K. L. Chien. 2012. "Effect of antidiabetic agents added to

metformin on glycemic control, hypoglycemia and weight change in patients with type 2

diabetes: a network meta-analysis." Diabetes, obesity & metabolism. doi:

10.1111/j.1463-1326.2012.01606.x.

Lu, Wendell, Qing Yang, Li Yang, Dana Lee, David D'Alessio, and Patrick Tso. 2012.

"Chylomicron Formation and Secretion is Required for Lipid-Stimulated Release of

Incretins GLP-1 and GIP." Lipids (bc43b27e-ef5e-ae67-c054-28f7583d85d7). doi:

10.1007/s11745-011-3650-1.

Lusis, A. J. 2000. "Atherosclerosis." Nature no. 407 (6801):233-41. doi: 10.1038/35025203.

Macisaac, Richard, and George Jerums. 2011. "Intensive glucose control and cardiovascular

outcomes in type 2 diabetes." Heart, lung & circulation no. 20 (6105db66-556b-326c-

9b40-9c2304d45a03):647-701. doi: 10.1016/j.hlc.2010.07.013.

Mallat, Z., A. Gojova, V. Brun, B. Esposito, N. Fournier, F. Cottrez, A. Tedgui, and H. Groux.

2003. "Induction of a regulatory T cell type 1 response reduces the development of

atherosclerosis in apolipoprotein E-knockout mice." Circulation no. 108 (10):1232-7.

doi: 10.1161/01.CIR.0000089083.61317.A1.

Marguet, D., L. Baggio, T. Kobayashi, A. Bernard, M. Pierres, P. Nielsen, U. Ribel, T.

Watanabe, D. Drucker, and N. Wagtmann. 2000. "Enhanced insulin secretion and

improved glucose tolerance in mice lacking CD26." Proceedings of the National

Academy of Sciences of the United States of America no. 97 (07c6e788-346a-2dd9-566e-

57abd9e5f466):6874-6883. doi: 10.1073/pnas.120069197.

Matikainen, N., S. Mänttäri, A. Schweizer, A. Ulvestad, D. Mills, B. Dunning, J. Foley, and M.

R. Taskinen. 2006. "Vildagliptin therapy reduces postprandial intestinal triglyceride-rich

lipoprotein particles in patients with type 2 diabetes." Diabetologia no. 49 (9):2049-

2106. doi: 10.1007/s00125-006-0340-2.

Matsubara, J., S. Sugiyama, K. Sugamura, T. Nakamura, Y. Fujiwara, E. Akiyama, H.

Kurokawa, T. Nozaki, K. Ohba, M. Konishi, H. Maeda, Y. Izumiya, K. Kaikita, H.

Sumida, H. Jinnouchi, K. Matsui, S. Kim-Mitsuyama, M. Takeya, and H. Ogawa. 2012.

"A dipeptidyl peptidase-4 inhibitor, des-fluoro-sitagliptin, improves endothelial function

Page 95: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

84

and reduces atherosclerotic lesion formation in apolipoprotein E-deficient mice."

Journal of the American College of Cardiology no. 59 (3):265-76. doi:

10.1016/j.jacc.2011.07.053.

Maury, E., and S. Brichard. 2010. "Adipokine dysregulation, adipose tissue inflammation and

metabolic syndrome." Molecular and cellular endocrinology no. 314 (d132956f-6b2b-

e92e-5d3f-b00e1b576538):1-17. doi: 10.1016/j.mce.2009.07.031.

Mayo, Kelly, Laurence Miller, Dominique Bataille, Stéphane Dalle, Burkhard Göke, Bernard

Thorens, and Daniel Drucker. 2003. "International Union of Pharmacology. XXXV. The

glucagon receptor family." Pharmacological reviews no. 55 (1861855a-8aa4-67ee-67a8-

8d129586585e):167-261. doi: 10.1124/pr.55.1.6.

McGill, H. C., Jr., C. A. McMahan, E. E. Herderick, A. W. Zieske, G. T. Malcom, R. E. Tracy,

and J. P. Strong. 2002. "Obesity accelerates the progression of coronary atherosclerosis

in young men." Circulation no. 105 (23):2712-8.

Meier, J., A. Gethmann, O. Götze, B. Gallwitz, J. Holst, W. Schmidt, and M. Nauck. 2006.

"Glucagon-like peptide 1 abolishes the postprandial rise in triglyceride concentrations

and lowers levels of non-esterified fatty acids in humans." Diabetologia no. 49 (3):452-

460. doi: 10.1007/s00125-005-0126-y.

Meier, Juris, Michael Nauck, Daniel Kranz, Jens Holst, Carolyn Deacon, Dirk Gaeckler,

Wolfgang Schmidt, and Baptist Gallwitz. 2004. "Secretion, degradation, and elimination

of glucagon-like peptide 1 and gastric inhibitory polypeptide in patients with chronic

renal insufficiency and healthy control subjects." Diabetes no. 53 (527b5c6a-ad98-cff0-

ea48-57d0e5f089ed):654-716.

Meir, K. S., and E. Leitersdorf. 2004. "Atherosclerosis in the apolipoprotein-E-deficient mouse:

a decade of progress." Arteriosclerosis, thrombosis, and vascular biology no. 24

(6):1006-14. doi: 10.1161/01.ATV.0000128849.12617.f4.

Mells, Jamie, Ping Fu, Shvetank Sharma, Darin Olson, Lihong Cheng, Jeffrey Handy, Neeraj

Saxena, Dan Sorescu, and Frank Anania. 2012. "Glp-1 analog, liraglutide, ameliorates

hepatic steatosis and cardiac hypertrophy in C57BL/6J mice fed a Western diet."

American journal of physiology. Gastrointestinal and liver physiology no. 302

(7a8890b8-471e-7a77-830b-bb4eba277a26):35. doi: 10.1152/ajpgi.00274.2011.

Mensenkamp, A. R., M. C. Jong, H. van Goor, M. J. van Luyn, V. Bloks, R. Havinga, P. J.

Voshol, M. H. Hofker, K. W. van Dijk, L. M. Havekes, and F. Kuipers. 1999.

"Apolipoprotein E participates in the regulation of very low density lipoprotein-

triglyceride secretion by the liver." The Journal of biological chemistry no. 274

(50):35711-8.

Mentlein, R. 1999. "Dipeptidyl-peptidase IV (CD26)--role in the inactivation of regulatory

peptides." Regulatory peptides no. 85 (db5efb24-40b1-9e8f-44c3-57aaad452215):9-33.

Menzaghi, C., V. Trischitta, and A. Doria. 2007. "Genetic influences of adiponectin on insulin

resistance, type 2 diabetes, and cardiovascular disease." Diabetes no. 56 (5):1198-209.

doi: 10.2337/db06-0506.

Mojsov, S., G. Weir, and J. Habener. 1987. "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." The Journal of clinical investigation no. 79 (2):616-625. doi:

10.1172/jci112855.

Molecular Mechanisms of Atherosclerosis. 2005. Edited by Joseph Loscalzo: Taylor & Francis.

Moore, Kathryn, and Ira Tabas. 2011. "Macrophages in the pathogenesis of atherosclerosis."

Cell no. 145 (60dc9887-ab1d-35ee-eb7e-9bbcd054a565):341-396. doi:

10.1016/j.cell.2011.04.005.

Page 96: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

85

"MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk

individuals: a randomised placebo-controlled trial." 2002. Lancet no. 360 (9326):7-22.

doi: 10.1016/S0140-6736(02)09327-3.

Murthy, S. N., R. C. Hilaire, D. B. Casey, A. M. Badejo, J. McGee, D. B. McNamara, P. J.

Kadowitz, and V. A. Fonseca. 2010. "The synthetic GLP-I receptor agonist, exenatide,

reduces intimal hyperplasia in insulin resistant rats." Diabetes & vascular disease

research : official journal of the International Society of Diabetes and Vascular Disease

no. 7 (2):138-44. doi: 10.1177/1479164109360269.

Nadeau, K. J., J. G. Regensteiner, T. A. Bauer, M. S. Brown, J. L. Dorosz, A. Hull, P. Zeitler, B.

Draznin, and J. E. Reusch. 2010. "Insulin resistance in adolescents with type 1 diabetes

and its relationship to cardiovascular function." The Journal of clinical endocrinology

and metabolism no. 95 (2):513-21. doi: 10.1210/jc.2009-1756.

Nadeau, K. J., and J. E. Reusch. 2011. "Cardiovascular function/dysfunction in adolescents with

type 1 diabetes." Current diabetes reports no. 11 (3):185-92. doi: 10.1007/s11892-011-

0180-4.

Nagashima, M., T. Watanabe, M. Terasaki, M. Tomoyasu, K. Nohtomi, J. Kim-Kaneyama, A.

Miyazaki, and T. Hirano. 2011. "Native incretins prevent the development of

atherosclerotic lesions in apolipoprotein E knockout mice." Diabetologia no. 54

(10):2649-59. doi: 10.1007/s00125-011-2241-2.

Nakashima, Y., A. S. Plump, E. W. Raines, J. L. Breslow, and R. Ross. 1994. "ApoE-deficient

mice develop lesions of all phases of atherosclerosis throughout the arterial tree."

Arteriosclerosis and thrombosis : a journal of vascular biology / American Heart

Association no. 14 (1):133-40.

Nathan, D. M., P. A. Cleary, J. Y. Backlund, S. M. Genuth, J. M. Lachin, T. J. Orchard, P.

Raskin, and B. Zinman. 2005. "Intensive diabetes treatment and cardiovascular disease

in patients with type 1 diabetes." The New England journal of medicine no. 353

(25):2643-53. doi: 10.1056/NEJMoa052187.

Nathanson, David, Ozlem Erdogdu, John Pernow, Qimin Zhang, and Thomas Nyström. 2009.

"Endothelial dysfunction induced by triglycerides is not restored by exenatide in rat

conduit arteries ex vivo." Regulatory peptides no. 157 (654a05a8-3818-c3a1-ae00-

d1fbf95d2f3f):8-21. doi: 10.1016/j.regpep.2009.07.003.

Nauck, M., M. Heimesaat, C. Orskov, J. Holst, R. Ebert, and W. Creutzfeldt. 1993. "Preserved

incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human

gastric inhibitory polypeptide in patients with type-2 diabetes mellitus." The Journal of

clinical investigation no. 91 (1):301-308. doi: 10.1172/jci116186.

Nawrocki, Andrea, Susanna Hofmann, Daniel Teupser, Joshua Basford, Jorge Durand, Linda

Jelicks, Connie Woo, George Kuriakose, Stephen Factor, Herbert Tanowitz, David Hui,

Ira Tabas, and Philipp Scherer. 2010. "Lack of association between adiponectin levels

and atherosclerosis in mice." Arteriosclerosis, thrombosis, and vascular biology no. 30

(c306f917-2b91-9ec7-0e9f-c05521c12f66):1159-1224. doi:

10.1161/atvbaha.109.195826.

Noyan-Ashraf, M. H., M. A. Momen, K. Ban, A. M. Sadi, Y. Q. Zhou, A. M. Riazi, L. L.

Baggio, R. M. Henkelman, M. Husain, and D. J. Drucker. 2009. "GLP-1R agonist

liraglutide activates cytoprotective pathways and improves outcomes after experimental

myocardial infarction in mice." Diabetes no. 58 (4):975-83. doi: 10.2337/db08-1193.

Nyström, Thomas, Mark Gutniak, Qimin Zhang, Fan Zhang, Jens Holst, Bo Ahrén, and Ake

Sjöholm. 2004. "Effects of glucagon-like peptide-1 on endothelial function in type 2

diabetes patients with stable coronary artery disease." American journal of physiology.

Page 97: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

86

Endocrinology and metabolism no. 287 (12609a2e-4e31-dee3-39cd-940470eabba8):15.

doi: 10.1152/ajpendo.00237.2004.

Oeseburg, Hisko, Rudolf de Boer, Hendrik Buikema, Pim van der Harst, Wiek van Gilst, and

Herman Silljé. 2010. "Glucagon-like peptide 1 prevents reactive oxygen species-induced

endothelial cell senescence through the activation of protein kinase A." Arteriosclerosis,

thrombosis, and vascular biology no. 30 (ed9ec34b-b3c7-d8f8-8c91-

d03daf7eeb04):1407-1421. doi: 10.1161/atvbaha.110.206425.

Oral, Elif, Vinaya Simha, Elaine Ruiz, Alexa Andewelt, Ahalya Premkumar, Peter Snell,

Anthony Wagner, Alex DePaoli, Marc Reitman, Simeon Taylor, Phillip Gorden, and

Abhimanyu Garg. 2002. "Leptin-replacement therapy for lipodystrophy." The New

England journal of medicine no. 346 (95f37c65-b5b1-d57b-00f0-b06acc1a22af):570-

578. doi: 10.1056/NEJMoa012437.

Orchard, T. J., J. C. Olson, J. R. Erbey, K. Williams, K. Y. Forrest, L. Smithline Kinder, D.

Ellis, and D. J. Becker. 2003. "Insulin resistance-related factors, but not glycemia,

predict coronary artery disease in type 1 diabetes: 10-year follow-up data from the

Pittsburgh Epidemiology of Diabetes Complications Study." Diabetes care no. 26

(5):1374-9.

Orskov, C., J. Andreasen, and J. Holst. 1992. "All products of proglucagon are elevated in

plasma from uremic patients." The Journal of clinical endocrinology and metabolism no.

74 (152c2d66-3b76-5b7a-2c86-57cb8997fc1b):379-463.

Orskov, C., A. Wettergren, and J. Holst. 1993. "Biological effects and metabolic rates of

glucagonlike peptide-1 7-36 amide and glucagonlike peptide-1 7-37 in healthy subjects

are indistinguishable." Diabetes no. 42 (96be2e5f-3463-1a95-0cf2-5277e4bf6f5f):658-

719.

Paffen, Elaine, and Moniek DeMaat. 2006. "C-reactive protein in atherosclerosis: A causal

factor?" Cardiovascular research no. 71 (f402a1ca-db1e-fd46-7083-b0bc0f246cab):30-

39. doi: 10.1016/j.cardiores.2006.03.004.

Park, H. Y., H. M. Kwon, H. J. Lim, B. K. Hong, J. Y. Lee, B. E. Park, Y. Jang, S. Y. Cho, and

H. S. Kim. 2001. "Potential role of leptin in angiogenesis: leptin induces endothelial cell

proliferation and expression of matrix metalloproteinases in vivo and in vitro."

Experimental & molecular medicine no. 33 (2):95-102.

Parker, Helen, Frank Reimann, and Fiona Gribble. 2010. "Molecular mechanisms underlying

nutrient-stimulated incretin secretion." Expert reviews in molecular medicine no. 12. doi:

10.1017/s146239940900132x.

Parlevliet, Edwin, Janny Schröder-van der Elst, Eleonora Corssmit, Kristen Picha, Karyn

O'Neil, Vedrana Stojanovic-Susulic, Tatiana Ort, Louis Havekes, Johannes Romijn, and

Hanno Pijl. 2009. "CNTO736, a novel glucagon-like peptide-1 receptor agonist,

ameliorates insulin resistance and inhibits very low-density lipoprotein production in

high-fat-fed mice." The Journal of pharmacology and experimental therapeutics no. 328

(a1057c20-2aa2-9b3d-0505-bb550b1072d4):240-248. doi: 10.1124/jpet.108.144154.

Paulson, K. E., S. N. Zhu, M. Chen, S. Nurmohamed, J. Jongstra-Bilen, and M. I. Cybulsky.

2010. "Resident intimal dendritic cells accumulate lipid and contribute to the initiation of

atherosclerosis." Circulation research no. 106 (2):383-90. doi:

10.1161/CIRCRESAHA.109.210781.

Pellissier, S., K. Sasaki, D. Le-Nguyen, D. Bataille, and C. Jarrousse. 2004. "Oxyntomodulin

and glicentin are potent inhibitors of the fed motility pattern in small intestine."

Neurogastroenterology and motility : the official journal of the European

Page 98: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

87

Gastrointestinal Motility Society no. 16 (4):455-63. doi: 10.1111/j.1365-

2982.2004.00528.x.

Persson, K., R. Gingerich, S. Nayak, K. Wada, E. Wada, and B. Ahrén. 2000. "Reduced GLP-1

and insulin responses and glucose intolerance after gastric glucose in GRP receptor-

deleted mice." American journal of physiology. Endocrinology and metabolism no. 279

(5):62.

Pischon, T., C. J. Girman, G. S. Hotamisligil, N. Rifai, F. B. Hu, and E. B. Rimm. 2004.

"Plasma adiponectin levels and risk of myocardial infarction in men." JAMA : the

journal of the American Medical Association no. 291 (14):1730-7. doi:

10.1001/jama.291.14.1730.

Plump, A. S., J. D. Smith, T. Hayek, K. Aalto-Setala, A. Walsh, J. G. Verstuyft, E. M. Rubin,

and J. L. Breslow. 1992. "Severe hypercholesterolemia and atherosclerosis in

apolipoprotein E-deficient mice created by homologous recombination in ES cells." Cell

no. 71 (2):343-53.

Qin, Xiaofa, Hui Shen, Min Liu, Qing Yang, Shuqin Zheng, Mary Sabo, David D'Alessio, and

Patrick Tso. 2005. "GLP-1 reduces intestinal lymph flow, triglyceride absorption, and

apolipoprotein production in rats." American journal of physiology. Gastrointestinal and

liver physiology no. 288 (6426c56c-a6a2-a0ff-84e8-bb44dd6a8e24):9. doi:

10.1152/ajpgi.00303.2004.

Ramnanan, C., D. Edgerton, G. Kraft, and A. Cherrington. 2011. "Physiologic action of

glucagon on liver glucose metabolism." Diabetes, obesity & metabolism no. 13 Suppl

1:118-143. doi: 10.1111/j.1463-1326.2011.01454.x.

Rask-Madsen, C., E. Buonomo, Q. Li, K. Park, A. C. Clermont, O. Yerokun, M. Rekhter, and

G. L. King. 2012. "Hyperinsulinemia Does Not Change Atherosclerosis Development in

Apolipoprotein E Null Mice." Arteriosclerosis, thrombosis, and vascular biology. doi:

10.1161/ATVBAHA.111.239558.

Rask-Madsen, C., Q. Li, B. Freund, D. Feather, R. Abramov, I. H. Wu, K. Chen, J. Yamamoto-

Hiraoka, J. Goldenbogen, K. B. Sotiropoulos, A. Clermont, P. Geraldes, C. Dall'Osso, A.

J. Wagers, P. L. Huang, M. Rekhter, R. Scalia, C. R. Kahn, and G. L. King. 2010. "Loss

of insulin signaling in vascular endothelial cells accelerates atherosclerosis in

apolipoprotein E null mice." Cell metabolism no. 11 (5):379-89. doi:

10.1016/j.cmet.2010.03.013.

Ray, K., C. Cannon, and E. Braunwald. 2007. "Recent trials of lipid lowering." International

journal of clinical practice no. 61 (13555abd-7d8a-f35a-a566-b57d022b68b0):1145-

1204. doi: 10.1111/j.1742-1241.2007.01425.x.

Raz, I., V. Fonseca, M. Kipnes, L. Durrwell, J. Hoekstra, M. Boldrin, and R. Balena. 2012.

"Efficacy and Safety of Taspoglutide Monotherapy in Drug-Naive Type 2 Diabetic

Patients After 24 Weeks of Treatment: Results of a randomized, double-blind, placebo-

controlled phase 3 study (T-emerge 1)." Diabetes care no. 35 (3):485-7. doi:

10.2337/dc11-1942.

Renard, C. B., F. Kramer, F. Johansson, N. Lamharzi, L. R. Tannock, M. G. von Herrath, A.

Chait, and K. E. Bornfeldt. 2004. "Diabetes and diabetes-associated lipid abnormalities

have distinct effects on initiation and progression of atherosclerotic lesions." The

Journal of clinical investigation no. 114 (5):659-68. doi: 10.1172/JCI17867.

Richter, G., O. Feddersen, U. Wagner, P. Barth, R. Göke, and B. Göke. 1993. "GLP-1 stimulates

secretion of macromolecules from airways and relaxes pulmonary artery." The American

journal of physiology no. 265 (76436e7c-b3d5-9638-c2f0-21a4b9318c93):81.

Page 99: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

88

Ritchie, S. A., M. A. Ewart, C. G. Perry, J. M. Connell, and I. P. Salt. 2004. "The role of insulin

and the adipocytokines in regulation of vascular endothelial function." Clinical science

no. 107 (6):519-32. doi: 10.1042/CS20040190.

Rizzo, M., A. A. Rizvi, G. A. Spinas, G. B. Rini, and K. Berneis. 2009. "Glucose lowering and

anti-atherogenic effects of incretin-based therapies: GLP-1 analogues and DPP-4-

inhibitors." Expert opinion on investigational drugs no. 18 (10):1495-503. doi:

10.1517/14728220903241633.

Roberge, J., and P. Brubaker. 1993. "Regulation of intestinal proglucagon-derived peptide

secretion by glucose-dependent insulinotropic peptide in a novel enteroendocrine loop."

Endocrinology no. 133 (1):233-273.

Roberge, J., K. Gronau, and P. Brubaker. 1996. "Gastrin-releasing peptide is a novel mediator of

proximal nutrient-induced proglucagon-derived peptide secretion from the distal gut."

Endocrinology no. 137 (6):2383-2391.

Rocca, A., and P. Brubaker. 1999. "Role of the vagus nerve in mediating proximal nutrient-

induced glucagon-like peptide-1 secretion." Endocrinology no. 140 (4):1687-1781.

Rodbell, M., L. Birnbaumer, S. L. Pohl, and F. Sundby. 1971. "The reaction of glucagon with its

receptor: evidence for discrete regions of activity and binding in the glucagon molecule."

Proceedings of the National Academy of Sciences of the United States of America no. 68

(5):909-13.

Ross, R. 1997. "Cellular and molecular studies of atherogenesis." Atherosclerosis no. 131 Suppl

(c4757ce7-1cf2-56ac-1463-9bad58b9e482):4.

———. 1999. "Atherosclerosis is an inflammatory disease." American heart journal no. 138

(451fb382-1ab3-9e2e-7415-9bad58b84ec9):20.

Rubenfire, Melvyn, Robert Brook, and Robert Rosenson. 2010. "Treating mixed hyperlipidemia

and the atherogenic lipid phenotype for prevention of cardiovascular events." The

American journal of medicine no. 123 (10):892-900. doi:

10.1016/j.amjmed.2010.03.024.

Ruderman, N., and C. Haudenschild. 1984. "Diabetes as an atherogenic factor." Progress in

cardiovascular diseases no. 26 (5):373-785.

Ruiz-Grande, C., C. Alarcón, A. Alcántara, C. Castilla, J. López Novoa, M. Villanueva-

Peñacarrillo, and I. Valverde. 1993. "Renal catabolism of truncated glucagon-like

peptide 1." Hormone and metabolic research = Hormon- und Stoffwechselforschung =

Hormones et métabolisme no. 25 (dd8feedc-a6af-b675-2b9e-57c88b13d514):612-618.

doi: 10.1055/s-2007-1002190.

Rus, H. G., R. Vlaicu, and F. Niculescu. 1996. "Interleukin-6 and interleukin-8 protein and gene

expression in human arterial atherosclerotic wall." Atherosclerosis no. 127 (2):263-71.

Samson, Susan, Erica Gonzalez, Vijay Yechoor, Mandeep Bajaj, Kazuhiro Oka, and Lawrence

Chan. 2008. "Gene therapy for diabetes: metabolic effects of helper-dependent

adenoviral exendin 4 expression in a diet-induced obesity mouse model." Molecular

therapy : the journal of the American Society of Gene Therapy no. 16 (649e7b7a-84b1-

d987-fcf9-924e799c3e85):1805-1817. doi: 10.1038/mt.2008.198.

Sauve, M., K. Ban, M. A. Momen, Y. Q. Zhou, R. M. Henkelman, M. Husain, and D. J.

Drucker. 2010. "Genetic deletion or pharmacological inhibition of dipeptidyl peptidase-4

improves cardiovascular outcomes after myocardial infarction in mice." Diabetes no. 59

(4):1063-73. doi: 10.2337/db09-0955.

Schieffer, B., T. Selle, A. Hilfiker, D. Hilfiker-Kleiner, K. Grote, U. J. Tietge, C. Trautwein, M.

Luchtefeld, C. Schmittkamp, S. Heeneman, M. J. Daemen, and H. Drexler. 2004.

"Impact of interleukin-6 on plaque development and morphology in experimental

Page 100: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

89

atherosclerosis." Circulation no. 110 (22):3493-500. doi:

10.1161/01.CIR.0000148135.08582.97.

Schjoldager, B., F. Baldissera, P. Mortensen, J. Holst, and J. Christiansen. 1988.

"Oxyntomodulin: a potential hormone from the distal gut. Pharmacokinetics and effects

on gastric acid and insulin secretion in man." European journal of clinical investigation

no. 18 (5):499-1002.

Schjoldager, B., P. Mortensen, J. Myhre, J. Christiansen, and J. Holst. 1989. "Oxyntomodulin

from distal gut. Role in regulation of gastric and pancreatic functions." Digestive

diseases and sciences no. 34 (9):1411-1420.

Seino, Y., U. Ikeda, M. Ikeda, K. Yamamoto, Y. Misawa, T. Hasegawa, S. Kano, and K.

Shimada. 1994. "Interleukin 6 gene transcripts are expressed in human atherosclerotic

lesions." Cytokine no. 6 (1):87-91.

Seongah, Han, Liang Chien-Ping, DeVries-Seimon Tracie, Ranalletta Mollie, L. Welch Carrie,

Collins-Fletcher Kadesha, Accili Domenico, Tabas Ira, and R. Tall Alan. 2006.

"Macrophage insulin receptor deficiency increases ER stress-induced apoptosis and

necrotic core formation in advanced atherosclerotic lesions." Cell metabolism no. 3

(5f1a2bec-cb50-d227-27a2-21a4b7eff980). doi: 10.1016/j.cmet.2006.02.008.

Shah, Z., T. Kampfrath, J. A. Deiuliis, J. Zhong, C. Pineda, Z. Ying, X. Xu, B. Lu, S. Moffatt-

Bruce, R. Durairaj, Q. Sun, G. Mihai, A. Maiseyeu, and S. Rajagopalan. 2011. "Long-

term dipeptidyl-peptidase 4 inhibition reduces atherosclerosis and inflammation via

effects on monocyte recruitment and chemotaxis." Circulation no. 124 (21):2338-49.

doi: 10.1161/CIRCULATIONAHA.111.041418.

Sierra-Honigmann, M. R., A. K. Nath, C. Murakami, G. Garcia-Cardena, A. Papapetropoulos,

W. C. Sessa, L. A. Madge, J. S. Schechner, M. B. Schwabb, P. J. Polverini, and J. R.

Flores-Riveros. 1998. "Biological action of leptin as an angiogenic factor." Science no.

281 (5383):1683-6.

Singhal, A., I. S. Farooqi, T. J. Cole, S. O'Rahilly, M. Fewtrell, M. Kattenhorn, A. Lucas, and J.

Deanfield. 2002. "Influence of leptin on arterial distensibility: a novel link between

obesity and cardiovascular disease?" Circulation no. 106 (15):1919-24.

Stratton, I. M., A. I. Adler, H. A. Neil, D. R. Matthews, S. E. Manley, C. A. Cull, D. Hadden, R.

C. Turner, and R. R. Holman. 2000. "Association of glycaemia with macrovascular and

microvascular complications of type 2 diabetes (UKPDS 35): prospective observational

study." BMJ no. 321 (7258):405-12.

Ta, Nga, Corinne Schuyler, Yanchun Li, Maria Lopes-Virella, and Yan Huang. 2011. "DPP-4

(CD26) inhibitor alogliptin inhibits atherosclerosis in diabetic apolipoprotein E-deficient

mice." Journal of cardiovascular pharmacology no. 58 (56523d4a-6bbb-f4d4-85a6-

e09af50beea0):157-223. doi: 10.1097/FJC.0b013e31821e5626.

Tangirala, R. K., E. M. Rubin, and W. Palinski. 1995. "Quantitation of atherosclerosis in murine

models: correlation between lesions in the aortic origin and in the entire aorta, and

differences in the extent of lesions between sexes in LDL receptor-deficient and

apolipoprotein E-deficient mice." Journal of lipid research no. 36 (11):2320-8.

Taylor, Addison, and George Bakris. 2010. "The role of vasodilating beta-blockers in patients

with hypertension and the cardiometabolic syndrome." The American journal of

medicine no. 123 (57f03adb-9bfe-1942-e1d0-b592523f5645):6. doi:

10.1016/j.amjmed.2010.04.015.

Tedgui, A., and Z. Mallat. 2006. "Cytokines in atherosclerosis: pathogenic and regulatory

pathways." Physiological reviews no. 86 (2):515-81. doi: 10.1152/physrev.00024.2005.

Page 101: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

90

Tenenbaum, A., E. Z. Fisman, M. Motro, and Y. Adler. 2006. "Atherogenic dyslipidemia in

metabolic syndrome and type 2 diabetes: therapeutic options beyond statins."

Cardiovascular diabetology no. 5:20. doi: 10.1186/1475-2840-5-20.

Terasaki, M., M. Nagashima, T. Watanabe, K. Nohtomi, Y. Mori, A. Miyazaki, and T. Hirano.

2012. "Effects of PKF275-055, a dipeptidyl peptidase-4 inhibitor, on the development of

atherosclerotic lesions in apolipoprotein E-null mice." Metabolism: clinical and

experimental. doi: 10.1016/j.metabol.2011.11.011.

Theodorakis, Michael, Olga Carlson, Spyros Michopoulos, Máire Doyle, Magdalena Juhaszova,

Kalliopi Petraki, and Josephine Egan. 2006. "Human duodenal enteroendocrine cells:

source of both incretin peptides, GLP-1 and GIP." American journal of physiology.

Endocrinology and metabolism no. 290 (3):9. doi: 10.1152/ajpendo.00326.2004.

Thorens, B., A. Porret, L. Buhler, S. P. Deng, P. Morel, and C. Widmann. 1993. "Cloning and

functional expression of the human islet GLP-1 receptor. Demonstration that exendin-4

is an agonist and exendin-(9-39) an antagonist of the receptor." Diabetes no. 42

(11):1678-82.

Tilg, H., E. Trehu, M. B. Atkins, C. A. Dinarello, and J. W. Mier. 1994. "Interleukin-6 (IL-6) as

an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and

soluble tumor necrosis factor receptor p55." Blood no. 83 (1):113-8.

Tilg, Herbert, and Alexander Moschen. 2006. "Adipocytokines: mediators linking adipose

tissue, inflammation and immunity." Nature reviews. Immunology no. 6 (6e04de05-ab1f-

f6c3-a7d6-a806bc0b366e):772-855. doi: 10.1038/nri1937.

Tolhurst, G., F. Reimann, and F. M. Gribble. 2008. "Nutritional regulation of glucagon-like

peptide-1 secretion." The Journal of Physiology no. 587. doi:

10.1113/jphysiol.2008.164012.

Tornehave, D., P. Kristensen, J. Romer, L. B. Knudsen, and R. S. Heller. 2008. "Expression of

the GLP-1 receptor in mouse, rat, and human pancreas." The journal of histochemistry

and cytochemistry : official journal of the Histochemistry Society no. 56 (9):841-51. doi:

10.1369/jhc.2008.951319.

Tremblay, A. J., B. Lamarche, C. F. Deacon, S. J. Weisnagel, and P. Couture. 2011. "Effect of

sitagliptin therapy on postprandial lipoprotein levels in patients with type 2 diabetes."

Diabetes, Obesity and Metabolism no. 13 (4da8bbfd-afd6-3564-976e-bc1c9aa6a8ad).

doi: 10.1111/j.1463-1326.2011.01362.x.

Turton, M. D., D. O'Shea, I. Gunn, S. A. Beak, C. M. Edwards, K. Meeran, S. J. Choi, G. M.

Taylor, M. M. Heath, P. D. Lambert, J. P. Wilding, D. M. Smith, M. A. Ghatei, J.

Herbert, and S. R. Bloom. 1996. "A role for glucagon-like peptide-1 in the central

regulation of feeding." Nature no. 379 (6560):69-72. doi: 10.1038/379069a0.

Unger, R., A. Ohneda, I. Valverde, A. Eisentraut, and J. Exton. 1968. "Characterization of the

responses of circulating glucagon-like immunoreactivity to intraduodenal and

intravenous administration of glucose." The Journal of clinical investigation no. 47

(1):48-113. doi: 10.1172/jci105714.

Ussher, John, and Daniel Drucker. 2012. "Cardiovascular Biology of the Incretin System."

Endocrine reviews (f223284e-4a58-ecab-9cf6-8c3a1d968f22). doi: 10.1210/er.2011-

1052.

Vance, D.E., J.E. Vance, and G. Bernardi. 2002. Biochemistry of Lipids, Lipoproteins and

Membranes. 4th ed. Amsterdam: Elsevier Science B.V.

Veillard, N. R., S. Steffens, G. Pelli, B. Lu, B. R. Kwak, C. Gerard, I. F. Charo, and F. Mach.

2005. "Differential influence of chemokine receptors CCR2 and CXCR3 in development

Page 102: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

91

of atherosclerosis in vivo." Circulation no. 112 (6):870-8. doi:

10.1161/CIRCULATIONAHA.104.520718.

Vella, A., P. Shah, A. S. Reed, A. S. Adkins, R. Basu, and R. A. Rizza. 2002. "Lack of effect of

exendin-4 and glucagon-like peptide-1-(7,36)-amide on insulin action in non-diabetic

humans." Diabetologia no. 45 (10):1410-5. doi: 10.1007/s00125-002-0924-4.

Virmani, Renu, Allen Burke, Andrew Farb, and Frank Kolodgie. 2006. "Pathology of the

vulnerable plaque." Journal of the American College of Cardiology no. 47 (8eb60f4b-

e9e3-d16a-e6e7-9c02c6767478):8. doi: 10.1016/j.jacc.2005.10.065.

Vittone, F., A. Liberman, D. Vasic, R. Ostertag, M. Esser, D. Walcher, A. Ludwig, N. Marx,

and M. Burgmaier. 2012. "Sitagliptin reduces plaque macrophage content and stabilises

arteriosclerotic lesions in Apoe (-/-) mice." Diabetologia no. 55 (8):2267-75. doi:

10.1007/s00125-012-2582-5.

Weber, Christian, and Heidi Noels. 2011. "Atherosclerosis: current pathogenesis and therapeutic

options." Nature medicine no. 17 (11):1410-1432. doi: 10.1038/nm.2538.

Wei, Y., and S. Mojsov. 1995. "Tissue-specific expression of the human receptor for glucagon-

like peptide-I: brain, heart and pancreatic forms have the same deduced amino acid

sequences." FEBS letters no. 358 (9ced5c0c-ca0d-274b-bc42-9164cd5a8078):219-243.

Wellen, Kathryn, and Gökhan Hotamisligil. 2005. "Inflammation, stress, and diabetes." The

Journal of clinical investigation no. 115 (a39445ab-58cf-3fb1-cfda-

a8092f943472):1111-1120. doi: 10.1172/jci25102.

Whiting, David, Leonor Guariguata, Clara Weil, and Jonathan Shaw. 2011. "IDF diabetes atlas:

global estimates of the prevalence of diabetes for 2011 and 2030." Diabetes research

and clinical practice no. 94 (3):311-332. doi: 10.1016/j.diabres.2011.10.029.

Whitman, S. C. 2004. "A practical approach to using mice in atherosclerosis research." The

Clinical biochemist. Reviews / Australian Association of Clinical Biochemists no. 25

(1):81-93.

Wojdemann, M., A. Wettergren, B. Sternby, J. J. Holst, S. Larsen, J. F. Rehfeld, and O. Olsen.

1998. "Inhibition of human gastric lipase secretion by glucagon-like peptide-1."

Digestive diseases and sciences no. 43 (4):799-805.

Xiao, C., R. H. Bandsma, S. Dash, L. Szeto, and G. F. Lewis. 2012. "Exenatide, a glucagon-like

peptide-1 receptor agonist, acutely inhibits intestinal lipoprotein production in healthy

humans." Arteriosclerosis, thrombosis, and vascular biology no. 32 (6):1513-9. doi:

10.1161/ATVBAHA.112.246207.

Xing, Z., J. Gauldie, G. Cox, H. Baumann, M. Jordana, X. F. Lei, and M. K. Achong. 1998. "IL-

6 is an antiinflammatory cytokine required for controlling local or systemic acute

inflammatory responses." The Journal of clinical investigation no. 101 (2):311-20. doi:

10.1172/JCI1368.

Yamagishi, S. I., D. Edelstein, X. L. Du, Y. Kaneda, M. Guzman, and M. Brownlee. 2001.

"Leptin induces mitochondrial superoxide production and monocyte chemoattractant

protein-1 expression in aortic endothelial cells by increasing fatty acid oxidation via

protein kinase A." The Journal of biological chemistry no. 276 (27):25096-100. doi:

10.1074/jbc.M007383200.

Yki-Jarvinen, H. 1990. "Acute and chronic effects of hyperglycaemia on glucose metabolism."

Diabetologia no. 33 (10):579-85.

———. 1997. "Acute and chronic effects of hyperglycaemia on glucose metabolism:

implications for the development of new therapies." Diabetic medicine : a journal of the

British Diabetic Association no. 14 Suppl 3:S32-7. doi: 10.1002/(SICI)1096-

9136(199708)14:3+<S32::AID-DIA442>3.0.CO;2-6.

Page 103: The Role of the Glucagon-Like Peptide-1 Receptor in ......Results: Exendin-4 treatment (10 nmol/kg/day) of high-fat diet-induced glucose-intolerant mice for 22 weeks did not significantly

92

Yvan-Charvet, L., T. A. Pagler, T. A. Seimon, E. Thorp, C. L. Welch, J. L. Witztum, I. Tabas,

and A. R. Tall. 2010. "ABCA1 and ABCG1 protect against oxidative stress-induced

macrophage apoptosis during efferocytosis." Circulation research no. 106 (12):1861-9.

doi: 10.1161/CIRCRESAHA.110.217281.

Yvan-Charvet, L., N. Wang, and A. R. Tall. 2010. "Role of HDL, ABCA1, and ABCG1

transporters in cholesterol efflux and immune responses." Arteriosclerosis, thrombosis,

and vascular biology no. 30 (2):139-43. doi: 10.1161/ATVBAHA.108.179283.

Zanchetti, A., E. A. Rosei, C. Dal Palu, G. Leonetti, B. Magnani, and A. Pessina. 1998. "The

Verapamil in Hypertension and Atherosclerosis Study (VHAS): results of long-term

randomized treatment with either verapamil or chlorthalidone on carotid intima-media

thickness." Journal of hypertension no. 16 (11):1667-76.

Zander, M., S. Madsbad, J. L. Madsen, and J. J. Holst. 2002. "Effect of 6-week course of

glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function

in type 2 diabetes: a parallel-group study." Lancet no. 359 (9309):824-30. doi:

10.1016/S0140-6736(02)07952-7.

Zoungas, S., and A. Patel. 2010. "Cardiovascular outcomes in type 2 diabetes: the impact of

preventative therapies." Annals of the New York Academy of Sciences no. 1212:29-40.

doi: 10.1111/j.1749-6632.2010.05837.x.


Recommended