79
Towards Examining FGF21 Secretion from Pancreatic Islets in a Microfluidic Device by Brenda Green A thesis submitted in conformity with the requirements for the degree of Master of Applied Science Graduate Department of Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Brenda Green (2012)

Towards Examining FGF21 Secretion from Pancreatic Islets ... · Type II diabetes is characterized by impaired insulin-secretion from pancreatic β- cells. A novel endocrine factor,

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Towards Examining FGF21 Secretion from Pancreatic Islets in a Microfluidic Device

    by

    Brenda Green

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

    Graduate Department of

    Institute of Biomaterials and Biomedical Engineering

    University of Toronto

    © Copyright by Brenda Green (2012)

  • ii

    Towards Examining FGF21 Secretion from Pancreatic Islets in a

    Microfluidic Device

    Brenda Green

    Master of Applied Science

    Institute of Biomaterials and Biomedical Engineering University of Toronto

    2012

    Abstract

    Type II diabetes is characterized by impaired insulin-secretion from pancreatic β- cells. A

    novel endocrine factor, fibroblast growth factor 21 (FGF21) is produced by the liver and

    improves pancreatic β- cell function and survival. However, a state of FGF21- resistance has

    been described in obese mice and humans.

    This thesis examines (1) whether pancreatic islets temporally express FGF21 and (2) if

    high glucose and fat reduce transcript expression of FGF21 receptors in islets. This thesis further

    characterizes a novel microfluidic device that will be used to collect and assay FGF21.

    Our data indicate that palmitate induces maximal production of FGF21 mRNA in β- cells

    after 16 hours of stimulation through PPARγ. Secondly, we report that FGF21, glucose and

    palmitate do not cause FGF21-resistance by down-regulating receptor expression. Overall, we

    present a mechanism for the induction of FGF21 from islets, and describe a microfluidic device

    for culture and secretion studies.

  • iii

    Acknowledgments

    I would like to thank my supervisor Professor Rocheleau for his guidance and

    encouragement throughout my Master’s thesis. He has taught me valuable research approaches,

    and thorough experimental techniques. I would also like to thank my committee members

    Professor Volchuk and Professor Sefton for their helpful comments and directions.

    In addition, I want to thank Svetlana Altamentova for all of the animal work, and all the

    Rocheleau lab members for being there to lend a helping hand. I would like to acknowledge the

    Simmons and Guenther labs for their assistance with microfluidic modeling and fabrication.

    Lastly, I want to thank my friends and family for their support and understanding while I

    have been studying. I could not have accomplished this without their encouragement.

  • iv

    Table of Contents

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

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

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

    List of Tables ............................................................................................................................... viii 

    List of Figures ................................................................................................................................ ix 

    List of Appendices .......................................................................................................................... x 

    Chapter 1 - Introduction .................................................................................................................. 1 1.1  Type II Diabetes .................................................................................................................. 1 1.1.1  Glucolipoxicity Model ................................................................................................... 2 1.2  FGFs and FGF21 ................................................................................................................. 4 1.2.1  Fibroblast Growth Factors.............................................................................................. 4 1.2.2  Fibroblast Growth Factor Receptors .............................................................................. 5 1.2.3  Klotho Co-Receptors ...................................................................................................... 7 1.2.4  Paracrine and Endocrine FGFs....................................................................................... 7 1.2.5  FGF Receptor Signaling Pathway .................................................................................. 9 1.2.6  FGF21 Expression and Function .................................................................................. 11 

    Chapter 2 – Related Literature ...................................................................................................... 14 2.1 FGF21 Secretion ................................................................................................................ 14 2.2 FGF21 Resistance .............................................................................................................. 17 

    Chapter 3 – Research Rationale .................................................................................................... 21 3.1 Rationale ............................................................................................................................ 21 3.2 Hypothesis and Objectives ................................................................................................. 22 

    Chapter 4 – Materials and Methods .............................................................................................. 23 4.1 Pancreatic Islet Isolation, Tissue Culture and Cell Culture ............................................... 23 4.2 Fatty Acid Preparation ....................................................................................................... 23 4.3 RNA Isolation, reverse transcription PCR and quantitative PCR ...................................... 23 4.4 Immunoflourescence .......................................................................................................... 24 4.5 Microfluidics ...................................................................................................................... 25 4.5.1 Device Fabrication and Design ....................................................................................... 25 4.5.2 Microfluidic COMSOL modeling ................................................................................... 26 4.5.3 Microfluidic device long- term islet culture .................................................................... 27 4.5.4 Microfluidic device image application ............................................................................ 27 4.6 NAD(P)H Imaging ............................................................................................................. 28 4.7 Spherotech Velocity Profiles and Flou4 Ca2+ Oscillations ................................................ 29 4.8 Statistical Analysis ............................................................................................................. 30 

  • v

    Chapter 5 – FGF21 Secretion and Resistance ............................................................................... 31 5.1 Introduction ........................................................................................................................ 31 5.2 Results ................................................................................................................................ 32 5.2.1 Verifying FGF21 Responsiveness ................................................................................... 32 5.2.2. FGF21 expression is higher in mouse islets compared to liver and WAT ..................... 33 5.2.3 Palmitate, Bezafibrate and Rosiglitazone induce FGF21 expression in Min6 β- cells

    and in islets ....................................................................................................................... 34 5.2.4. Palmitate- induces FGF21 expression in a temporal manner in Min6 β- cells .............. 36 5.3 Discussion .......................................................................................................................... 39 

    Chapter 6 – Microfluidic Device for Islet Culture ........................................................................ 41 6.1 Introduction ........................................................................................................................ 41 6.2 Results ................................................................................................................................ 42 6.2.1 The bypass microfluidic device is designed to hold islets in series while allowing media exchange through the tissue ............................................................................... 42 6.2.3 Pressure drop induces intercellular flow through the islet ............................................ 45 6.2.4 Islets cultured in the bypass microfluidic device have enhanced EC morphology relative to islets cultured in a dish ................................................................................. 48 6.2.5 Islets cultured in the bypass microfluidic device for 48 hours show glucose- stimulated Ca2+ oscillations ........................................................................................... 49 6.3 Discussion .......................................................................................................................... 53 

    Chapter 7 – Conclusion ................................................................................................................. 55 

    Appendix A – FGF21 Circadian Response ................................................................................... 56 A.1 Introduction ....................................................................................................................... 56 A.2 Results ............................................................................................................................... 57 

    Appendix B – Microfluidic Calcium Staining with Fluo4 and β-Cyclodextrin ............................ 59 B.1 Introduction ....................................................................................................................... 59 B.2 Results ............................................................................................................................... 59 

    Appendix C – Metabolic Gene Analysis and Insulin Secretion ................................................... 61 C.1 Introduction ....................................................................................................................... 61 C.2 Results ............................................................................................................................... 62 

    Chapter 8 – References ................................................................................................................. 65 

  • vi

    List of Abbreviations

    Acronyms

    ACC Acetyl-CoA Carboxylase Akt Serine/ threonine protein kinase B ATF6 Activating transcription factor 6 ATP Adenosine Triphosphate BMHH Imaging Buffer BSA Bovine Serum Albumin Ca2+ Intracellular calcium CD31 Cluster of differentiation 31 CDCA Chenodeoxycholic Acid CPT1 Carnitine Palmitoyl Transferase I CT Cycle Threshold for qPCR DAG Diacylglycerol DIO Diet induced obese mice EC Endothelial cell ECM Extracellular matrix ELISA Enzyme-linked Immunosorbent Assay ER Endoplasmic reticulum ERK1/2 Extracellular signal-regulated kinase ERO1 ER oxidoreductin 1 FACoAs Free fatty acid long- chain acyl coenzyme A esters FBS Fetal bovine serum FFA Free Fatty Acids FFT Fast fourier transform FGF21 Fibroblast Growth Factor 21 FGFR Fibroblast Growth Factor Receptor FXR Farnesoid X receptor HEPES 4(2-hydroxyethyl)-1-piperazineethanesulfonic acid HSPG Heparin Sulfate Proteogylcans Idh1 Isocitrate dehydrogenase 1 IgG Immunoglobulin G IRE1α Inositol-requiring 1α JNK JUN N-terminal kinase KATP ATP- sensitive potassium channel KLB Klotho- β MAPK Mitogen-activated Protein Kinase NAD(P)H Aggregate of NADH and NADPH NADH Nicotinamide Adenine Dinucleotide NADPH Nicotinamide Adenine Dinucleotide Phosphate

  • vii

    NF-κB Nuclear transcription factor κB NNT Nicotinamide nucleotide transhydrogenase iNOS Inducible nitric oxide synthase PECAM Platelet endothelial cell adhesion molecule PERK Protein kinase- like ER kinase PC Pyruvate carboxylase PDI Protein disulphide isomerase PDMS Polydimethylsiloxane Pdk4 Pyruvate dehydrogenase lipoamide kinase isozyme 4 PI3K Phosphoinositide 3-kinase PLC Phospholipase C PPAR Peroxisome Proliferator-activated Receptor ROS Reactive oxygen species RPMI Cell and tissue culture media RT-PCR Reverse-Transcription Polymerase Chain Reaction RZD Rosiglitazone Spry Sprouty proteins STAT Signal Transducer and Activator of Transcription TCA Tricarboxylic acid cycle TZD Thiazolidinedione UCP2 Uncoupling protein 2 UPR Unfolded protein response VEGF Vascular endothelial growth factor WAT White Adipose Tissue ZT Zeitgeber time

  • viii

    List of Tables

    Table 1.1 – Fibroblast growth factor subfamilies ………………………...……………..…. 4

    Table 1.2 – Endocrine FGFs and Klotho ……………….……………………………….… 7

    Table 5.1 – In vivo and in vitro concentrations of FGF21 ………………...…………….… 32 Table 6.2.1 – Fluid parameters used to determine the pressure drop across the islet holding area in the direct- flow and bypass microfluidic devices…………..……………… 48 Table 6.2.2 – Shear stress calculated around the periphery of an islet in the direct flow and bypass microfluidic devices ………………………………………..……………………… 51

  • ix

    List of Figures

    Figure 1.1. Pancreatic islet of Langerhan………………………………….……….......................1 Figure 1.2. Fibroblast growth factor subtypes .............................................................................. 5 Figure 1.3. FGFR structure and dimerization ............................................................................... 6 Figure 1.4. Endocrine FGFs ........................................................................................................... 8 Figure 1.5. FGFR signaling network ........................................................................................... 10 Figure 1.6. Anatomic FGF21 qPCR expression profiles in mouse tissues from C57BL6 mice. . 11 Figure 1.7. Relative FGF21 mRNA expression levels in metabolic tissue of C57BL6 lean,  diet induced obese and ob/ob mice ............................................................................................... 12 Figure 1.8. Pancreatic β- cell metabolic pathways ....................................................................... 13 Figure 2.1. Protein levels of FGF21 in the supernatant of liver cells .......................................... 14 Figure 2.2. CDCA bile acids induce hepatic FGF21 secretion from HepG2 cells ...................... 15 Figure 2.3. Model of the PPARγ - FGF21 regulatory pathway in WAT tissue .......................... 16 Figure 2.4. FGF receptor and KLB mRNA expression in WAT of lean and diet- induced obese mice ....................................................................................................................... 17 Figure 2.5. Temporal mRNA expression profile of FGF21 in the liver ...................................... 18 Figure 2.6. Simplified model of a pancreatic islet consisting of clustered pancreatic β- cells…..20 Figure 4.1. Representative microfluidic device used for image analysis ..................................... 28 Figure 4.2. Oscillations and power spectral density plot of glucose- stimulated Ca2+ responses in pancreatic islets ....................................................................................................... 30 Figure. 5.2.1. Mitochondrial NAD(P)H dose response for islets treated with FGF21 ................ 32 Figure. 5.2.2. FGF21 expression is higher in mouse islets compared to liver and WAT ............ 33 Figure. 5.2.3. Palmitate induces FGF21 expression in β- cells, α- cell and islets ....................... 35 Figure. 5.2.4. Palmitate induces FGF21- expression in a temporal manner ................................ 36 Figure. 5.2.5. FGF21, glucose and palmitate treatment do not change FGF21, FGF receptor and co-receptor expression levels. ............................................................................................... 38 Figure 6.2.1. Direct- flow and bypass microfluidic device prototypes ....................................... 46 Figure 6.2.2. Comsol CFD velocity and pressure profiles through the microfluidic devices ...... 44 Figure 6.2.3. Media exchange through islets converges at 30 seconds as the flow rate increases ........................................................................................................................................ 46 Figure 6.2.4. Islets cultured in the short- pass device have enhanced EC morphology relative to islets cultured in a dish ................................................................................................. 49 Figure 6.2.5. Spherotech beads flowing in the short- pass microfluidic device………...............50 Figure 6.2.6. Periods of Ca2+ oscillations in islets cultured for 48 hours in a dish and short-pass device ............................................................................................................................................ 52 Figure A.1.1. The experimental design for examining the FGF21- circadian response .............. 56 Figure A.2.1. Islet mitochondrial and cytoplasmic NAD(P)H responses to glucose ................... 58 Figure B.2.1. β-Cyclodextrin increases the penetration depth of Fluo4 into the islet……..........60 Figure C.2.1. FGF21 does not alter the mRNA expression of UCP2, NNT, PC, Pdk4 or Idh1 in mouse islets………………………………………………………………………………………63 Figure C.2.2. Palmitate and glucose- stimulated insulin secretion……………………………...64

  • x

    List of Appendices

    Appendix A – FGF21- Circadian Response …………………………………….…...….56

    Appendix B – Microfluidic Calcium Staining with Fluo4 and β-Cyclodextrin…..……..59

    Appendix C – Metabolic Gene Analysis and Insulin Secretion…………………………61

  • 1

    Chapter 1 - Introduction

    1.1 Type II Diabetes Diabetes mellitus is a group of metabolic diseases, which are characterized by high blood

    sugar. This is a complex disease that involves the endocrine pancreas, an important organ

    composed of the islets of Langerhans (Figure 1.1). Islets contain β- cells, the cells responsible for

    the production and secretion of the hormone insulin.

    Figure 1.1. Pancreatic islet of Langerhan. Islets are cell aggregates composed of insulin- secreting β- cells, α-, δ- , ε- , PP- cells, endothelial cells, nerves and extracellular matrix. The endocrine cells (α, δ, ε, PP) secrete glucagon, somatostatin, ghrelin and pancreatic polypeptide, respectively. Typically, β- cells consist of 75% of the islet, α- cells 15%, δ- cells 5%, PP cells 4% and ε- cells

  • 2

    We chose to examine fibroblast growth factor 21 (FGF21) due to its therapeutic potential

    for type II diabetes. FGF21 is shown to have positive effects on fatty acid metabolism in liver,

    white adipose tissue and pancreatic islets [5, 14].

    1.1.1 Glucolipoxicity Model

    The clinical state of type II diabetes is often accompanied by elevated blood glucose and

    fatty acid levels, creating a glucolipotoxic environment in the β- cell. Excess carbohydrates and

    fat in the circulation cause hyperinsulinemia and chronic insulin resistance, ultimately resulting

    in β- cell failure [8]. Several pathways have been implicated in glucolipotoxicity including

    endoplasmic reticulum (ER) stress, oxidative stress and the activation of inflammatory pathways

    in pancreatic islets [6, 7]. Prentki et al. (2002) describe a role for malonyl- CoA, peroxisome

    proliferator-activated receptors (PPARα and -γ) in the development of glucolipotoxicity.

    The ER is responsible for protein synthesis, folding and assembly of many proteins, and

    also plays an important role in sensing cellular stress [6]. Chronic glucose and lipid exposure

    increase the demand for protein folding, creating an imbalance between the protein- folding

    capacity of the ER. Unfolded or misfolded proteins accumulate in the ER lumen, creating a

    condition called ER stress. Eukaryotic cells have developed the unfolded- protein response

    (UPR) to cope with stressful conditions and the protein- folding defect. ER sensors inositol-

    requiring 1α (IRE1α), protein kinase- like ER kinase (PERK) and activating transcription factor

    6 (ATF6) sense unfolded proteins and trigger a pathway to inhibit transcription or translation

    processes. If the cell fails to prevent ER stress, the UPR will initiate apoptosis through the CHOP

    transcription factor [6].

    The signaling pathways in the UPR and inflammation are connected to the production of

    reactive oxygen species (ROS) and the activation of nuclear transcription factor (NF-κB) and

    JUN N-terminal kinase (JNK). ROS are small molecules that are highly reactive due to the

    presence of unpaired electrons and the accumulation of these molecules results in a state of

    oxidative stress [6]. Protein folding in the ER consumes energy for the formation of intra- and

    intermolecular disulphide bonds. The electron transport during disulphide bond formation is

    driven by ER proteins: protein disulphide isomerase (PDI) and ER oxidoreductin 1 (ERO1).

    ERO1 creates an oxidizing environment involving electron transfer from PDI to molecular

    oxygen. The transfer of electrons to molecular oxygen results in the production of ROS. The

  • 3

    buildup of ROS is particularly detrimental to the pancreatic islet, due to low levels of intrinsic

    antioxidant defenses. Excess ROS generation may initiate an inflammatory response [6].

    The inflammatory response occurs when cells of the immune system sense cellular

    damage, and trigger the release of inflammatory substances, including cytokines, free radicals

    and hormones. Several inflammatory cytokines (TNF-α, IL-6) cause ER stress and activate the

    UPR [6, 9]. The PERK pathway of the UPR can relieve oxidative stress by triggering the

    induction of oxidant- detoxifying enzymes, including glutathione and NAD(P)H- quinone

    oxidoreductase. Altogether, β- cell glucolipotoxicity triggers the unfolded- protein response in

    the ER which is linked to oxidative stress and inflammation.

    Prentki et al. (2002) describe a glucolipotoxicity model that implicates malonyl- CoA and

    PPARα and-γ. Malonyl-CoA is a metabolic signaling molecule that controls the relative flux of

    fatty acid oxidation and esterification. When nutrient levels are high, glucose is metabolized to

    citrate, which is converted to malonyl-CoA in the cytoplasm. This is the first committed step of

    fatty acid synthesis. Malonyl-CoA also acts to inhibit the transport of fatty acids into the

    mitochondria, thus preventing fatty acid oxidation [8]. The combination of elevated free fatty

    acids (FFAs) and glucose increase the concentration of FFA- derived long- chain acyl-CoA

    esters (FACoAs) in the cytoplasm, which cause lipoapoptosis through nitric oxide cytotoxicity

    [8]. Nitric oxide is known to form complexes in islets that result in an inhibitory effect on the

    tricarboxylic acid cycle (TCA) and ATP production [31].

    PPARs are nuclear receptor transcription factors that regulate diverse aspects of lipid

    metabolism [10]. PPARα stimulates lipid catabolism mainly in the liver by upregulating genes

    involved in fatty acid oxidation [10]. On the other hand, PPARγ causes increased lipid uptake

    and lipogenic processes in adipocytes [11]. Circulating FFAs are natural agonists to PPARs.

    Prentki et al. (2002) show that elevated glucose decreases the expression of PPARα, and induces

    the expression of PPARγ; thus, favoring the lipid synthesis pathway. This leaves only one

    option for FFAs, to undergo esterification, which then causes progressive lipid accumulation and

    β- cell failure [8].

    The lipid partitioning model of β- cell failure in type II diabetes is considered to be

    complementary to the oxidative stress hypothesis. Saturated fatty acids such as palmitate

    increase the production of ROS in rat islets [8]. Both models reveal the biochemical basis of β-

  • 4

    cell adaptation and failure in the progression of type II diabetes. It is therefore critical to study

    the cell mechanisms that regulate β- cell activity during high nutrient loading and develop a

    therapeutic to maintain mass and function.

    1.2 FGFs and FGF21

    1.2.1 Fibroblast Growth Factors

    Fibroblast growth factors (FGFs) are members of the largest family of growth factor

    ligands, comprising 22 members (Table 1.1, Figure 1.2). FGFs are structurally related

    polypeptides that are necessary for embryonic development, proliferation, wound healing, and

    angiogenesis; additionally they can function as hormones that regulate metabolism [12]. Human

    FGFs are 150- 300 amino acid polypeptides with a conserved core of 120 amino acids exhibiting

    30- 60% homology [12].

    Most FGFs function in a paracrine fashion. These FGFs are secreted glycoproteins that

    are generally sequestered to the extracellular matrix (ECM), as well at the cell surface by

    heparan sulphate proteoglylcans (HSPGs). The cell surface HSPGs stabilize FGF ligand-

    receptor interaction, forming a ternary complex with FGF receptors (FGFR) [13].

    Endocrine FGFs, such as FGF21, are of recent interest due to their homeostatic and

    metabolic effects [14]. They are able to act over a distance due to their low affinity for HSPGs;

    however, they require Klotho- β (KLB) as a co-receptor for signaling.

    Table 1.1 – Fibroblast growth factor subfamilies (Figure 1.2)

    Intracrine Intracellular signaling proteins that do not bind FGF receptors and regulate voltage gated sodium channels.

    Paracrine Canonical FGFs that mediate biological responses as extracellular proteins by binding and activating FGFRs with heparan sulphate as a cofactor. Paracrine FGFs act as growth and differentiation factors during development.

    Endocrine Hormone like- FGFs that mediate biological responses in an FGFR- dependent manner. These FGFs bind to heparan sulphate with very low affinity, which enables them to function in an endocrine manner [15].

  • 5

    Figure 1.2. Fibroblast growth factor subtypes. The subtypes consist of intracrine, paracrine and endocrine members. Phylogenetic analysis suggests that FGF genes can be arranged into seven subfamilies, containing two to four members each. Branch lengths are proportional to the evolutionary distance between each gene [12].

    1.2.2 Fibroblast Growth Factor Receptors

    The FGF receptors consist of an extracellular ligand domain composed of two or three

    immunoglobulin- like domains (IgG), a single transmembrane helix domain, and an intracellular

    domain with tyrosine kinase activity (Figure 1.3) [16]. The IgG domain is an essential

    determinant of ligand- binding specificity. FGF receptors signal as dimers, where ligand-

    dependent dimerization leads to a conformational shift in receptor structure that activates the

  • 6

    intracellular kinase domain [13]. The FGF receptors are encoded by four genes (Fgfr-1 to Fgfr-

    4), but because of alternative splicing of Fgfr-1, Fgfr-2 and Fgfr-3, seven prototype receptors are

    generated (1b, 1c, 2b, 2c, 3b, 3c and 4) [16]. The specificity of the FGF-FGFR interaction is

    established by the different ligand-binding capacities of the receptor isoforms and tissue- specific

    expression of ligands, receptors and other transmembrane proteins (such as KLB) [13].

    Figure 1.3. FGFR structure and dimerization. (A) FGF binds and is sequestered to the extracellular matrix. (B) FGF signal transduction is initiated upon binding of the FGF ligand in conjunction with heparan sulphate to form a ternary complex. The result is trans- autophosphorylation and activation of the tyrosine kinase, which facilitates second messenger signaling. (C) FGF receptor complex consists of extracellular Ig domains, single transmembrane domain, and a cytoplasmic tyrosine kinase domain [16].

    A B C

  • 7

    1.2.3 Klotho Co-Receptors

    The Klotho gene encodes a single-pass transmembrane protein. Klotho forms complexes

    with FGFRs and increases their affinity to endocrine FGFs. These receptor complex formations

    are required to induce phosphorylation and signal transduction. The expression of Klotho

    proteins and FGFR splice variants determines the specificity of endocrine FGF signaling [15]. It

    has been shown that FGF21 binds Klotho- β (KLB) and FGFR1c and 2c in liver, white adipose

    tissue (WAT) and pancreas. KLB has a relatively large extracellular domain with 11 putative N-

    glycosylation sites, two inactive glycosyl hydrolase-1 domains, and a relatively short C-terminal

    tail [17].

    1.2.4 Paracrine and Endocrine FGFs

    Paracrine FGFs function in development, cell proliferation and migration by influencing

    the intracellular signaling events of neighboring cells. The range of FGF signaling is regulated in

    part by its affinity for the extracellular matrix and in part by dimerization of some FGFs [12].

    Examples of paracrine FGFs include FGF4 and FGF8, which have essential roles in blastocyst

    formation and gastrulation.

    Endocrine FGFs function through FGFRs and act over long distances as hormone- like

    factors (Table 1.2). These hormones have been recently appreciated as endocrine factors (in the

    past 5 years), in contrast to paracrine FGFs, which have been a focus of research over the last 40

    years [18]. There is a growing interest in understanding the effects of endocrine FGFs.

    Table 1.2 – Endocrine FGFs and Klotho (Figure 1.4)

    FGF15/ 19 FGF15/19 are orthologous genes in vertebrates where FGF15 is found in mice and FGF19 is found in humans. FGF15/19 is secreted from the ileum upon feeding. These FGFs act on hepatocytes to reduce bile acid synthesis through suppression of transcription factors. They are also shown to be involved in cell proliferation. Binds Klotho- β and FGFR4 found in the liver and gallbladder.

    FGF21 FGF21 is secreted by the liver and adipocytes and is shown to regulate energy homeostasis (glucose and lipid metabolism). Binds Klotho- β and FGFR1c and 2c in liver, WAT and pancreas.

    FGF23 FGF23 is secreted by bone, and is involved in phosphate and vitamin D homeostasis in the kidney. Binds Klotho- α and FGFR1c in the kidney [15].

  • 8

    Figure 1.4. Endocrine FGFs. FGF signaling is regulated by the expression of Klotho co- receptors. FGF15/19 is secreted from ileum upon feeding and acts on the liver and gallbladder via Klotho- β and FGFR4. FGF21 is secreted from the liver upon fasting and acts on WAT through Klotho- β and FGFR1c/2c. FGF23 is secreted from the bone in response to vitamin D and acts on the kidney though Klotho- α and FGFR1c [15].

    Ileum  Liver Bone 

    FGF15/19  FGF21 FGF23

    FGFR4 βKlotho  βKlotho αKlotho FGFR1cor 2c

    FGFR1c

    Bile acid homeostasis Bile acid synthesis (Liver) ↓ Bile acid transport (Ileum) ↓ 

    Energy homeostasis Glucose uptake (WAT) ↑

    Phosphate & Vitamin D homeostasis 

    Phosphate excretion (Kidney) ↑Vitamin D synthesis (Kidney) ↓

  • 9

    1.2.5 FGF Receptor Signaling Pathway

    The FGF- FGFR ternary complex formation results in trans- autophosphorylation of the

    receptor tyrosine kinase domains. The phosphorylated tyrosine residues function as docking sites

    for adaptor proteins that may also be directly phosphorylated by FGFR, leading to the activation

    of multiple signaling pathways (Figure 1.5) [13].

    FGF binding to its receptor and co-receptor results in the activation of four key downstream

    signaling pathways: (1) RAS-RAF-MAPK, (2) PI3K-AKT, (3) STAT and (4) PLCγ/ Ca2+ [12].

    There are several mechanisms that attenuate FGFR signaling which are only partly understood

    [13]. Sprouty (Spry) proteins, SEF family members and Stat transcription factors can modulate

    receptor signaling at several points.

    Spry proteins are thought to function in a dominant negative manner, by competing for

    adaptor protein GRB2 binding, and preventing SOS-mediated RAS activation, or by directly

    binding to RAF and blocking subsequent MAPK signaling [13].

    SEF may function at multiple levels including a transmembrane form that can directly

    interact with FGFRs and an intracellular form capable of inhibiting ERK1/2 phosphorylation.

    STAT signaling transducer can translocate to the nucleus and affect transcription of target

    genes (such as growth inhibitor, p21).

  • 10

    Figure 1.5. FGFR signaling network. The signal transduction network downstream of fibroblast growth factor receptors (FGFRs), along with negative regulators. Following ligand binding and receptor dimerization, the kinase domains transphosphorylate each other, leading to the docking of adaptor proteins and the activation of four key downstream pathways:

    (1) RAS–RAF–MAPK [cell growth, differentiation, survival] (2) PI3K–AKT [proliferation, apoptosis] (3) Signal transducer and activator of transcription (STAT) [transcription activity] (4) Phospholipase Cγ (PLCγ) [intracellular calcium concentrations]

    DAG, diacylglycerol; FRS2α, FGFR substrate 2α; GRB2, growth factor receptor-bound 2; IP3, inositol triphosphate; P, phosphorylation; PIP2, phosphatidylinositol-4,5-biphosphate; PKC, protein kinase C; Sos, son of sevenless [13].

    FGF signaling pathways

  • 11

    1.2.6 FGF21 Expression and Function

    Fibroblast growth factor 21 is an endocrine polypeptide that is a promising clinical

    candidate for the treatment of type II diabetes [19, 20, 21]. FGF21 is a significant hormonal

    regulator of glucose, lipids and energy balance. Endogenous FGF21 facilitates insulin-

    independent uptake of glucose in adipocytes and promotes lipolysis [22, 23]. Our lab has also

    demonstrated FGF21- responsiveness in pancreatic islets [5]. The relative mRNA tissue

    expression in C57BL6 mice is presented in Figure 1.6 [24]. FGF21 is present is most tissues,

    with elevated levels in the pancreas, testis, and liver. This thesis specifically explores FGF21

    expression in the pancreatic islet α- and β- cells.

    Figure 1.6. Anatomic FGF21 qPCR expression profiles in mouse tissues from C57BL6 mice. mRNA expressed relative to 18S rRNA [24].

    The mRNA expression of FGF21 is examined in diet- induced obese (DIO) and ob/ob

    mice. DIO mice are fed a high- fat diet for 8- 12 weeks. They become obese, moderately

    hyperglycemic and develop impaired glucose tolerance. Typical serum glucose concentrations

    and free fatty acids levels are ~9 mM and 0.5 mM, respectively [25]. Ob/ob mice are a mutant

    strain that lack the hormone leptin, which controls appetite. These mice develop high blood sugar

    (15- 20 mM) and increased levels of insulin. Both models are used in studies of type II diabetes.

    Relative

     mRN

    A expression 

       FGF21 

    Eye

    Bra

    in S

    tem

    Cer

    ebel

    lum

    Cer

    ebru

    mC

    orpu

    s St

    riatu

    mO

    lfact

    ory

    Bul

    bSp

    inal

    Cor

    dH

    ypot

    hala

    mus

    Pitu

    itary

    Adr

    enal

    Thyr

    oid

    Panc

    reas

    Tong

    ueSt

    omac

    hD

    uode

    num

    Jeju

    num

    Ileum

    Col

    onG

    all B

    ladd

    e rLi

    ver

    Kid

    ney

    BA

    TW

    AT

    Sple

    enTh

    ymus

    Ova

    ryU

    teru

    sEp

    idid

    ymus

    Prep

    utia

    l Gla

    ndPr

    osta

    teSe

    min

    al V

    esic

    les

    Test

    isVa

    s D

    efer

    ens

    Aor

    taH

    eart

    Lung

    Bon

    eM

    uscl

    eSk

    in

  • 12

    FGF21 expression levels are significantly induced in the liver of ob/ob mice compared to

    adipose and pancreatic tissue (Figure 1.7) [26].

    Figure 1.7. Relative FGF21 mRNA expression levels in metabolic tissue of C57BL6 lean, diet induced obese and ob/ob mice. Tissues were collected from ad libitum fed lean (open bars, 8 and 16 wk old), DIO (gray bars, 16 wk old), or ob/ob mice (black bars, 8 wk old). Data were normalized to the cyclophilin control gene and are expressed as fold change in FGF21 mRNA levels relative to the level seen in the liver of lean mice [26].

    The expression level of FGF21 in the pancreas of lean mice is affected by the diet. This

    led us to consider FGF21 signaling in pancreatic islets treated with excess nutrients. In islets,

    FGF21 causes a reduction in acetyl CoA carboxylase (ACC) levels, which we postulate leads to

    improved fatty acid transport and oxidation [5]. As shown in Figure 1.8, high glucose and fat

    cause a build up of citrate which is generated from the tri-carboxylic acid cycle (TCA). Citrate is

    converted to malonyl CoA, in the presence of ACC, for fatty acid synthesis. Malonyl CoA

    inhibits fatty acid transport into the mitochondria by blocking carnitine palmityl transferase

    (CPT1), thereby blocking mitochondrial fatty acid oxidation [27]. Overall, ACC regulates the

    levels of malonyl CoA, to ultimately regulate the levels of fatty acid synthesis and fatty acid

    oxidation in the cell.

    Relative

     mRN

    A expression 

  • 13

    The TCA cycle produces NADH as a key energy intermediate. NADH is ultimately

    converted to cellular energy in the form of ATP. NADPH is consumed during fatty acid

    synthesis. The aggregate signal of NADH and NADPH (NAD(P)H) is a readout of the metabolic

    state of the cell.

    Figure 1.8. Pancreatic β- cell metabolic pathways

    FGF21 improves pancreatic β- cell function and survival by activation of ERK1/2 and

    Akt signaling pathways [20]. Wente et al. (2006) showed that in vitro FGF21 treatment of islets

    isolated from healthy rats enhanced insulin mRNA and protein levels compared to the control.

    FGF21 treatment also caused rapid phosphorylation of ERK1/2 and Akt. It has been previously

    demonstrated that FGF21 is mitogenically inactive in vitro when tested with FGF- sensitive cell

    lines [20]. Thus, FGF21 promotes β- cell survival and function, without inducing mitogenicity.

    Fatty AcylCoA 

    Pyruvate 

    Malonyl CoA 

    Citrate Palmitate

    ACC

    Acetyl CoA

    Citrate

    Glucose Metabolism

    Fatty Acid Oxidation

    Fatty Acid Synthesis 

    Cytoplasm  Mitochondria

    TCA Cycle 

    ↑NADH

    ↑ NADH 

    ↑ NADH

    ↓ NADPH 

    CPT1 

  • 14

    Chapter 2 – Related Literature

    2.1 FGF21 Secretion

    FGF21 is known to be mainly secreted from the liver via PPARα and acts as an endocrine

    factor [28]. Free fatty acids (FFA) induce the expression of PPARα, resulting in the downstream

    production of FGF21 (Figure 2.1) [29]. This thesis explores PPAR- induced FGF21 transcript

    expression in islets and β- cells.

    Figure 2.1. Protein levels of FGF21 in the supernatant of liver cells. Human liver hepatocellular carcinoma cells (HepG2) are stimulated with an 0.16 mM FFA mixture (palmitic, oleic and linoleic acid) for 1, 2, 4, 8 and 24 h. Results are expressed as means (± sem) *P

  • 15

    Figure 2.2. CDCA bile acids induce hepatic FGF21 secretion from HepG2 cells. Cells were incubated with 100 μM CDCA for 6 or 12 hours. The level of FGF21 and albumin in the culture medium was measured by Western analysis [30]

    FGF21 induction in adipocytes occurs via PPARγ agonists during the fed-state. PPARγ

    induction has beneficial effects for lipotoxic human islets, through antioxidant properties and

    reduction of iNOS [31]. Dutchak et al. (2012) decided to investigate the mechanisms of FGF21

    secretion from adipocytes.

    PPARγ agonists include thiazolidinediones (TZDs): rosiglitazone and pioglitazone.

    PPARγ activity is regulated by posttranslational modification, including phosphorylation and

    sumoylation. Small Ubiquitin-like Modifier or SUMO proteins are a family of small proteins that

    are covalently attached to and detached from other proteins in cells to modify their function.

    Sumoylation of PPARγ at K107 blocks its transcriptional activity. Dutchak et al. (2012) report

    that FGF21 inhibits the sumoylation of PPARγ; thus, sustaining its own activity in a feed-

    forward manner (Figure 2.3). PPARγ - induced FGF21 enhances adipocyte differentiation,

    insulin sensitization, promotes proper lipid distribution and stimulates PPARγ transcriptional

    activity.

    Relative

     FGF21 

    Protein Secretion 

    6 h            12 h 

    Human HepG2 

    FGF21 

    Albumin 

  • 16

    Figure 2.3. Model of the PPARγ - FGF21 regulatory pathway in WAT tissue [32].

    FGF21 mRNA and protein levels were induced by rosiglitazone in WAT but not in liver

    of wild type mice. Conversely, FGF21 mRNA and protein levels were increased by the PPARα

    agonist in the liver, but not WAT. The PPARα agonist, GW7647, significantly increased plasma

    FGF21 concentrations; however, rosiglitazone had no effect [32]. This suggests that FGF21 is

    secreted from WAT but acts locally in an autocrine or paracrine fashion instead of entering the

    circulation. Kharitonenkov et al. (2011) describe FGF21 as an endocrine factor secreted from the

    liver; however, Dutchak et al. (2012) hypothesize that FGF21 is secreted from WAT but is

    unable to enter the circulation due to the interactions with the WAT extracellular matrix. Thus,

    FGF21 was postulated to act as an autocrine or paracrine factor, similar to conventional FGFs.

    Johnson et al. (2009) also describes an autocrine role for FGF21 in exocrine pancreas and liver.

    Acinar cell FGF21 mRNA expression markedly increased during pancreatitis in vitro. The

    activation of FGF21 was an immediate cell autonomous result to pancreatic damage, leading to

    an autocrine signal that reduced the expression of early response factor, Erg1 [64].

    This led us to hypothesize that pancreatic islets express and secrete FGF21 to signal in an

    autocrine manner. We further hypothesize that this signaling pathway may be diminished during

    diabetes due to FGF21 resistance.

    TZDs

    Insulin sensitization, side effects 

     FGF21

    SUMO‐PPARγ (inactive) 

    PPARγ(active)

  • 17

    2.2 FGF21 Resistance

    FGF21 improves β- cell function and survival by stimulating peripheral glucose and lipid

    disposal [33]. However, a state of FGF21 resistance has been described in obese mouse models

    and in obese humans [21, 34].

    Fisher et al. report that the expected beneficial effects of endogenous FGF21 to increase

    glucose tolerance and reduce circulating triglycerides are absent in obesity. Expression of

    FGFR1 and KLB were significantly downregulated in WAT of obese mice (Figure 2.4), most

    likely contributing to impaired FGF21 signaling [21].

    Figure 2.4. FGF receptor and KLB mRNA expression in WAT of lean and diet- induced obese mice. Data for each gene are calculated using 36B4 as a reference gene [21].

    FGF21 is secreted from the liver via PPARα in circadian fashion [28]. Figure 2.5

    illustrates the FGF21 circadian mRNA levels for mice fed with a fasting diet containing PPARα

    agonist, compared to mice fed with a high-fat ketogenic diet. Mice were maintained under light

    cycle of 12 hrs lights on: 12 hrs lights off (lights on at ZT0) and fed ad libitum 5 days. The fold

    change in FGF21 mRNA level is blunted in mice fed a high-fat diet. The peak to baseline ratio of

    the mice fed a high-fat diet is reduced compared to the fasting mice. The baseline FGF21 levels

    are also increased significantly in the high-fat case.

       FGFr1    FGFr2    FGFr3    FGFr4       KLB

    FGFr/36B

    4 mRN

  • 18

    Figure 2.5. Temporal mRNA expression profile of FGF21 in the liver. Mice are fed a fasting diet (left) and high-fat ketogenic diet (right). Open circles represent control mice, and closed circles represent treated mice. The maximal value for control mice is expressed as 100% [10, 70].

    Yu et al. (2011) recently published results showing circadian rhythm of FGF21 in 36 lean

    and obese volunteers. FGF21 levels increase during periods of fasting (overnight) and are

    reduced during feeding (daytime). The magnitude of the nocturnal rise was blunted in obese

    individuals (peak to baseline ratios were 2 vs. 4 in lean individuals). The baseline FGF21

    concentrations were much higher in obese than lean people.

    Coskun et al. (2008) detected a prominent effect of exogenous FGF21 treatment on

    endogenous FGF21 mRNA expression in liver, adipose and pancreatic tissue of DIO mice.

    FGF21 transcript levels were substantially lowered in each of these tissues after FGF21

    administration. These studies demonstrate that FGF21 and FGFR expression levels, and

    circadian rhythm of FGF21 are diminished during obesity suggesting an FGF21- resistant state.

    Fisher et al. (2010) measured ERK1/2 phosphorylation as a reporter of FGF signaling. In

    obese mice, they found that there was a severe impairment in the ability of FGF21 to activate

    signaling pathways through induction of phospho- ERK1/2 in liver and WAT. Therefore, we

    hypothesized that FGF21- resistance may occur in pancreatic islets and interfere with FGF21

    expression, secretion and autocrine signaling. Despite the evidence for FGF21- resistance, Hale

    et al. (2012) report that the phospho- ERK1/2 response was only partly attenuated in liver and

    WAT of obese mice. This area remains controversial and further studies are required to elucidate

    FGF21 resistance.

    0 6 12 18 24 0

    2000

    4000

    6000 mRN

    A level (%) 

    mRN

    A level (%) 

  • 19

    2.3 Microfluidic Devices for Pancreatic Tissue Culture

    Microfluidic devices are systems that process small (10-4 to 10-3 liters) amounts of fluids,

    using channels with dimensions of tens to hundreds of micrometers [35]. These low cost systems

    offer a variety of useful features including the ability to use very small quantities of reagents,

    short time for analysis and small footprints for the analytical devices. The effects that become

    dominant in microfluidics include laminar flow, diffusion, fluidic resistance and surface area to

    volume ratio [36]. The laminar flow through microfluidics is a result of low Reynolds number by

    design (< 2,300) and enables the controlled application of shear stress and the delivery of

    multiple laminar streams in the absence of mixing. When two fluids come together in a

    microchannel, they do not mix convectively; instead they flow in parallel, without eddies or

    turbulence. The only mixing that occurs is the result of the diffusion of molecules across the

    interface of the fluids [35]. We have also shown that microfluidic devices used for islet culture

    and analysis provide benefits such as enhanced nutrient delivery to the center of the tissue and

    control of flow conditions. Microfluidic devices are also used for periodic effluent collection

    (such as insulin) from the cultured tissue [37]. Due to small volumes, the effluent becomes

    concentrated in the exit stream.

    The devices are commonly made of polydimethylsiloxane (PDMS), an organic silicon

    based compound. PDMS is chosen for tissue culture due to its inert, non-toxic, and optically

    clear properties. PDMS is also inherently permeable to gas, and therefore fully compatible with

    living samples.

    Johansson et al. (2006) showed that β- cell proliferation is closely associated with

    vascular growth. Pancreatic islets are cell aggregates that largely consist of endocrine cells and

    vascular endothelial cells (ECs) [38]. β- cells form the core of the islet however every β- cell is

    adjacent to several β- cells as well as capillary ECs (Figure 2.6).

  • 20

    Figure 2.6. Simplified model of a pancreatic islet consisting of clustered pancreatic β- cells (green) and endothlial cells (red). Glucose enters the capillary blood vessels, and diffuses into the cells of the islet. Islet β- cells secrete insulin in response to glucose, where it travels through the blood vessels to the rest of the body [39].

    Sankar et al. (2011) examined EC morphology in pancreatic islets cultured in a

    microfluidic flow environment. Islets cultured in this device had preserved EC- density and -

    connected length throughout 24 and 48 hours in culture. The devices were designed to mimic in

    vivo hemodynamics using flow rates between 3 and 6 ml/24 hours. Microfluidic tissue culture

    presents several advantages over conventional culture in a dish, such as improved EC-

    morphology and enhanced nutrient delivery to the center of the islet.

    β- cells respond to glucose through closure of the ATP- sensitive potassium channel

    (KATP), membrane depolarization, Ca2+ influx through voltage gated calcium channels, and

    subsequent insulin secretion [40]. Sankar et al. (2011) observed a mutated glucose- stimulated

    Ca2+ response with the Flou4 AM dye in device- cultured islets. The peripheral β- cells of the

    islet experienced flow- induced damage due to shear stress, preventing the detection of Ca2+

    oscillations. We aim to design a microfluidic device for islet culture that reduces shear stress

    around the islet, allowing detection of Ca2+ oscillations, while maintaining enhanced delivery of

    media to the centre of the tissue and EC morphology. Furthermore, the device will be custom-

    designed for islet secretion studies.

          Glucose 

          Glucose 

    Insulin

    Insulin 

    Blood Vessel 

    Pancreatic Islet

  • 21

    Chapter 3 – Research Rationale

    3.1 Rationale

    The glucolipotoxicity model of type II diabetes describes that β-cell dysfunction develops

    when both glucose and circulating fatty acids are high. Elevated glucose and fatty acids cause

    toxicity in islets, which may be associated with type II diabetes. The mechanism leading to β-cell

    glucolipotoxicity is described by oxidative stress and lipid partitioning models [7, 8]. We aim to

    determine the regulation of FGF21- signaling in β- cells during glucolipotoxicity.

    FGF21 is produced by the liver through induction of PPARα. It is secreted into the

    circulation by hepatocytes, due to its low affinity for heparin sulfate proteoglycans in the

    extracellular matrix, where it stimulates glucose- uptake and promotes lipolysis [13]. FGF21 is

    induced in WAT via PPARγ during the fed state and acts in a feed-forward autocrine loop to

    sustain its own activity [32]. Johnson et al. (2009) show that pancreatic acinar cell FGF21

    expression immediately increases in response to cerulein- induced pancreatitis in mice. Thus, it

    is possible that pancreatic islets respond to glucolipotoxicity by generating FGF21. This action

    could preserve β- cell survival and function in an autocrine manner.

    Excess circulating nutrients appear to diminish the beneficial effects of FGF21, leading to

    reduced circadian rhythms, phospho- ERK1/2 levels and FGF21 and FGF receptor expression

    levels [21, 34]. Given that FGF resistance could affect mRNA expression and secretion

    pathways, we examined whether excess nutrients alter FGF21 and FGF receptor expression

    levels in pancreatic islets.

    Pancreatic islets secrete low quantities of hormones (5.5 - 15 ng/ml of insulin collected in

    a 500 μl volume over 40 minutes). Our goal was to use a microfluidic device for the long- term

    culture and assay of FGF21 secretion from pancreatic islets. Previous devices presented several

    problems, such as non- uniform flow around islets, challenges identifying individual islets and

    exposing the islets to high shear stress [37]. We designed a device that provided enhanced flow

    through the islets and reduced shear stress around the islet periphery.

  • 22

    3.2 Hypothesis and Objectives

    FGF21 is a metabolic hormone that is controlled by the nutritional state and influences

    glucose and lipid metabolism. In WAT, FGF21 is an inducible, fed- state autocrine factor that

    functions in a feed- forward loop [32]. Despite the beneficial effects of FGF21, Fisher et al.

    (2010) and Yu et al. (2011) describe a state of FGF21 resistance in obese mice and humans.

    1. We hypothesized that fatty acids induce FGF21 transcript expression (mRNA) in β- cells

    in a temporal manner.

    Objectives

    1. Determine if palmitate, oleate, linoleate, PPARα agonist (bezafibrate) or PPARγ

    agonist (rosiglitazone) induce the expression of FGF21 in β- cells and islets

    2. Compare FGF21 expression in mouse islets, liver, WAT, β- cells and α- cells

    3. Examine the temporal induction of FGF21 in β- cells

    4. Design a microfluidic device for pancreatic islet culture and effluent collection

    2. We hypothesized that high glucose and high fat cause FGF21 resistance in pancreatic

    islets by down-regulating the mRNA expression of FGF21, FGFR1c and KLB.

    Objectives

    1. Determine if elevated levels of FGF21, glucose or palmitate reduce the expression

    levels of FGF21, FGF receptor and co-receptor in islets

  • 23

    Chapter 4 – Materials and Methods

    4.1 Pancreatic Islet Isolation, Tissue Culture and Cell Culture

    Ex vivo culturing of pancreatic mouse islets were performed for quantitative PCR

    (qPCR), ELISA and microfluidic analysis. Animal procedures were approved by the Animal

    Care Committee of the University Health Network, Toronto, Ontario, Canada in accordance with

    the policies and guidelines of the Canadian Council on Animal Care (Animal Use Protocol

    #1531). The C57BL6 mice strain is a common inbred strain widely used for models of human

    disease due to its availability and robustness. Pancreatic islets were isolated from 8- to 12- week-

    old C57BL6 male mice by using collagenase digestion (Roche) [41]. Islets were subsequently

    cultured in the islet media (full RPMI 1640 medium supplemented with 11 mM glucose, 10%

    FBS, and 5 U/ml penicillin-streptomycin).

    Mouse insulinoma 6 (Min6) β- cells are a cell- line commonly used as a model for

    pancreatic β- cells [42]. The next most abundant cell type in the islet is the glucagon- secreting

    α- cells. The adenoma- derived α- cell line αTC1 is used for diabetes research due to its glucagon

    biosynthesis and cytokine sensitivity [43]. Min6 β- cells and αTC1 cells were cultured in RPMI

    1640 media supplemented with 5 mM glucose, 1% FBS and 5 U/ml penicillin-streptomycin.

    Cells are treated with 100 ng/ml of recombinant human FGF21 (R&D Systems).

    4.2 Fatty Acid Preparation

    Palmitate is prepared by initially dissolving 0.128 g of palmitic acid (Sigma Aldrich) in

    0.1 M NaOH at 70oC to make 100 mM solution. Next, the solution was diluted to 2 mM in 2%

    fatty acid free BSA (BioShop), and incubated for 15 minutes at 60oC. Finally, the 2 mM solution

    was diluted to 0.2 mM in cell culture or islet media. Oleate and linoleate were purchased as

    BSA- conjugated stock solutions (Sigma Aldrich) and added directly to the cell culture media.

    4.3 RNA Isolation, reverse transcription PCR and quantitative PCR

    RNA isolation, reverse transcription (RT) PCR and quantitative (q) PCR were performed

    for mouse islets, liver, WAT and α- and β- cell lines. Islets were hand- picked from RPMI 1640

    culture media into an Eppendorf tube where they were centrifuged and washed with 1x PBS. A

    small piece of liver and WAT was excised from the mouse, minced and homogenized. Cells

  • 24

    were grown to 70- 80% confluency and then washed with 1x PBS. The tissues and cells were

    lysed and the nucleic acid was extracted using TRIzol reagent and chloroform. RNA was

    extracted using the Qiagen RNA extraction kit, and the quantity and quality were determined by

    measuring the absorbance at 260 and 280 nm. Reverse transcription to create the cDNA template

    was carried out using a PCR thermal cycler (10 minutes at 25oC, 2 hours at 37oC followed by 5

    minutes at 85oC). The forward and reverse primers used for PCR amplification of mouse FGF21

    are 5’-AGATCAGGGAGGATGGAACA-3’ and 5’-TCAAAGTGAGGCGATCCATA-3’. PCR

    amplification was performed with Taq DNA polymerase (Sigma Aldrich) with the following

    reaction steps: 95oC for 1 minute, 95oC for 30 seconds, 51oC for 30 seconds, 72oC for 30

    seconds, 34 cycles, 72oC for 10 minutes. The cDNA was run on a 1.7% agarose gel.

    Quantitative PCR was set up in duplicate with primers specific to FGF21, FGFR1c,

    FGFR2c and KLB (Applied Biosystems: FGF21 Mm00840165_g1, FGFR1c Mm00438930_m1,

    FGFR2c Mm01269938_m1, KLB Mm00473122_m1). The mRNA expression level was

    determined relative to actin (Applied Biosystems 4352933E) by measuring the cycle threshold

    (CT) value. The CT represents the cycle at which there is the first detectable increase in cDNA

    fluorescence. Reactions with a CT greater than 38 cycles were determined to be below the limit

    of detection. CT values typically varied between 33-36 cycles required for DNA amplification

    (out of a total of 40 cycles).

    4.4 Immunoflourescence

    Islets were stained with PECAM-1 (platelet endothelial cell adhesion molecule 1)

    antibodies after 48- hour culture in a dish and in the short-pass device. Islets cultured in the dish

    were loaded into a device for PECAM-1 staining. Islets were fixed with 2% paraformaldehyde

    for 1 hour at 100 μl/h in the device and then blocked with 0.1% Triton X, 10% normal goat

    serum in 1x PBS on ice for 4 hours at 25 μl/h. Islets were stained with primary antibody (1:150

    dilution of rat anti- mouse CD31, BD Pharmigen) for at least 12 hours at 2 μl/h and then blocked

    for 1 hour at 100 μl/h. Islets were stained with secondary antibody (1:500 dilution of Alexa Fluor

    633 goat anti-rat IgG, Invitrogen) for 3 hours at 25 μl/h and then rinsed with 1x PBS for ½ hour

    at 100 μl/h. Islets were then imaged using confocal microscopy using 633 nm excitation. Islet

    endothelial cell (EC) length was analyzed using the Simple Neurite tracer macro for Fiji. The

    total EC connected length was normalized to the islet periphery and plotted as EC length/

  • 25

    perimeter. Islet EC fractional area was detected by adjusting the threshold for 5- 10 z-slices

    through each islet and measuring the average fractional intensity.

    4.5 Microfluidics

    4.5.1 Device Fabrication and Design

    Microfluidic channels were designed using Adobe Illustrator CS4. Channels were drawn

    to actual size using Adobe Illustrator in white color, and the background was kept in black color.

    Designs were printed to acetate film. Device masters were fabricated at the University of

    Toronto Microfluidics Foundry. Corning glass slides were used as the master substrate. The

    slides were washed sequentially using isopropanol, acetone, and isopropanol and then

    immediately dried using an air gun. The cleaned glass slides were placed on a 65oC hot plate for

    15 minutes to dry. SU8 photo-resist was used to coat the glass substrate slides. The 125 μm layer

    was spun using SU8-2100 at settings, step 1 at 500 rpm, ramp 5 seconds, dwell 10 seconds, and

    step 2 at 2,500 rpm, ramp 5 seconds, dwell 30 seconds. The subsequent SU8-2100 coat was soft

    baked at 65oC for 5 minutes, 95oC for 30 minutes, 65oC for 5 minutes, and cooled at room

    temperature for 10 minutes. The 125 μm features were added by applying the corresponding

    master film negative, with channel features exposed to UV for 16 seconds with 13 mW of power.

    The post-UV exposure bake was done at 65oC for 5 minutes, 95oC for 12 minutes, 65oC for 5

    minutes, and cooled at room temperature for 30 minutes.

    The UV exposed channel features of the SU8 coating were thermally cross linked, and

    rendered insoluble to the MicroChem SU8 liquid developers. The masters were finally developed

    by soaking in the SU8 developer solution in glass trays agitated on an orbital shaker for 15

    minutes. The masters were left to dry at room temperature overnight.

    Microfluidic devices were fabricated using elastomer polydimethylsiloxane (PDMS)

    (Dow Corning Sylgard) [44]. The PDMS silicone elastomer base and the curing reagent were

    poured into P100 dishes at a 10:1 ratio (35 g to 3.5 g), mixed in a fume hood, and vacuum

    desiccated for removal of bubbles. The mixtures were subsequently poured over top of the

    master slides containing positive moulds of channels. The dishes were desiccated again to

    remove bubbles and baked at 80oC for 3 hours in a vacuum oven for curing. The PDMS devices

    were peeled off from the master moulds gently, and cut into appropriate sizes for bonding onto

  • 26

    No. 1 thickness 24 × 50-mm coverslips (VWR Scientific). Prior to bonding, cover slips were

    cleaned using methanol, and dried using β wipes. PDMS devices were first hole-punched at the

    ends of channels. The hole-punched devices were placed channel side up on a glass slide, and

    clear tape was used to clean the channel side. The cleaned PDMS device and dried cover slip

    were simultaneously oxygen plasma treated (Harrick Plasma Cleaner) at high power for 1

    minute. The irreversible bonding was performed immediately following oxygen plasma

    treatment, and the bonded device was left on an 80oC hotplate for 30 minutes. Circular wells

    were bonded as a second layer to the PDMS device to allow a reservoir for media during image

    analysis. Finally, tygon tubing was inserted into the channel ends at punch holes.

    The direct- flow device is designed with main channel dimensions: 300 μm in width, and

    125 μm in height. The width expands to 1800 μm in the islet-holding area. The average islet

    diameter is 150 μm; thus, the holding area was designed to accommodate this size. The bypass

    device main channel dimensions are 300 μm in width and 125 μm in height. The islet holding

    area is 300 width x 300 μm length to accommodate only 1 islet. The diameter of the nozzle is 50

    μm. Two prototypes are considered with different bypass lengths: short-pass (0.5 cm) and long-

    pass (1 cm). The islets were loaded into the device using gravity flow and imaged using confocal

    microscopy.

    4.5.2 Microfluidic COMSOL modeling

    The microfluidic device was drawn in AutoCAD, and imported into COMSOL

    Multiphysics as a DXF file. The 2- dimensional device was extruded to 3- dimensions by

    defining the channel height. The islet was positioned in the holding area and extruded as a 3-

    dimensional sphere. Laminar flow of water was chosen as the fluid type, as an approximation of

    the islet media. The wall boundaries, inlet and outlet were defined and the finite element mesh

    was set to free tetrahedral. The velocity and pressure profiles were created in the results section.

    We modeled the device in COMSOL assuming that the nozzle is fully blocked, in order to

    provide a visual representation of the system. This assumption is based on the observation that

    the flow rate through the islet is significantly lower than the flow rate through the main channel

    (Table 6.2.2).

  • 27

    4.5.3 Microfluidic device long- term islet culture

    Device-treated islets were loaded into the microfluidic devices using gravity flow shortly

    after isolation and incubated in a custom-built desk-top incubator [37]. Briefly, the microfluidic

    device was submerged in a stirred water bath at 37oC with flow driven by a syringe pump. The

    reservoir media was also submerged in a separate water-bath maintained just above 37oC to

    reduce formation of air bubbles in the microfluidic channel. A cap of mineral oil was placed on

    top of the media in the reservoir to reduce evaporation and drift in pH. A bubble- trap was also

    used to minimize bubble formation in the channels. To create the bubble trap, we hole-punched a

    second hole in the inlet channel, and inserted Tygon tubing. During islet culturing, the bubble-

    trap tubing was raised above the device. The bubbles preferentially entered the bubble- trap tube

    and allowed media to continually flow through the main channel. Islets were cultured in flow for

    48 hours. Post- culture the islets were stained with Flou4 for Ca2+ oscillations and PECAM-1 for

    endothelial cell detection.

    4.5.4 Microfluidic device image application

    The PDMS microfluidic devices were used in all imaging experiments to hold islets

    stationary and allow subsequent controlled flow of stimulation media. Prior to imaging, islets

    were loaded into the microfluidic device, subsequently the device was mounted into the 37oC

    temperature controller chamber and taped in place. The inlet and outlet tubing was fed through

    the exit holes on the side of the chamber, and the chamber was mounted onto the microscope

    stage over the 20x objective or the 40x oil immersion objective. The channel was brought into

    focus using the transmitted light. Prior to loading islets, the channel was washed using 2 mM

    glucose BMHH media (BMHH imaging media: 125 mM NaCl, 5.7 mM KCl, 2.5 mM CaCl2, 1.2

    mM MgCl2, 10 mM HEPES, pH 7.4) from the outlet towards the inlet using a 22 mm gauge

    blunt end needle- tipped syringe.

    Air bubbles were removed by applying pressure while washing the channel with BMHH.

    After three washes, the presence of bubbles in the channel was checked by visual examination

    through the eyepiece. Using the inlet tubing, the islets were loaded into the microfluidic device.

    It was important not to overload the channel with islets, or to allow gravity flow to continue for

    an extended period of time as loaded islets could be pushed underneath the drop wall or against

    the channel wall. The outlet tube was connected to a syringe on the digital syringe pump. The

  • 28

    inlet tube was clamped to allow media to flow through the well (Figure 4.1). Loaded islets were

    subjected to a 5 minute reset flow to allow reorientation in the channel.

    Outlet 

    Inlet 

    Islet holding area 

    Well

    Figure 4.1. Representative microfluidic device used for image analysis. The device was placed over the 20x or 40x objective lens and then washed with BMHH media (stained red in this image), and bubbles were removed from the channels by applying pressure through the inlet. Islets were flown in through the inlet tube by gravity flow. When the islets reached the holding area, the outlet tube was connected to a syringe pump and the inlet tube was clamped. The pump pulled media through the well at a constant rate.

    4.6 NAD(P)H Imaging

    Islets were loaded into a microfluidic device and stimulated with increasing glucose

    concentration while measuring changes in the levels of NAD(P)H. Changes in NAD(P)H levels

    were determined using live cell 2-photon microscopy. Endogenous NADH and NADPH are both

    autofluorescent at 705 nm, with an emission spectra of 380- 550 nm, giving off an aggregate

    signal of NAD(P)H. Stimulating the islets with a higher glucose concentration (20 mM) caused a

    rise in NAD(P)H signal with changes in intensity directly proportional to the level of glucose

    metabolism. The fold NAD(P)H response was expressed relative to the 2 mM baseline.

    NAD(P)H images are collected for islets treated with increasing glucose concentrations.

    Syringe Pump Outlet

    Well Inlet

    Islet holding area

  • 29

    ImageJ was used to quantify the mean intensity of NAD(P)H images by selecting and

    measuring the intensity of 20 small circular regions of interest on each islet. Regions were

    selected at random while avoiding nuclei and saturated pixels. Five regions were also measured

    at a distance from each islet to obtain average background intensity. The mean NAD(P)H

    intensity of each islet was calculated by averaging the 20 intra-islet measurements and

    subtracting the average background.

    4.7 Spherotech Velocity Profiles and Flou4 Ca2+ Oscillations

    Islets were loaded into the short- pass microfluidic device, and Spherotech beads

    (carboxyl fluorescent pink, 5.3 μm in diameter, 13x dilution, Spherotech Inc) were introduced

    into the device at a flow rate of 9.6 ml/ 24 hours. The beads were imaged using confocal

    microscopy with the 543 nm excitation wavelength. Time- series imaging was used to capture

    the flow of beads around the islet. The distance of the streaks was measured using Zeiss imaging

    software, and divided by the scan time in order to calculate the velocity.

    Islets were cultured in RPMI 1640 media (11 mM glucose, 10% FBS) in a dish (control)

    or short- pass device for 48 hours. Post- culture, islets were stained with 4 μM Fluo4 AM Ca2+

    dye (Invitrogen) for 2 hours in either the dish or short- pass device. For the β- cyclodextrin Flou4

    experiments, the islets were stained with 4 µM Fluo4 for 2 hours on the same day as isolation in

    the dish or short- pass device. The 4 µM Fluo4 was dissolved with 90 mM 2-hydoxypropyl β –

    cyclodextrin (Sigma Aldrich). Islets were incubated with 10 mM glucose for 15 minutes and then

    imaged using the 488 nm excitation wavelength. The penetration depth was analyzed by

    measuring the intensity profile across the islet periphery, and excluding the data that were less

    than 10% of the maximum intensity. This allowed us to uniformly assess the penetration depth in

    different islets.

    The islet Ca2+ oscillations were analyzed with ImageJ (Figure 4.2 A). The z-axis intensity

    profile was plotted through the image time-series. The periods of oscillations were determined

    with Matlab fast fourier transform (FFT). The FFT estimates the frequencies in the oscillation

    data from a discrete set of values sampled at a fixed rate. The sample frequency was ~1 Hertz (1

    sample/ second). A plot of power spectral density versus frequency (periodogram) was generated

    (Figure 4.2 B). The FFT generates a symmetrical graph; where any frequency on the left side

    was mirrored by the frequency on the right side. The period was then calculated as the inverse of

  • 30

    the frequency. One representative region was chosen in a single islet to obtain an estimate of the

    islet Ca2+ frequencies.

    Figure 4.2. Oscillations and power spectral density plot of glucose- stimulated Ca2+ responses in pancreatic islets. (A) Pancreatic islets were stained with 4 µM Fluo4 dye for 2 hours, and introduced into a microfluidic device for imaging. Islets were treated with 10 mM glucose for 15 minutes and the intensity changes were recorded using time- series imaging over 93 seconds. (B) Fast fourier transform plot of the data in (A) generated in Matlab. One frequency peak was detected at 0.07813 s-1. This corresponds to a period of 12.8 seconds.

    4.8 Statistical Analysis

    For experiments with two treatments, one-way and two- way student’s t-test were

    performed to determine significance. Data with p values < 0.05 were considered significant.

    For experiments with multiple treatments, a one-way ANOVA followed by the Tukey

    multiple comparisons test was used to assess statistical significance. Parings with p values <

    0.05 were accepted as statistically significant.

    0        16      31      47       62      78       93 

    A B

    Mean Intensity 

    Power Spe

    ctral D

    ensity 

    Sampling Time (s) Frequency (Hz)0           0.2         0.4          0.6         0.8          1.0

    55 

    65 

    75 

    85 

    200

    400

    600

  • 31

    Chapter 5 – FGF21 Secretion and Resistance

    5.1 Introduction

    FGF21 is secreted from hepatocytes due to induction of PPARα and FXR whereas

    secretion from adipocytes results from activation of the PPARγ [29, 30]. Circulating free fatty

    acids, such as palmitate, oleate and linoleate, are natural agonists to PPARα and -γ in hepatic and

    colon tissue, respectively [29, 45]. Dutchak et al. (2012) also demonstrated that FGF21 acts in a

    feed- forward loop in adipocytes to sustain the activity of PPARγ.

    We examined whether islets express FGF21 in response to excess nutrients and if this

    expression is temporally regulated. Mai et al. (2009) indicate that the relative FGF21 mRNA

    expression levels in liver cells treated with fatty acids increases over a period of 8 hours. This

    corresponded with increased FGF21 protein levels. Concurrently, Badman et al. (2007) reported

    that isolated hepatocytes treated with fatty acids show significant FGF21- induction after 24

    hours. Thus, we determined the palmitate- induced FGF21 mRNA expression after 4, 8, 16 and

    24 hour treatment to find the maximal induction.

    The elevated circulating levels of FGF21 in obese subjects may cause desensitization or

    resistance of this hormone, thereby diminishing its metabolic and autocrine signaling effects

    [34]. FGF21 causes a change in expression level of itself and its receptors in WAT and liver in

    obese models [21, 46]. To address FGF21 resistance, we cultured islets with excess nutrients and

    measured the change in FGF21, FGF receptor and co- receptor mRNA expression levels.

    In the future, we aim to measure the temporal secretion of FGF21 from islets. Sankar et

    al. (2011) demonstrated effluent collection from islets using a microfluidic device. Thus, we

    custom- designed a microfluidic device for our application. This new device has the ability to

    hold islets stationary in individual traps and reduce shear stress around the periphery of the islets.

    The device was designed to concentrate the effluent from islets after long- term culture.

  • 32

    5.2 Results

    5.2.1 Verifying FGF21 Responsiveness

    Initially, the islet responsiveness to FGF21 was assessed by creating a dose response

    curve. Typical FGF21 plasma concentrations vary in different studies, as seen in Table 5.1 [20,

    47, 48, 49, 50, 51], thus we aimed to establish an effective concentration of FGF21. Islets were

    treated with increasing levels of FGF21, and the autofluorescent NAD(P)H levels were

    measured. The NAD(P)H response provides a readout of the metabolic state of the cell,

    indicating the level of the TCA cycle activity. These results indicate that islets remain responsive

    to FGF21 as the concentration increases (Figure 5.2.1). The NAD(P)H response is reduced at a

    concentration of 100 ng/ml, therefore this concentration was chosen for future experiments.

    Table 5.1 – In vivo and in vitro concentrations of FGF21 FGF21 conc (ng/ml) In vivo Mice [48, 50] 0.2- 13 Human [51] 0.1- 5 In vitro β- cells [20] 1000 Adipocytes [47] 4000 BaF3 cells [49] 400- 800

    Figure. 5.2.1. Mitochondrial NAD(P)H dose response for islets treated with FGF21. Pancreatic islets were treated with 0, 1, 10, 100 and 1000 ng/ml of recombinant human FGF21 in standard RPMI 1640 media supplemented with 11 mM glucose and 10% FBS for 48 hours. Post- culture, islets were stimulated with 20 mM glucose and the NAD(P)H responses were recorded. The fold NAD(P)H was expressed relative to the 2 mM glucose baseline.

    FGF21 concentration (ng/ml)0         1       10      100    1000

    1.3 

    1.4 

    1.5 

    1.6 

    Fold NAD(P)H 

  • 33

    5.2.2. FGF21 expression is higher in mouse islets compared to liver and WAT

    FGF21 transcript is predominately found in the pancreas and hepatic tissues of mice

    (Figure 1.6) [24]. To determine FGF21 transcript expression in endocrine pancreas, we

    compared the expression level of FGF21 by reverse transcriptase PCR (RT-PCR) in islets

    relative to liver, WAT, Min6 β- cells and αTC1 cells. We observed more defined bands in islets

    and liver compared to WAT, α- and Min6 β- cells (Figure 5.2.2 A). We further examined

    transcript expression by quantitative PCR (qPCR). The qPCR data in Figure 5.2.2 B shows that

    the highest expression of FGF21 occurs in islets. This result corresponds with literature results

    which show elevated FGF21 expression levels in the pancreas [24].

    Islets1      2     3     4 

    Liver1      2     3    

    WAT1      2     3    

    Gapdh

    αTC1 cells1       2     

    Min6 β‐cells1      2

    FGF21

    Figure. 5.2.2. FGF21 expression is higher in mouse islets compared to liver and WAT. (A) RNA was isolated from pancreatic islets, liver, WAT of C57BL6 mice and αTC1 and Min6 β- cells and reverse transcribed. The RT-PCR reaction was set up with FGF21 and Gapdh primers. The numbers above each gel indicate the samples assayed. (B) qPCR data representing expression levels of FGF21 relative to actin for 3 independently assayed mice. Exception: WAT qPCR was performed with only 2 mice (One-way ANOVA *P

  • 34

    5.2.3 Palmitate, Bezafibrate and Rosiglitazone induce FGF21 expression in Min6 β- cells and in islets

    Elevated circulating levels of FGF21 are present in models of obese mice [21]. We

    therefore investigated whether the saturated fatty acid (palmitate) induces the expression of

    FGF21 in Min6 β- cells and αTC1 cells. As seen in Figure 5.2.3. A, palmitate appears to increase

    FGF21 transcript expression in α- and β- cells after 16 hours of treatment.

    Subsequently, we determined if palmitate, unsaturated fatty acids (oleate or linoleate),

    and PPARα/γ agonists induced FGF21 expression in the Min6 β- cell line (Figure 5.2.3 B).

    Saturated fatty acids such as palmitate were previously shown to cause lipoapoptosis in human β-

    cells; however, unsaturated fatty acids (oleate and linoleate) actually prevent stearate- induced

    lipoapoptosis [52, 53]. FGF21 is produced in the liver via PPARα, and in WAT through PPARγ

    induction [10]; thus, we included bezafibrate (PPARα agonist) and rosiglitazone (PPARγ

    agonist) as positive controls.

    We observed a quantitative induction of FGF21 transcript in Min6 β- cells stimulated

    with palmitate, bezafibrate and rosiglitazone for 24 hours (Figure 5.2.3. B). However, no

    induction was observed with oleate or linoleate. We similarly observed a more moderate

    increase in pancreatic islets in response to palmitate and robust responses to both PPAR agonists

    (Figure 5.2.3 C). Our data suggest that α- and β- endocrine cells respond to saturated fatty acids

    by producing FGF21 in a PPAR- dependent manner. We postulate that elevated levels of FGF21

    could then act in a protective manner to diminish glucolipotoxicity.

  • 35

    Figure 5.2.3. Palmitate induces FGF21 expression in β- cells, α- cells and islets. (A) Min6 β- cells and αTC1 cells were treated with 0.2 mM palmitate for 16 hours in RPMI 1640 media containing 5 mM glucose and 1% FBS. RNA isolation was performed and the RNA was reverse transcribed. The cDNA PCR reaction was set up with FGF21 and Gapdh primers. The numbers above each gel indicate the samples assayed. (B & C) qPCR data for Min6 β- cells and pancreatic islets. Min6 β- cells and islets were cultured for 24 hours in RPMI 1640 media with 5 mM glucose, 1% FBS and 0.2 mM palmitate, oleate or linoleate, 250 μM bezafibrate or 5 μM rosiglitazone. The qPCR data represents expression levels relative to actin for 3 independently assayed and treated β- cell samples and mice (One-way ANOVA *P

  • 36

    5.2.4. Palmitate- induces FGF21 expression in a temporal manner in Min6 β- cells

    To determine the temporal effect of saturated fatty acid- induction of FGF21, we treated

    Min6 β- cells for 4, 8, 16 and 24 hours with 0.2 mM palmitate. The data indicate that maximum

    induction of FGF21 occurs after 16 hours of treatment, followed by decreased expression after

    24 hours of treatment (Figure 5.2.4). FGF21 expression may be attenuated after 16 hours due to

    translocation of STAT (signal transducer and activator of transcription) to the nucleus to affect

    transcription [13].

    Figure. 5.2.4. Palmitate induces FGF21- expression in a temporal manner. Min6 β- cells were cultured for 4, 8, 16 and 24 hours in RPMI 1640 media containing 5 mM glucose and 1% FBS with 0.2 mM palmitate, 250 μM bezafibrate or 5 μM rosiglitazone. The qPCR data represents expression levels relative to actin for 3 independently assayed β- cell samples (One way ANOVA *P

  • 37

    5.2.5 Elevated levels of FGF21, glucose and palmitate do not change FGF21, FGF receptor and co-receptor mRNA expression levels in islets

    The ability of FGF21 to increase glucose tolerance and reduce circulating triglycerides is

    absent in models of obesity, suggesting a state of FGF21 resistance [21]. FGF21 resistance can

    occur at many different levels such as:

    Enzymatic inactivation of FGF21

    Alterations in the FGF21 receptor mRNA expression

    Alterations in FGF21 protein trafficking to the cell membrane

    Modifications to the FGF21 receptor

    Alterations in the metabolic pathways down-stream of FGF21 receptors [54]

    Fisher et al. (2010) and Coskun et al. (2008) report that FGF21 and FGF receptor

    expression levels were significantly down- regulated in the WAT of obese mice. Hence, we

    examined whether excess nutrients reduce receptor and co- receptor expression in pancreatic

    islets. A reduction in FGF receptor expression levels would hypothetically reduce FGF21

    autocrine signaling [21].

    Pancreatic islets were cultured in the presence of elevated levels of FGF21, palmitate or

    glucose. Concentrations were chosen to represent serum levels typically found in diabetic obese

    mice [55, 56, 57] (100 ng/ml FGF21, 20 mM glucose and 0.2 mM palmitate). The results show

    that elevated nutrients do not cause FGF21, FGF receptor or co- receptor down- regulation

    (Figure 5.2.5), suggesting that FGF21 resistance does not occur in islets through reduced

    receptor expression. Future studies are needed to confirm alternative mechanisms of resistance.

  • 38

    Figure. 5.2.5. FGF21, glucose and palmitate treatment do not change FGF21, FGF receptor and co-receptor expression levels. (A) Islets were cultured for 24 hours in RPMI 1640 media containing 11 mM glucose and 10% FBS the absence (control) and presence of 100 ng/ml FGF21. (B & C) Islets were cultured for 24 hours in RPMI 1640 media containing 10% FBS with 5, 11 and 20 mM glucose. (D & E) Islets were cultured for 24 hours in RPMI media containing 5 mM glucose and 10% FBS in the absence (control) and presence of 0.2 mM palmitate. The qPCR data represents expression levels relative to actin for the islets from 3 independently assayed and treated mice. Exception: (B & C) results were taken from 2 independently assayed mice.

    A

    B C D E

            R

    elative mRN

    A expression 

            R

    elative mRN

    A expression 

    FGF21    FGFR1c   FGFR2c       KLB

    ControlFGF21

    FGFR1c    KLB   FGFR1c    KLB   

     1 

     1 

    0

    1

    2

     1 

    Glucose (mM)  Glucose (mM) Palmitate (mM)  Palmitate (mM) 5      11      20  5      11      20 0      0.2  0      0.2

  • 39

    5.3 Discussion

    We report that FGF21 mRNA expression levels are higher in islets compared to liver and

    WAT of lean C57BL6 mice (Figure 5.2.2). These data imply that pancreatic islets could play an

    important role in whole body FGF21 production and/or act as an autocrine factor.

    Elevated circulating levels of fat are accompanied by excess FGF21 in the serum [21,

    34]. This led us to examine the effect of fatty- acid induction of FGF21 expression. Our results

    show that saturated fatty acids (palmitate), but not unsaturated fatty acids (oleate or linoleate),

    induce the expression of FGF21 in Min6 β- cells and in islets (Figure 5.2.3). This fits with our

    hypothesis that elevated levels of fat induce FGF21 expression and secretion in islets. Palmitate

    increases the production of ROS in rat islets [8], thus FGF21 may lower oxidative and ER stress

    by reducing the accumulation of fatty acid intermediates. The maximum induction of FGF21

    occurs after 16 hours of palmitate treatment (Figure 5.2.4), followed by a decli