PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE . DATE DUE 5/08 K:IProj/Aoc&Pres/CIRCIDaIeDuoIindd ———,——-— PAN C AND M PANCREATIC B-CELL FATTY ACID METABOLISM AND MODULATION OF FUNCTION IN RESPONSE TO GLUCOLIPOTOXICITY By Christopher D. Green A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Biochemistry & Molecular Biology 2009 r MODEL} TyPc funCliOn in nhhiddh decreased in nun.mm increased lit Activation 0f gene express 0813. H6“? hyperglycemi andGSlS. Li expression of synthesis and together with 1 enhanced base funnier of hi increased iipoi as diacyigiycei proteins reduce ABSTRACT PANCREATIC B-CELL FATTY ACID METABOLISM AND MODULATION OF FUNCTION IN RESPONSE TO GLUCOLIPOTOXICITY By Christopher D. Green Type 2 diabetes is associated with gradual diminishment of pancreatic islet B-cell function in response to chronic hyperglycemia and elevated plasma free fatty acids (FFAs), defined as glucolipotoxicity. The effects of glucolipotoxicity on B-cells include decreased insulin gene expression, diminished glucose-stimulated insulin secretion (G818), and ultimately decreased B-cell mass. Loss of B-cell GSIS has been linked to increased lipogenic gene expression and triacylglyceride (TAG) accumulation. Activation of the liver X receptor (LXR) transcription factor further increases lipogenic gene expression and TAG accumulation but increases both basal insulin release and G318. Here, INS-1 B-cells treated with the LXR agonist T0901317 during chronic hyperglycemia increased lipogenic gene expression, de novo synthesis of TAG, and basal and G818. LXR-activated INS-1 cells exhibited increased fatty acid (FA) oxidation and expression of genes involved in mitochondrial B-oxidation. Inhibition of fatty acyl-CoA synthesis and mitochondrial B-oxidation blocked the elevated basal insulin release. Thus, together with the rapid turnover of TAG in LXR-activated cells, these results indicate that enhanced basal insulin release involves oxidation of fatty acyl-CoAs generated during turnover of neutral lipid pools. Increased synthesis and turnover of TAG suggested increased lipolysis of complex lipids and the generation of lipid signaling molecules such as diacylglycerol. In this manner, inhibition of TAG turnover and diacylglycerol binding proteins reduced the LXR-mediated increase in G818. LXR acli synthesis and 61“ limiting enlimcs to be elerated in rat islets had red llL'FA synthesi: B-cell response 1 Ms. cause tox integrity, \s'hicl‘ siRVAs and ads was examined f (16:0). Knocki lNS-l cells to SCD2 increase toxicity. Elosl “Slim-9) and 0"eberrpressio Palmitate-indu Elorl.6 and E maintaining b; In Con mmOVEr and s LXR activation in INS-1 cells also increased monounsaturated F A (MUFA) synthesis and elevated stearoyl-CoA desaturases (SCD) l and 2 gene expression, rating- limiting enzymes in MUFA synthesis. SCD] and 2 gene expression were then identified to be elevated in pre-diabetic Zucker diabetic fatty (ZDF) rat islets, whereas diabetic ZDF rat islets had reduced expression of SCDl, SCD2 and Elovl-6, a FA elongase involved in MUFA synthesis. These findings suggested SCDs and Elovl-6 could be involved in the B-cell response to metabolic load. Elevated exogenous FFA levels, particularly saturated F As, cause toxic effects to B-cells that include altered endoplasmic reticulum (ER) integrity, which is linked to induction of ER stress responses and apoptosis. Using siRNAs and adenoviral constructs, altered SCD or Elovl-6 gene expression in INS-l cells was examined for its effects on ER stress and apoptosis mediated by exogenous palmitate (16:0). Knockdown of SCDs decreased MUFA synthesis and increased susceptibility of INS-1 cells to palmitate-induced ER stress and apoptosis, whereas over-expression of SCD2 increased palmitate desaturation to palmitoleate (16:1,n-7) and reduced palmitate toxicity. Elovl-6 knockdown decreased palmitate conversion to stearate (18:0) and oleate (18:1,n-9) and tended to reduce palmitate-induced ER stress and apoptosis, while Elovl-6 over-expression increased synthesis of 18:0 and 18:1,n-9 and increased susceptibility to palmitate-induced toxicity. Further studies showed that coordinated expression of SCDs, Elovl-6 and Elovl-S, which elongates 16:1,n-7 to vaccenate (18:1,n-7), is required for maintaining balanced de novo synthesis of n-7 versus n-9 MUFAs. In conclusion, these studies demonstrate that modulation of TAG synthesis and turnover and MUFA synthesis significantly alters the B-cell response to glucolipotoxicity. T'n' number of teaching in friendship. . like to thanl for their gui Dr. Busik ar. Fourth. l apr members oft Katrina Linn Fifth. my file memorable, e Last b and my bf 01hr school 1 “on “3”“ “won, and OUr new ]j_ would n01 be p Acknowledgements This disseration would not be possible without the support and help from a number of people. First, I would like to sincerely thank my advisor Dr. Olson for teaching me how to do scientific research and writing, as well as providing advice, friendship, and encouragement related to research and life in general. Second, I would like to thank my committee members Dr. Jump, Dr. Benning, Dr. DeWitt and Dr. LaPres for their guidance throughout my dissertation research. Third, I would also like to thank Dr. Busik and Dr. Parameswaran for their helpful discussions and technical support. Fourth, I appreciate the technical guidance, support, and friendship of all past and present members of the Olson, Jump, Busik, Parameswaran and Uhal labs, especially Diana Ye, Katrina Linning, Jinghua Xu, Yun Wang, Barbara Christian and Madelina Opreanu. Fifth, my friends outside of lab have made this journey through grad school fun and memorable, especially Trevor Barkham and Scott Brock. Last but not least, I would like to express my sincere appreciation to my parents and my brother and his wife for their unconditional support throughout my life and grad school. I would also like to sincerely express my deep appreciation and graditude for the warm support, encouragement, and help of my wife Hui during this dissertation research and our new life together. Without the unwavering support of these people, this work would not be possible. iv TABLE OF List of Tables. List of Figures List of Abbres INTRODL'C T Chapter 1. Li 1. Fatty Acid 1 1.1. Fatty A 1.2. Modifit 1.2.1. Dc 1.2.2. E1 13.EXOgcn 1.4. De Km 1.5 Gi}‘cen 1.6. Transcr 1.6.1. C5 1.6.2. St 1.6.3. Li 1.7. Fairy A 2. Pancreatic 1 2.1. Mechar 22- Regula 2.3. MiiOCh 2'4- Genera TABLE OF CONTENTS List of Tables ....................................................................................... viii List of Figures ....................................................................................... ix List of Abbreviations ............................................................................... xi INTRODUCTION ................................................................................... 1 Chapter 1. Literature Review 1. Fatty Acid Metabolism and Regulation of Lipogenic Gene Expression ............... 3 1.1. Fatty Acid Structure and Classification ................................................... 3 1.2. Modification of Fatty Acid Structure ...................................................... 4 1 .2.1 . Desaturation ............................................................................ 4 1.2.2. Elongation .............................................................................. 5 1.3. Exogenous Fatty Acid Uptake and Essential Fatty Acids .............................. 8 1.4. De Novo Fatty Acid Synthesis ............................................................. 9 1.5. Glycerol 3-Phosphate Pathway of Triacylglycerol Synthesis ......................... 12 1.6. Transcriptional Regulators of Lipogenesis ............................................. 14 1.6.1. Carbohydrate Response Element Binding Protein .............................. 14 1.6.2. Sterol Regulatory Element Binding Protein-1c ................................... 15 1.6.3. Liver X Receptors .................................................................... 16 1.7. Fatty Acid Oxidation ...................................................................... 18 2. Pancreatic B-Cell Insulin Secretion and Role of Fatty Acid Metabolism ............ 20 2.1. Mechanism of B-Cell Insulin Secretion ................................................. 20 2.2. Regulation of Fatty Acid Metabolism in Pancreatic fi-cells .......................... 22 2.3. Mitochondrial Anaplerosis/Cataplerosis ................................................ 24 2.4. Generation of Lipid Signaling Molecules ............................................... 25 2.4.1. Role of Malonyl-CoA ............................................................... 25 2.4.2. Free Fatty Acid, Long Chain Acyl-CoA, and Diacylglycerol .................. 26 2.5. Lipolysis and Glycerolipid/Fatty Acid Cycling ........................................ 28 2.5.1. Neutral Glycerolipid Lipolysis ..................................................... 28 2.5.1.1. Hormone-Sensitive Lipase .................................................... 28 2.5.1.2. Adipose Triacylglyceridc Lipase .................................... ,,, ...... 29 2.5.2. Phospholipases C and D ............................................................. 30 2.5.3. Phospholipase A2 ............................................................... ,,,. . .32 3. Glucose and Fatty Acid Mediated Pancreatic fi-Cell Dysfunction .................... 34 3.1. Glucotoxicity vs. Glucolipotoxicity in Type 2 Diabetes .............................. 34 3.2. Mechanisms of B-Cell Failure ............................................................ 35 3.2.1. Endoplasmic Reticulum Stress ..................................................... 35 3.2.2. Oxidative Stress ...................................................................... 36 3.2.3. Malonyl-CoA Inhibition of Fatty Acid Oxidation and Lipid Accumulation ................................................................................................. 37 U) (J) ('71 L) I-J . 'J‘ 4.. -". H C)... L.) L» J D) b) J Lu b) J l J I... b U U 4. Statemen Chapter 2. Methods... 1. Mater 2. F atty : 3. Huma 4. Anima 5. [NS] 6. siRVA 7. Adena 8. RNA 5 9. Weste 10. lnsul 1]. Glue- 12. Com 13. Palm l4. Stati: Chapter 3. Abstract. . . lmmductio “"18 ..... 3.1. Effe 3.2.4. Dysregulated Glycerolipid/Fatty Acid Cycling .................................. 38 3.2.5. Apoptosis-Mediated Loss of B-Cell Mass ........................................ 39 3.3. Evidence and Mechanisms of B-cell Compensation ................................... 42 3.3.1. SREBP-lc and Liver X Receptors ................................................. 42 3.3.2. Glycerolipid/Fatty Acid Cycling .................................................... 43 3.3.3. Monounsaturated Fatty Acid Synthesis ........................................... 43 4. Statement of Problem and Specific Aims ................................................... 45 Chapter 2. Materials and Methods .............................................................................................. 47 1. Materials ...................................................................................... 47 2. Fatty acid preparation ..................................................................... 47 3. Human islets ................................................................................. 48 4. Animals and islet isolation ................................................................ 48 5. INS-l cell culture ............................................................................ 49 6. siRNA treatment ............................................................................ 49 7. Adenovirus preparation ................................................................... 50 8. RNA analysis ................................................................................. 51 9. Western blot analysis ...................................................................... 55 10. Insulin secretion studies .................................................................. 55 ll. Glucose utilization studies ............................................................... 55 12. Complex lipid and fatty acid analysis ................................................. 56 13. Palmitate oxidation .......................................... . .............................. 57 14. Statistical analysis ......................................................................... 57 Chapter 3. Elevated Insulin Secretion From Liver X Receptor-Activated Pancreatic B-Cells Involves Increased de Nova Lipid Synthesis and Triacylglyceride Turnover .............................................................................. 58 Abstract .............................................................................................. 58 Introduction ........................................................................................ 59 Results ................................................................................................ 62 3.1. Effect of glucose and LXR activation on SREBP-l and lipogenic gene expression ................................................................................... 62 3.2. Effect of glucose and LXR activation on lipogenesis ................................. 65 3.3. Effect of LXR activation on FA profile ................................................. 66 3.4. Impact of LXR activation on basal and glucose-stimulated insulin secretion. .....71 3.5. Role of FA oxidation in elevated basal insulin release ............................... 74 3.6. Role of TAG turnover in elevated insulin release ..................................... 77 Discussion ........................................................................................... 85 Chapter 4. Stearoyl-CoA Desaturase Modulates Palmitate-Induced Endoplasmic Reticulum Stress and Apoptosis in Pancreatic B-ells ........................ 91 Abstract .............................................................................................. 91 Introduction ......................................................................................... 92 Results ................................................................................................ 95 4.1. Rat islet and INS-1 cell FA desaturase and elongase gene expression profiles. ...95 vi ‘ 10;. "' My fi< ink fiffii; _i .. . v ‘ .' ..’. f9 ::E. vs 2" ,9 ? «-‘ ..:_ ; ; VJ .,. L). ’I I. ' Ea. I‘. ' r ._3‘ '19 .0 ?'=. O“ z. -t. I:.. -1 I;l- 1.0, .v: 137. It, 0' D 4d- .3: ‘5.- 51'1”". 0‘ o. I D r ”o"! O M '. ‘ '2‘!“ 'b'i‘. '3‘. ‘Vp'-"'-9‘J‘t.i manamngcam; 4.2. PA de: 4.3. Knock 4.4. Suscep 4.5. SC D k 4.6. Over: and m: 4.7. Effect: 4.8.8631! diac) actisation ..... Discussion. . . . Chapter 5. R M Abstract ...... introduction . Results ........ 5.1. Regulg 5.2. Elevate ratio or 5.3. siRNA Speciti. 5.4. Over-e ML’FA Discussion... . Chapter 6. G BIBLIOGRA. 4.2. FA desaturase and elongase gene expression in ZDF rat islets ...................... 99 4.3. Knockdown of SCD and Elovl-6 gene expression modulate MUFA synthesis. 103 4.4. Susceptibility to palmitate-induced ER stress is increased by SCD knockdown .............................................................................................................................. 106 4.5. SCD knockdown impacts susceptibility to palmitate-induced apoptosis ......... 107 4.6. Over-expression of SCD2 and Elovl-6 differentially modulate MUFA synthesis and markers of ER stress ................................................................ 113 4.7. Effects of SCD2 and Elovl—6 over-expression on palmitate-induced apoptosis .............................................................................................. 114 4.8. Elevated CHOP expression by SCD knockdown coincides with increased diacylglycerol formation and involves Ca2+-dependent PKC activation ....... 120 Discussion ............................................................................................ 123 Chapter 5. Role of Fatty Acid Elongases in Determination of De Novo Synthesized Monounsaturated Fatty Acid Species ......................................... 128 Abstract ............................................................................................ 128 Introduction ....................................................................................... 129 Results .............................................................................................. 131 5.1. Regulation of FA elongase and desaturase genes by glucose ....................... 131 5.2. Elevated glucose increases abundance of de novo synthesized F As and alters the ratio of saturated to monounsaturated FA ............................................. 132 5.3. siRNA knockdown of elongases and desaturases modulates the synthesis of specific FA species ....................................................................... 136 5.4. Over-expression of Elovl-S or Elovl-6 leads to selective synthesis of specific MUFA species ............................................................................ 141 Discussion .......................................................................................... 147 Chapter 6. General Conclusions and Future Studies .................................... 151 BIBLIOGRAPHY ................................................................................ 1 5 7 vii Table 2.1... Table 2.2... Table 2.3... Table 4.1... List of Tables Table 2.1 ............................................................................................. 52 Table 2.2 ............................................................................................. 53 Table 2.3 ............................................................................................. 54 Table 4.1 ............................................................................................ 101 viii Figure 1. Figure 1. Figure 1.. Figure l.-‘ Figure 1.5 Figure 1.6 Figure 1.7 Figure 3.4.. Figure 3.5.. Figure 3.6... Hflm37u Figure 3.8... Figure 4.1.... List of Figures Figure 1.1 .............................................................................................. 7 Figure 1.2 ............................................................................................. 11 Figure 1.3 ............................................................ i ................................. 13 Figure 1.4 ............................................................................................. 17 Figure 1.5 ............................................................................................. 21 Figure 1.6 ............................................................................................. 33 Figure 1.7 ............................................................................................. 41 Figure 3.1 ............................................................................................. 63 Figure 3.2 ............................................................................................. 68 Figure 3.3 ............................................................................................. 70 Figure 3.4 ............................................................................................. 73 Figure 3.5 ............................................................................................. 76 Figure 3.6 ............................................................................................. 81 Figure 3.7 ............................................................................................. 83 Figure 3.8 ............................................................................................. 85 Figure 4.1 ............................................................................................. 97 Figure 4.2 ........................................................................................... 102 Figure 4.3 ........................................................................................... 105 Figure 4.4 ........................................................................................... 109 Figure 4.5 ........................................................................................... 112 Figure 4.6 ........................................................................................... 1 16 Figure 4.7 ........................................................................................... 119 ix Figure Figure .' Figure 5 I It Figure Figure 5. Figure 5. Figure 6.. Figure 4.8 ........................................................................................... 123 Figure 5.1 ........................................................................................... 133 Figure 5.2 ........................................................................................... 136 Figure 5.3 ........................................................................................... 141 Figure 5.4 ........................................................................................... 146 Figure 5.5 ........................................................................................... 148 Figure 6.1 ........................................................................................... 155 AA ABC AC C Ad ADP AGPAT AMP AHPK ATF CEBP CHOP C‘nRE ChREBP CPT DAG .151) £161) AA ABC ACC Ad AGPAT AMPK _ ATF ATGL ATP B-gal bHLH C E C/EBP CHOP ChRE ChREBP CPT DAG ASD A6D List of Abbreviations arachidonic acid ATP-binding cassette acetyl-CoA carboxylase adenovirus adenosine 5 ’-diphosphate acylglycerol-3-phosphate acyltransferase adeonsine monophosphate AMP-activated protein kinase activating transcription factor adipose triglyceride lipase adenosine 5’-triphosphate B—galactosidase basic helix-loop helix cholesterol ester CCAAT/enhancer-binding protein C/EBP homologous protein carbohydrate response element carbohydrate response element binding protein carnitine palmitoyl transferase diacylglyceride delta-5 desaturase delta-6 desaturase xi A90 DGAT DNA E1011 ER FA FAS FFA GK GLL’TZ GPAT GSlS HSL INS-l IREI JNK KREB LCAD LC~C0A LDL LPA L-PK LuQ A9D DGAT DNA Elovl ER FA FAS FFA GK GLUT2 GPAT GSIS HSL INS-1 IREl JNK KREB LCAD LC-CoA LDL LPA L-PK Luc delta-9 desaturase diacylglyerol acyltransferase deoxyribonucleic acid fatty acid elongase endoplasmic reticulum fatty acid fatty acid synthase free fatty acid glucokinase glucose transporter 2 glycerol-3-phosphate acyltransferase glucose-stimulated insulin secretion hormone sensitive lipase rat insulinoma cell line inositol requiring ER-to-nucleus signal kinase 1 c-Jun N-terminal kinase Kreb’s Ringer bicarbonate buffer long chain acyl CoA dehydrogenase long chain-acyl-coenzyme A low density lipoprotein lysophophatidic acid liver pyruvate kinase luciferase xii L-\'GC C LXR ME hiLX thPAT ML'FA NADH NADPH PA PAP PC PDH PERK PKC PLA2 PLC PPARo PL'FA W R RVA ROS RP137 G RYR L-VGCC LXR ME MLX thPAT MUF A NADH NADPH PA PAP PC PDH PERK PKC PLA2 PLC PPAROt PUFA qPCR ROS RPL32 L-type voltage-gated calcium channel liver X receptor malic enzyme Max-like protein X mitochondrial GPAT monounsaturated fatty acid B-nicotinamide adenine dinucleotide B-nicotinamide adenine dinucleotide phosphate phosphatidic acid 3 phosphatidic acid phosphatase pyruvate carboxylase pyruvate dehydrogenase double-stranded RNA-activated kinase (PKR)-like ER kinase protein kinase C phospholipase A2 phospholipase C peroxisome proliferator activated receptor-or polyunsaturated fatty acid quantitative PCR ribonucleic acid reactive oxygen species ribosomal protein L32 retinoid X receptor xiii TO TED VLAD \lDl. Xbp 1 ZDF ZF 16:1,n.7 18:0 18:1,n-7 1811.n-9 1333,.n-6 30:33-9 ~04 11.6 i 20.5 [1,3 ’ 3236M SCAP SCD SREBP TAG TCA T0 T2D VLAD VLDL Xbpl ZDF ZF 16:0 16: 1 ,n-7 18:0 18: 1,n-7 18:1,n-9 18:2,n-6 18:3,n-6 20:3,n-9 20:4,n-6 20:5,n-3 22:6,n-3 SREBP cleavage activating protein stearoyl-CoA desaturase sterol regulatory element binding protein triacylglyceride tricarboxylic acid acyle T0901317 type 2 diabetes very long chain acyl CoA dehydrogenase very low density lipoprotein X-box binding protein 1 Zucker diabetic fatty rat Zucker fatty rat palmitic acid palmitoleic acid stearic acid vaccenic acid oleic acid linoleic acid linolenic acid mead acid arachidonic acid eicosapentaenoic acid docosahexeanoic acid xiv Ty projected diabetes re cell insulir insulin to dl'Sregulate elevated p fimCllOn, glucolipoto giucose-stir majm €03] mechanism Mm glue01113010 INTRODUCTION Type 2 diabetes (T2D) accounts for more than 90% of diabetes cases, which is projected to affect 300 million people worldwide by 2025 (1, 2). Whereas type 1 diabetes requires insulin administration due to the complete absence of pancreatic islet B- celliinsulin secretion, T2D results, in part, from the inability of B-cells to secrete enough insulin to overcome insulin resistance and maintain glucose homeostasis. As dysregulated glucose metabolism develops, sustained periods of hyperglycemia and elevated plasma free fatty acids (FFAs) contribute to the progressive loss of B-cell function, collectively termed glucolipotoxicity (3). The adverse effects of glucolipotoxicity on B-cells include decreased insulin gene expression, diminished glucose-stimulated insulin secretion (GSIS), and ultimately the loss of B-cell mass (4). A major goal of our lab is to understand how these effects develop and to identify mechanisms to protect from B-cell dysfunction. Modulation of fatty acid (FA) metabolism in B-cells in response to glucolipotoxicity is essential for maintaining proper function. Chronic exposure of B- cells to elevated levels of glucose or glucose plus FFAs increases the storage of FA in triacylglyceride (TAG) and diminishes GSIS (3). Thus, accumulation of TAG in B-cells has been associated with the pathogenesis of B-cell dysfunction. Evidence has emerged, however, for protection of B-cells from glucolipotoxicity by modulating the expression and activity of genes involved in de nova FA synthesis (lipogenesis) and TAG storage. This includes regulation of lipogenic genes via liver X receptor (LXR) activation and genes involved in monounsaturated FA synthesis (5, 6). This dissertation provides insight into the mechanism of elevated insulin secretion from LXR-activated B-cells r . -<- d -.5 r {uL-IFC» ’ 4 g-12..&o.a‘~::; u. guano 0-: ~ .Q\: o I- o I 05:75; o~. to)? fig; ‘10,.- . O- r :55; .- swat-twp." . '3""T'~" ‘3' " 7" "" _ 5.324%“ manger . ,7 ‘xy'- ' 'v13 0 i l i 1 h under ck desaturas fi-ceils tc that cault ,4: I ! I i y. . ' ' . < - 21'! '1 =9" -..«....._,.. - .w_.-,_.. under chronic hyperglycemia, and examines the roles of altered expression of FA desaturase and elongase genes involved in monounsaturated FA synthesis in response of B-cells to exogenous saturated F As. The findings of this research identify mechanisms that could be utilized to prevent the onset of B-cell failure and T2D. Chapter 1. Fafl)‘ A 1.1. F arty Fat stabilizing signal Iran structure. group and hydrocarbo saturated F represented unsaturated monounsatt iPL'FAS), C, denoting 1h, end_ For e: carbons, One PLfFA SUCh the first do“ consisrs Of a Chapter 1. Literature Review 1. Fatty Acid Metabolism and Regulation of Genes Involved in Lipogenesis 1.1. Fatty Acid Structure and Classification Fatty acids (FAs) serve numerous essential biological functions, such as stabilizing cellular membranes, providing energy storage depots, and participating in signal transduction. The efficacy of these functions is largely dependent on the FA structure. FAs are composed of hydrocarbon chains of various lengths with a methyl group and a carboxyl group residing at opposite ends of each chain. FAs with hydrocarbon chains that are completely saturated with hydrogen atoms are termed saturated FAs. An example of a saturated FA is the sixteen-carbon FA palmitic acid, represented as 16:0. Modified FAs containing one or more double bonds are termed unsaturated FAs. The number of double bonds further categorizes these FAs into monounsaturated FAs (MUFAs), containing one double bond, and polyunsaturated FAs (PUF As), containing two or more double bonds. Unsaturated FAs are characterized by . denoting the position of the carbon of the first double bond, counting from the methyl end. For example, the MUFA oleic acid is represented as 18:1,n-9, as it has eighteen carbons, one double bond, and the double bond is nine carbons from the methyl end. A PUFA such as arachidonic acid. (20:4,n-6) has four double bonds and only the position of the first double bond is listed. As in all cells, the cellular FA composition of islet B-Cells consists of a broad range of saturated and unsaturated FAs, the concentrations of which are under tight regulation. .J.‘ 25;” _ 1.2. Mod 1.2.]. De: M reductase. cis-double addition 0 called stea double bor involved ir. acid (18:1,r ('7). ASD ; (20:339.). 2 and MD ar brain and he and B'cells (_ Stear Particular;y I include four hamster {SC} SUbStraie spec 4 p reference 101 stearic acid ( 1.2. Modification of Fatty Acid Structure 1.2.1. Desaturation Modification of F As by oxidative desaturation uses NADH-cytochrome b5 reductase, cytochrome b5, and a desaturase to convert a single carbon-carbon bond to a cis-double bond (Figure 1.1). In mammals, the FA desaturases capable of enzymatic addition of a cis-double bond include delta 5 desaturase (ASD), A6D, and A9D, also called stearoyl-CoA desaturase (SCD). The ‘A’ refers to the position of the carbon the double bond is added, counting from the carboxyl end. The ASD and A6D genes are involved in PUFA synthesis. Substrates preferentially desaturated by the A6D are oleic acid (18:1,n-9) and the C18 and C24 PUFAs, whereas ASD prefers C20 PUFA substrates (7). ASD and A6D activities [are essential for the synthesis of the PUFAs mead acid (20:3,n-9), arachidonic acid (20:4,n-6), and docosahexaenoic acid (22:6,n-3) (7, 8). ASD and A6D are ubiquitously expressed with the highest level found in liver, followed by brain and heart (9). The ASD and A6D genes were identified to be expressed in rat islets and B-cells (10), but a unique role in B-cell function has not been demonstrated. Stearoyl-CoA desaturase (SCD) is the rate-limiting enzyme for MUFA synthesis, particularly palmitoleate (16:1,n-7) and oleate (18:1,n-9). SCD isoforms cloned thus far include four from mouse (SCD1—4) (11-14), two in rat (SCDl and 2) (15), three in hamster (SCD1-3) (16), and three in human (SCDl, 2, and 5) (17-19). Analysis of substrate specificity using microsomal fractions from cells over-expressing SCD 1, 2, or 4 demonstrated the ability to desaturate C13-Cl9 saturated FAs with a particular preference towards stearic acid (18:0) (20). SCD3 desaturated C13-C16 FAs but not stearic acid (18:0), suggesting that SCD3 should be redefined as a palmitoyl-CoA desaturase (3 enhanced ca; fold higher 1 oleic acid (1 lipid storage expression in formation ( 1 1 SCD2 is expr is restricted t. for the skin ( (13). AlthOi and vvax ester IiPld metabo ellPicssion pr Cell function 1 1.2.2. EIOnga FA e1. calaij'zEd in 1‘. using NADP} The me‘llmir; which are Sub m OlBe2 rat, an desaturase (20). SCD] is highly expressed in adipose tissue and liver, tissues with enhanced capacity for neutral lipid storage (12). The activity of SCD] is more than 2- fold higher for stearic acid than palmitic acid (20). This in turn increases synthesis of oleic acid (18:1,n-9) and correlates with the requirement of SCD] activity for neutral lipid storage, as oleic acid is the preferred substrate for TAG storage (21). SCD2 expression in mice is primarily found in the brain, presumed to be important for myelin formation (11), and is required for lipid synthesis during early development (22). Human SCD2 is expressed at high levels in the brain and whole pancreas (19). SCD3 expression is restricted to skin sebaceous glands and is thought. to be involved in making wax esters for the skin (14, 23, 24). The mouse SCD4 isoform is expressed specifically in the heart (13). Although studies have identified direct roles for SOD] and SCD3 in lipid storage and wax ester synthesis, respectively, the significance of the SCD2 and SCD4 isoforms in lipid metabolism remains unclear. More specifically to the islet B-cell, the gene expression profile of SCDs and the contributions of specific SCD isoforms to normal [3- cell function have not been determined. 1.2.2. Elongation FA elongation by addition of a malonyl-CoA C2 unit to a fatty acyl-CoA is catalyzed in four steps: condensation of the fatty acyl-CoA with malonyl-CoA, reduction using NADPH, dehydration, and reduction to the fatty acyl-CoA product (Figure 1.1). The rate-limiting condensation step is catalyzed by various FA elongases, the activities of which are substrate dependent. Seven subtypes of FA elongases have been identified in mouse, rat, and human, and are referred to as e_longation 9f yery long chain fatty acids (Elo‘ chain barrr’e. mainl} PL‘FAs 34'). T11 level bei palmitole tissues (3 of oleic characteriz Tn Preparations C18 ML'FA mammalian r Elongases in i (Elovl-1 to 7) (www.cnsembl.org). Elovl-l, 3, and 4 elongate a broad range of very long chain FAs (>C20) and are involved in sphingolipid synthesis, brown adipose and skin barrier function, and retinal function, respectively (25-30). Elovl-2 and Elovl-5 are mainly involved in n-3 and n-6 PUFA synthesis, with Elovl—2 elongating C20 and C22 PUFAs and Elovl-5 elongating C18 PUFAs as well as palmitoleic acid (16:1,n-7) (31- 34). The expression of Elovl-2 and Elovl-5 was detected in most tissues with the highest level being in the liver (35). Elovl-6 is capable of elongating C12-16 saturated FAs and palmitoleic acid (16:1,n-7) (31, 32). Although the expression of Elovl-6 is low is most tissues (35), its activity significantly regulates stearic acid (18:0) synthesis, the precursor of oleic acid (18:1,n-9) (36). Elovl-7 substrates and expression have not been characterized. In regards to MUFA synthesis, FA elongation assays using microsomal preparations demonstrated Elovl-5 and Elovl-6 to elongate C16 FAs for the generation C 18 MUFAs. The roles of these genes in the de nova synthesis of MUFAs in intact mammalian cells, however, have not been addressed. In addition, a unique role for FA elongases in B-cells is not known. ~ww NADH 4 NAD NADH:C b5 Re NADH + H)£’ (Fe 2+8)“ Stearoyl- -CoA + 02 NAD“ Fe3+ Fe3+ (HEX Oleoyl-CoA + H20 NADH: Cytochrome Cytochrome b5 Stearoyl-CoA b5 Reductase Desaturase Fatty acyl-CoA ml w' a... 3-Ketoacyl-CoA —1m 3-Hydroxyacly-COA _i D...yd...... Trans-2, 3-enoyl-COA TER Elongated Fatty acyl-CoA Figure 1.1. Reaction diagrams for FA desaturation through the stearoyl-CoA desaturase complex and general FA elongation. Microsomal FA desaturation occurs through the transfer of two electrons from NADH to oxygen to produce a desaturated F A- CoA and water. Microsomal FA elongation occurs through the following sequential reactions: 1) FA-CoA condensation with malonyl-CoA to form 3-ketoacyl—COA, 2) 3- ketoacyl-CoA reduction to 3-hydroxyacyl-COA using NADPH, 3) 3-hydroxyacyl-COA dehydration to trans-2,3-enoyl-COA, 4) trans-2,3-enoyl-COA reduction to acyl-CoA. KAR, 3-ketoacyl-COA reductase. TER, trans-2,3-enoyl-C0A reductase. 13.Ex038"° A ma. derived from ‘ FAs (C-g- 31b complexes cor cholesterol. 3. respectively. after digesting . lumen of the in and 2-monoacy glycerol and FF the}: are used 11 within lipoprotei both exogenous lipcproteins, par LDL), are cleav e tissues (44). FFF TAG and cholestc theliver(45). Sui Either facilitated ' membrane. Dietary F linolenic acid ( 13 1.3. Exogenous Fatty Acid Uptake and Essential Fatty Acids A major source of intracellular FAs comes from transport of exogenous FA derived from circulating chylomicrons, lipoproteins, free FAs (FFAs), and protein bound FAs (e.g. albumin). ChylomiCrons and lipoproteins consist of lipid droplet/protein complexes composed of triacylglycerol (TAG), cholesterol esters, glycerophospholipids, cholesterol, and apolipoproteins, and are secreted from the intestine and liver, respectively. FAs incorporated into chylomicrons primarily originate from dietary fat afier digesting a meal. TAG, the predominant source of dietary FA (37), is cleaved in the lumen of the intestine by pancreatic lipase at the sn-1 and sn-3 positions to produce FF As and 2-monoacylglcyerol (2-MAG) (3 8-40), the latter of which can be further cleaved to glycerol and FFA (41). MAG and FFAs are then transported inside enterocytes where they are used to reassemble TAG for chylomicron formation (42, 43). Lipid droplets within lipoproteins that are secreted from the liver contain a mixture of FAs derived from both exogenous and de nova (see next section) sources. Circulating chylomicrons and lipoproteins, particularly very low density and low density lipoproteins (VLDL and LDL), are cleaved by lipoprotein lipase to release FFAs for storage and use in peripheral tissues (44). FFAs are also released into the circulation during fasting by activation of TAG and cholesterol ester hydrolysis in lipid storage depots, such as adipose tissue and the liver (45). Subsequent uptake of exogenous F FAs at peripheral tissues occurs through either facilitated transport by FA transport proteins or by diffusion of the plasma membrane. Dietary FAs are the source of the essential FAs linoleic acid (18:2,n-6) and ' linolenic acid (18:3,n-3). Essential FAs are defined as essential because animals lack the 312D and A151) linoleic acid and docosahexeanoic ssnthesis of ar elongation activ from 18:3.n-3 a peroxisomal B-o 1.4. De .\'ovo F: De navo FAs that are n‘ mainly from the COA and malon in and out of t moon of ti mitochondrial l mitochondria, ) into “Fill-Co; for em)“ into i EDA carbox}’1a Fan}, acid syn elgngaliOn and PTOdUCe The $8 lllCleaSeg d8 ’70 A12D and A15D enzymes required for the synthesis of n-6 and n-3 FAs. Conversion of linoleic acid and linolenic acid to other PUFAs, such as arachidonic acid (20:4,n-6) and docosahexeanoic acid (22:6,n-3), is important for normal cellular function. Endogenous synthesis of arachidonic acid from 18:2,n—6 requires sequential desaturation and elongation activities of A6D, Elovl-5, and ASD, while docosahexeanoic acid synthesis from 18:3,n-3 additionally requires elongation by Elovl-2, desaturation by A6D, and peroxisomal B-oxidation (Figure 1.2) (7). 1.4. De Novo Fatty Acid Synthesis De nava synthesis of FAs characterizes the intracellular, cytosolic production of FAs that are made entirely fi'om intracellular acetyl-CoA and malonyl-CoA, derived mainly from the tricarboxylic acid cycle‘(TCA). The accumulation of cytosolic acetyl- CoA and malonyl-CoA is highly dependent upon glucose metabolism and the carbon flux in and out of the TCA cycle. Exposure of cells to elevated glucose levels increases transport of the glycolytic product pyruvate into the mitochondria, conversion of mitochondrial pyruvate to citrate via the TCA cycle, and export of citrate from the mitochondria. ATP-citrate lyase (ACL) then catalyzes the cleavage of cytosolic citrate into acetyl-CoA and oxaloacetate, the latter of which can be converted back to pyruvate for entry into the mitochondria. Carboxylation of ACL-derived acetyl-CoA via acetyl- CoA carboxylase (ACC) provides the necessary FA elongation substrate malonyl-CoA. Fatty acid synthase (FAS) performs all the enzymatic reactions necessary for FA elongation and subsequently utilizes one acetyl-CoA and seven malonyl-CoA units to produce the saturated FA palmitic acid (16:0) (46, 47). Together, elevated glucose increases de nova FA synthesis through elevated carbon flux through the TCA cycle and =Ii ‘ “wrawxr 4- *1 export Of C ll palmitate. De n through [we desaturation either Elovl- oleic acid (1 FA produce reducing SC (I) snthesis a: excessive lg nova FA 8}; (30:33-9) deSaturati 0r export of citrate, which can be sequentially converted through ACL, ACC and FAS to palmitate. De nova or exogenously derived palmitic acid can be used for MUFA synthesis through two pathways: Elovl-6 elongation to stearic acid (18:0) followed by SCD desaturation to oleic acid (18:1,n-9); SCD desaturation to palmioleic acid (16:1,n-7) and either Elovl-6 or Elovl-5 elongation to vaccenic acid (18:1,n-7) (Figure 1.2). Synthesis of oleic acid (18:1,n-9) has been demonstrated to be important for storing excess saturated FA produced after exposure to a high carbohydrate diet, an effect that is blocked by reducing SCD] gene expression (48). Thus, enhanced de nova FA synthesis drives the synthesis and storage of MUF As as a mechanism to prevent the accumulation of excessive levels of endogenous FFA. During conditions of essential FA deficiency, de nova FA synthesis also provides MUFAS as substrates for PUFA synthesis of mead acid (20:3,n—9) through sequential A6D desaturation, Elovl-l elongation, and ASD desaturation of oleic acid (Figure 1.2) (8). 10 -- 2 W "fur a‘o-J bun—w... Glucos 1 N6: N3 : E F Figure 1.2. deTlVed paln to stearic act 10 133111110191" 'Ssential FA prOCeSSed IC elongaSeS E deficiency, Elm-LL and Glycalysis A TP Citrate Lyase Acetyl CaA Carboxylase Fatty Acid Synthase I Elongase Glucose —-> 16 0 —> 18:0 Elovl6 A9D Essential fatty acid A9D S CD Deficiency & Obesity SCD .............. 13 1'19 —>:18: 2—>20: 2->2o: 31 Elongase A6D ' 5.3;]; " ' ' 351'; ' ' " 16:1n7 ———> 18:1n7 ElovlS/Elovl6 Diet __________________ 5”" k-—--—”'”'“"“--—‘-‘:.~g‘ N6: 18:2 +18:3->2o:3->2o:4zh 22:43.1 24:4—>24:5 ------- > 22:5 A6D Elovl5 A5D Elov12 Elovl2 A6D pp-ax N3: 18:3 —>rs:4->20.4—>20: 5—>22: 5:224: 5:224: 62:?22:6 ‘-- -“'"” ‘ ----------—--—‘—’— ” “‘ "’ ~ ‘ ‘--- -——’ -—----———--——— Essential Fatty Acids Figure 1.2. Synthesis of FAs de novo and from exogenous essential FAs. Glucose- derived palmitic acid (16:0) can be further modified by two pathways: Elovl—6 elongation to stearic acid (18:0) and SCD desaturation to oleic acid (18:1,n-9); or SCD desaturation to palmioleic acid (16:1,n-7) and Elovl-5/Elovl-6 elongation to vaccenic acid (18:1,n-7). Essential FAs from the diet, linoleic acid (18:2,n-6) and linolenic acid (18:3,n-3), are processed to long chain unsaturated FAs through the desaturases A6D and ASD, the elongases Elovl-5 and Elovl-2, and peroxisomal [ii-oxidation. During essential FA deficiency, de nova synthesized 18:1,n-9 is converted to mead acid (20:3,n-9) by A6D, Elovl-1, and ASD. Modified from Fatty Acid Regulation of Gene Transcription (8). 11 15. GlycerOI TAG siniheslS of phOSPhate’ a i first step, 3” SEmhaase. 2 ac) ltransl‘s’rasc 1.3). 5511111551 membranes. 35 (DA is transfer (AGPATSL 10C Dephogphorylal produces diacyl svnthesis of g1} addition of a thii The rate of de through both dire ofgenes involvec 1.5. Glycerol 3-Phosphate Pathway of Triacylglycerol Synthesis TAG serves as a storage depot for excess intracellular FFAs. In eukaryotes, synthesis of TAG de nova proceeds through stepwise FA acylation of glycerol 3- phosphate, a product of the glycolytic intermediate dihydroxyacetone phosphate. In the first step, an activated FFA, fatty acyl-CoA (FA-CoA) formed from an acyl-CoA synthetase, is esterified onto glycerol 3-phosphate by glycerol-3-phosphate acyltransferase (GPAT) at the sn-1 position to form lysophophatidic acid (LPA) (Figure 1.3). Synthesis of LPA occurs at the mitochondrial and endoplasmic reticulum (ER) membranes, as GPAT isoforms are found at both locations (49, 50). Next, another FA- CoA is transferred to LPA by a family of l-acylg1ycerol-3-phosphate acyltransferases (AGPATs), located in the ER membrane, to form phosphatidic acid (PA) (51, 52). Dephosphorylation of PA by a PA phosphatase (PAP) 1, also referred to as lipin, produces diacylglyceride (DAG) (53). Both PA and DAG can be utilized for the synthesis of glycerolphospholipids. The final reaction in TAG synthesis involves the addition of a third F A-CoA to DAG, catalyzed by a DAG acyltransferase (DGAT) (53). The rate of de nova TAG synthesis is highly affected by substrate availability and, through both direct and indirect mechanisms, substrate-induced transcriptional regulation of genes involved in FA synthesis and storage as described below. 12 i -s-‘ {:3 ngun: 1.3. is esteriiled acyltransfera or the ER 1 PhOSphaIe a dePhOSphOr}. FA'COA is e maciilgll'ceri Simi'lesis (Pi, Glycerol-3-phosphate Acyl-CoA Acyl-CoA GP AT Endoplasmic Mitochondria GPAT reticulum A l-C A LPA Cy ° AGPAT Endoplasmic reticulum PI, PG, CL 4—- PA PAP Endoplasmi creticulum Pi PC, PE, PS <——- DAG Acyl-CoA DGAT TAG Figure 1.3. Glycerol-3 phosphate pathway for triacylglyceride synthesis. A F A-CoA is esterified to glycolysis-derived glycerol-3 phosphate by glycerol-3 phosphate acyltransferase (GPAT) to form lysophosphatidic acid (LPA) at either the mitochondrial or the ER membrane. A second FA-CoA is esterified to LPA by acylglycerol-3 phosphate acyltransferase (AGPAT) to form phosphatidic acid (PA). PA is then dephosphorylated to diacylglycerol (DAG) by PA phosphatase (PAP). Finally, a third FA-CoA is esterified to DAG by diacylglycerol acyltransferase (DGAT) to form the triacylglyceride (TAG). PA and DAG can also be directly used for phospholipid synthesis (PI, PG, CL, PC, PE, and PS). 13 1.6. TranSC 1.6.1. C arb Glue glycolysis. shunt. F lux activator of activate the basic helix Binding of ( 0f genes our genes involv [0 Contain ( GPAT (th1 that glucose I the ChREBP LPK. and Ml of TC A Cycle of citrate and Storage of FA 31112 thPAT 1.6. Transcriptional Regulators of Lipogenesis 1.6.1. Carbohydrate Response Element Binding Protein Glucose metabolism is initiated through uptake, phosphorylation, and subsequent glycolysis. In addition, glycolytic intermediates can enter the hexose monophosphate shunt. Flux through the hexose monophosphate shunt produces xyulose 5-phosphate, an activator of protein phosphatase 2A that has been proposed to dephosphorylate and activate the carbohydrate response element binding protein (ChREBP) a member of the basic helix loop helix leucine zipper (bHLH/leu zip) transcription factor family (54). Binding of ChREBP and its heteromeric partner Max-like protein X (Mlx) to promoters of genes containing carbohydrate response elements (ChREs) activates transcription of genes involved in glucose metabolism and de nova FA synthesis (55, 56). Genes known to contain ChREs include L-pyruvate kinase (LPK), ACC, FAS, and mitochondrial GPAT (thPAT) (57-59). Studies in liver using a dominant negative Mlx demonstrated that glucose transporter 2 (GLUT2), malic enzyme (ME), and SCDl are also regulated by the ChREBP/Mlx complex (55). Enhanced pyruvate production by increased GLUT2, LPK, and ME expression increases pyruvate entry into the mitochondria and generation of TCA cycle intermediates. Accumulation of TCA cycle intermediates favors the efflux of citrate and conversion to acetyl-CoA by ACL that is driven towards synthesis and storage of FAS via increased ChREBP/Mlx-mediated expression of ACC, FAS, SCDl, and thPAT. l4 1.6.2. 5“” Sic transcript}.C conditiOnS‘ with the Si SREBP ins bHLH'kU .' tranSP'Oned include SRE Eenes im01 " studies” am intracellular 3 SREBP‘IC Pl protein abund the SREBP SC is still unclea: glucose and 5}" ACC, FAS, Eh. increases the r: shuttling of TC «5 manner, intake a. (lien glucose to insulin secreted fr 1.6.2. Sterol Regulatory Element Binding Protein-1c Sterol regulatory element binding proteins (SREBPs) are bHLH/leu zip transcription factors and key regulators of lipid synthesis (60). Under non-stimulated conditions, inactive and unprocessed SREBPs reside in the ER membrane and interact with the SREBP cleavage activating protein (SCAP) and Insig-2 (61-63). Activation of SREBP involves translocation of the SREBP/SCAP complex to the Golgi where the bHLH/leu zip domain is cleaved by site 1 and site 2 proteases and subsequently transported into the nucleus to initiate gene transcription (64, 65). Isoforms of SREBP include SREBP-1a, -1c, and -2. SREBP-1c is a major activator of the expression of genes involved in FA synthesis, whereas SREBP-2 induces genes involved in cholesterol synthesis and SREBP-1a activates genes for both FA and cholesterol synthesis (60). Low intracellular sterol levels initiate processing of SREBP-1a and -2 but not -lc (66, 67). SREBP-1c processing is initiated by insulin through decreased gene expression and protein abundance of Insig-2 (68), which releases its interaction with SCAP and allows the SREBP/SCAP complex to translocate to the Golgi (62, 63). Although the mechanism is still unclear, insulin-mediated SREBP-1c processing increases the metabolism of glucose and synthesis of FAs through binding to promoters of glucokinase (GK), ACL, ACC, FAS, Elovl-6, SCD l and 2, and thPAT (59, 69-76). Elevated GK expression increases the rate-limiting step of glycolysis, while the latter enzymes increase the shuttling of TCA cycle intermediates into FAs that are stored in complex lipids. In this manner, intake of dietary carbohydrates, specifically simple sugars, activates ChREBP to divert glucose to storage as FAS, and this process is enhanced through SREBP-lo by insulin secreted from glucose-stimulated pancreatic B-cells. 15 ‘V 1.6.3. Liver .\' Choic’S' and i2 membc LXRs are acti‘ LXR “35 Mei isoform is Will vvhereas LXRT; receptor (RXR transcription 01‘ rate-limiting en. ABCGI, plasmé of genes involve ChREBP and S synthetic agoni: atherosclerotic p' liter, hovvever, c. 10“ density lipOp induced SREBP- have reduced ex illustrates that L“ 1.6.3. Liver X Receptors Cholesterol and lipid homeostasis are also regulated by liver X receptors (LXR) or and [3, members of the nuclear hormone receptor superfamily of transcription factors. LXRs are activated by intracellular oxysterols (cholesterol intermediates), and recently, LXR was identified to directly bind and be activated by glucose (77). The LXRor isoform is highly expressed in the liver, intestine, adipose tissues, and macrophages, whereas LXRB is ubiquitously expressed (78). LXR heterodimerizes with its retinoid X receptor (RXR) partner, which then bind LXR response elements and activate transcription of genes involved in cholesterol catabolism and efflux such as CYP7A1, the rate-limiting enzyme for bile acid synthesis, and ATP-binding cassette (ABC) A1 and ABCGl, plasma membrane transporters involved in sterol transport (70, 79). Promoters of genes involved in de nova FA synthesis that contain LXR response elements include ChREBP and SREBP-10 as well as FAS, SCD], SCD2, and others (80-84). The synthetic agonist T0901317 also activates LXRs, and its use in mice reduced atherosclerotic plaques caused by cholesterol accumulation (82). LXR activation in the liver, however, caused a significant increase in TAG accumulation and secretion of very low density lipoproteins (85). In addition, LXR was shown to be required for insulin- induced SREBP-1c transcription (86), and LXR or/B -/- mice fed a high carbohydrate diet have reduced expression of some genes involved in FA synthesis (87). Together this illustrates that LXR is a key mediator of de nova synthesis and storage of FAs. 16 “ u',‘ j. . o 9 _. t 1‘- O T. .9. 9': E" 2”; €' . ‘1 D ,D . . ..- .~". T'- I C . 2 . O.‘ .a, :1». *5 3. ..,... .1' V E '. O’ (-2. .v V a n, .-..‘ I 11:: .,. 1‘! .‘g ¢ , r. 2.: t 3, 4- .‘ A14 ' .0. ‘4‘" mks-l, 4 N O ' i; . -‘ IrWerfigm ,y.,..,,g, Glu Insulins LX Figure 1.4, illSlliln. Dum Slum (Harp) a S.l'ltthesis of Glucose GKi Glucose 6-phosphate glycolysis L-PK pyruvate HMP m TCA cycle Glucose —>ChREBP citrate Insulin ACL - ace l-CoA t W LXR ACC malonyl-CoA \ FAS \ 16:0 Elovl-61 . SCD1I2 1621/1821 TAG Figure 1.4. Transcriptional regulation of lipogenic gene expression by glucose and insulin. During conditions of elevated glucose, flux through the hexose monophosphate shunt (HMP) activates ChREBP to induce L-type pyruvate kinase (L-PK) and drive the synthesis of pyruvate through glycolysis. Insulin secreted into the circulation from glucose-stimulated B-cells increases SREBP-1c processing, which can induce glucokinase to enhance glucose metabolism. Both ChREBP and SREBP-1c activation lead to the induction of genes involves in de nova synthesis and storage of FAs. LXRs can also bind and be activated by glucose and subsequently stimulate lipogenesis idirectly through ChREBP and SREBP-1c and directly through F AS and SCDl/2. 1.7. F3“? ‘1 In H through 5’0 from 21 FA-‘ utilized b). I OH Camlilnc camlllne fur the F,\-cami [0 Ver} long- medium I0 W transported to During decrease. \Vhit‘ cycle generatio cellular energy phosphorylatior reducing cytosol dependenl on car lo FA-carniline b—O» Huibited bv ma’ relieve CPT-l in ATP. 1.7. Fatty Acid Oxidation In mammals, mitochondria and peroxisomes carry out chain shortening of F As through B-oxidation. This process involves the multi-step enzymatic removal of C2 units from a F A-CoA to produce a shortened F A-CoA and acetyl-CoA, the latter of which is utilized by mitochondria to generate ATP. Mitochondrial B-oxidation of FA is dependent on camitine palmitoyltransferase-l (CPT-1)—mediated conversion of FA-CoAs to FA- camitine for transport into the mitochondria. Inside the inner mitochondrial membrane, the FA-carnitine is converted back to FA-CoA by CPT-2, and B-oxidation reduces short to very long-chain FA-CoAs completely to acetyl-CoAs. Peroxisomes, however, reduce medium to very long-chain FA-CoAs to shorter chain FA-CoAs and acetyl-CoA that are transported to the mitochondria for further [Bi—oxidation (88). During periods of nutrient deprivation or fasting, TCA cycle intermediates decrease, which leads to the induction of FA oxidation. Reduced glycolytic and TCA cycle generation of ATP causes an increase in the AMP/ATP ratio and activation of the cellular energy sensor AMP activated protein kinase (AMPK) (89, 90). AMPK-mediated phosphorylation inhibits ACC and activates malonyl-COA decarboxylase, effectively reducing cytosolic levels of malonyl-COA (89, 90). Mitochondrial B—oxidation of FA is dependent on carnitine palmitoyltransferase-l (CPT-l)-mediated conversion of FA-CoAs to FA-carnitine for transport into the mitochondria. CPT-l activity is allosterically inhibited by malonyl-CoA (91). Thus, reduced TCA cycle flux and AMPK activation relieve CPT-l inhibition by malonyl-CoA to allow for FA oxidation and the generation of ATP. 18 Ge proliferato PPARa id. regulates 3 glucose reg intracellular mitochondri Wifli RXR It may compel. Genes involved in peroxisomal B—oxidation are induced by the peroxisome proliferator activated receptor-a (PPARa) transcription factor. Natural ligands of PPARa identified thus far include FFAs, FA-CoAs and glucose (92, 93). PPARa also regulates some genes participating in mitochondrial B-oxidation (94). Together, direct glucose regulation of PPARa could possibly serve to prevent accumulation of excess intracellular FFAs during exposure to elevated levels of carbohydrates by elevating mitochondrial and peroxisomal FA oxidation. Similar to LXR, PPARa heterodimerizes with RXR to initiate transcription, suggesting that activation of these opposing pathways may compete for RXR. 19 2. Pancr 2.1. 519‘“ P2 secreting- of insulin gl}‘CO§€n ‘ metabolism lasting UP I 97). It is u dependent Uj through gb'c phosphon'lati potassium (K- activation of ‘ indirectly trigg cells in respons acids, amino 3C1 granule exocnos are less understr ofFA metabolis GSIS. (99, m3. 2. Pancreatic B-Cell Insulin Secretion and Role of Fatty Acid Metabolism 2.1. Mechanism of B-Cell Insulin Secretion Pancreatic B-cells respond to a rise in circulating blood glucose levels by secreting insulin, such as after digesting a carbohydrate-containing meal. The secretion of insulin into the circulation, in turn, signals peripheral tissues to store glucose as glycogen or lipid. Glucose-stimulated insulin secretion (GSIS) requires uptake and metabolism of the sugar and occurs in two phases; an initial first-phase of insulin release lasting up to ten minutes and an amplification second-phase that begins thereafter (95- 97). It is well accepted that the first-phase and initiation of the second-phase of GSIS is dependent upon an increased ratio of ATP/ADP, which results from the flux of glucose through glycolysis, the tricarboxylic acid (TCA) cycle, and mitochondrial oxidative phosphorylation (95, 97). The increased ATP/ADP ratio inhibits ATP-sensitive potassium (K+ATP) channel opening and causes depolarization of the plasma membrane, activation of voltage-dependent Ca2+ channels, and influx of Ca2+ that directly and indirectly triggers insulin release (Fig) (95-98). Stimulation of insulin secretion from [3- cells in response to glucose can be potentiated by other stimulatory effectors (e.g. fatty acids, amino acids, and incretin hormones) that generate secondary messengers to signal granule exocytosis (99-102). Mechanisms involved in the amplification phase of GSIS are less understood. Evidence has emerged, however, for glucose-stimulated regulation of FA metabolism and the generation of lipid signaling molecules in the mechanism of GSIS. (99, 103, 104). 20 iGluc ATP ~Sensit K’ a Channel Figure 1.5. m glucose-stimula Pancreatic fi-cel lhl'ough glycoly; me Of ATP to 153$ to depolar glanules are felt gTEmUIe exOCYIO 1‘ GIUcose ATP -Sensitive K+ o ...... 2+ .............. .Ca \ .. . KinaSes Figure 1.5. Mechanism of the ATP-sensitive K+ channel-dependent pathway of glucose-stimulated insulin secretion (GSIS). Exogenous gluocose is taken up into pancreatic B-cells through a glucose transporter (Glut2). Next, metabolism of glucose through glycolysis and the mitochondrial (Mito.) TCA cycle generates ATP and alters the ratio of ATP to ADP. The ATP-sensitive K+ channel is then inhibited by ATP, which leads to depolarization of the plasma membrane and an influx of Ca2+. Finally, insulin granules are released by both direct and indirect actions of Ca2+ on proteins involved in granule exocytosis. 21 2.2. RegU'atiO The P3 glucose over a high Km of b minimal actixi production of l negligible lact mitochondrial elei'ated gluco: ACC-mediated ”0). In regard: glycerol 3-phos backbone of gly- in B-cells coincir cells elevated gli 35 ChREBP and f SREBP-l (114,115). on ACC. FAS, Elm bl SREBP-1c ox' although both L Will-red for ma? incleaSed the EXY ‘ l 2.2. Regulation of Fatty Acid Metabolism in Pancreatic B-cells The pancreatic B-cell is innately designed to sense and efficiently metabolize glucose over a wide range of physiological concentrations (3-20 mM), due in part to the high Km of both GLUT2 (17 mM) and GK (8 mM) (103). As glucose levels rise, minimal activity through the hexose monophosphate shunt aids in driving the glycolytic production of pyruvate (105). Together with low pyruvate conversion to lactate, due to negligible lactate dehydrogenase (105), pyruvate in the [ES-cell is directed towards mitochondrial TCA cycling. Studies demonstrate that stimulation of B-cells with elevated glucose subsequently increases mitochondrial efflux of TCA intermediates, ACC-mediated generation of malonyl-CoA, and the synthesis and storage of FAs (106- 110). In regards to [ii-cell FA storage, a considerable portion of glucose is metabolized to glycerol 3-phosphate, from glycolytic dihydroxyacetone phosphate, for use in the backbone of glycerolipids (99). As in the liver, glucose-stimulated de novo FA synthesis in B-cells coincides with increased expression of L-PK, ACC, and FAS (110-113). In [3- cells elevated glucose has been shown to induce the binding of ChREBP to L-PK as well as ChREBP and SREBP-1c binding to FAS (111, 112). SREBP-1c activity is essential for glucose induction of lipogenic genes in B-cells (114, 115). Over-expression of SREBP-1c in a B-cell line induced the expression of ACC, FAS, Elovl-6, SCD], and SCD2 (114), and in rat islets, increased ACC and FAS by SREBP-1c over-expression correlated with elevated TAG synthesis (116). In B-cells, although both LXRor and [3 are expressed (5), LXRB is the predominant isoform and is required for maintenance of B-cell function (117). Pharmacological activation of LXRs increased the expression of ABCAl, involved in cholesterol efflux, as well as SREBP-1c, 22 GK. ACC elexated 5’ 3:11.111}! 0‘ expression Sim regulation b CPT-l and 1 oxidation thi augmented if (130). Anolh reduction of oxidative pho which leads to (121-123). El- protein expressi possibly througl 01 B-cells to ex thereby blocking Overall, directing excess ms 10 dean; rel ' ~ ' 3110115}le beta Bulls. GK, ACC, and F AS, resulting in intracellular TAG accumulation (5, 118). In addition, elevated glucose was shown to increase LXRor translocation into the nucleus (119), the activity of which could mediate the glucose-induced increase in ChREBP gene expression observed in B-cells. Similar to FA synthesis, FA oxidation in B-cells is subject to multiple levels of regulation by glucose. Of particular importance is the interaction of malonyl-CoA with CPT-1 and inhibition of mitochondrial FA uptake. Although high glucose reduces FA oxidation through the generation of malonyl-COA, FA oxidation can be significantly augmented by over-expression of CPT-1 during exposure to high glucose conditions (120). Another mechanism for inhibiting FA oxidation is through generation of ATP, or reduction of the AMP/ATP ratio through enhanced glycolysis and mitochondrial oxidative phosphorylation. This is associated with decreased B-cell AMPK activity, which leads to reduced ACC phosphorylation and increased ACC-derived malonyl-CoA (121-123). Elevated glucose also causes a significant decrease in PPARor gene and protein expression as well as promoter binding to genes involved in FA oxidation (124), possibly through the decrease in AMPK activity (125). In contrast to glucose, exposure of B-cells to exogenous FAS increases CPT-1 and decreases ACC gene expression, thereby blocking glucose-stimulated repression of mitochondrial FA oxidation. Overall, these mechanisms allow the pancreatic B-cell to function properly by directing excess exogenous carbon in the form of glucose into FAs for storage or excess FFAs to degradation via FA B-oxidation. The following sections describe the relationship between the regulation of FA metabolism and the mechanism of GSIS from [ES-cells. 23 2.3. Mitocho Enhar pyruvate has dehydrogenas oxaloacetate ( initiating the : ending with c describes the r are fully repler glucose-stimul. OfPl‘TUVate to intennediates g secretion, inclu. primary produc Sl‘mllesis of p.“ mala‘e‘Pb’rux'atc oxaloacetate-p},r derived from ( intermediate, 1: DINADPH COr‘ identified to be 35 l0 which p\ 2.3. Mitochondrial Anaplerosis/Cataplerosis Enhanced glucose flux through glycolysis generates pyruvate. Mitochondrial pyruvate has two options for entry into the TCA cycle: conversion by pyruvate dehydrogenase (PDH) to acetyl-CoA or conversion by pyruvate carboxylase (PC) to oxaloacetate (105, 126-128). PDH and PC activities provide the necessary substrates for initiating the sequential synthesis of TCA cycle intermediates beginning with citrate and ending with conversion of malate to oxaloacetate to reinitiate the cycle. Anaplerosis describes the replenishment of TCA cycle intermediates. After TCA cycle intermediates are fully replenished, exit of carbons from the cycle is defined as cataplerosis. In B-cells, glucose-stimulated anaplerosis via PC activity is high, metabolizing approximately half of pyruvate to oxaloacetate (105, 126-128). This anaplerosis/cataplerosis of TCA cycle intermediates generates second messengers thought to be involved in signaling for insulin secretion, including the cataplerotic products NADPH, malonyl-CoA and FA (103). The primary production of NADPH via anaplerosis/cataplerosis is pyruvate cycling, the re- synthesis of pyruvate fi'om cycle intermediates. Pyruvate cycling processes include the malate-pyruvate, citrate-pyruvate, isocitrate/alpha-ketoglutarate-pyruvate, and oxaloacetate-pyruvate shuttles (127, 129-135). Studies using [U-13C]-g1ucose supported this idea showing that two pyruvate “pools” exist in B-cells (127, 132). One pool is derived from glycolytic pyruvate and another is synthesized from a TCA cycle intermediate. It is hypothesized that NADPH serves as a second messenger as the level of NADPH correlates with GSIS, and gluctaredoxin—l, an NADPH target, was recently identified to be involved in GSIS (130, 136, 137). At this time, however, it is still unclear as to which pyruvate cycling process is most important. Nevertheless, it is clear that 24 .Eflucose'sun1L (100,106' I: 2.4. Gen era“ 2.4.1. R0“ 0' GluC'0 citrate. 3) con [M from 3“) of malonl'l'CC FAS and lead: molecules. 9 generation Of T by over-exp”: reduces GSIS 1 increases and formation of e accumulate “ht 140,141). ln s cytosolic malon estenfication, . physiologicalh COA plays an 1’ FA oxidation, participate in in glucose—stimulated cataplerosis activity is high as it drives synthesis of malonyl-CoA (100, 106, 129, 138), the precursor for de novo FA synthesis. 2.4. Generation of Lipid Signaling Molecules 2.4.1. Role of Malonyl—CoA Glucose-stimulated anaplerosis/cataplerosis increases 1) mitochondrial efflux of citrate, 2) conversion of citrate to acetyl-CoA by ACL, and 3) the generation of malonyl- CoA fiom acetyl-CoA by ACC. The role of malonyl-CoA in GSIS is based on the ability of malonyl-CoA to inhibit CPT-1 activity (91). This blocks mitochondrial Iii-oxidation of FAS and leads to the accumulation of FFAs, FA-CoAs, DAG and other lipid signaling molecules. ACC gene expression is naturally high in B-cells, allowing for rapid generation of malonyl-CoA prior to GSIS (106-108, 113, 139). Increasing FA oxidation by over-expression of CPT-1 or knockdown of ACC gene expression significantly reduces GSIS (120, 140, 141). Additionally, knockdown and over-expression of PPARa increases and decreases GSIS, respectively (142, 143). Thus, GSIS involves the formation of endogenous FAs and lipid signaling molecules, which are allowed to accumulate when malonyl-COA levels are sufficient to block FA oxidation (100, 138, 140, 141). In support of this concept, reducing malonyl-CoA levels by over-expressing cytosolic malonyl-COA decarboxylase (MCDc) increased FA oxidation, decreased FA esterification, and reduced GSIS in the presence of endogenous FFA, a more physiologically relevant state (100). The above studies show that synthesis of malonyl- CoA plays an important role in GSIS by blocking the elimination of FA-CoAs through FA oxidation, resulting in the accumulation of lipid signaling molecules that can participate in insulin secretion. 25 2.4.2. Fret lnh accumulall and DAG. syndiesis. and release number of i lntr: synthesized lipids, prodi elevated glu. molecules as GSIS (145.). 1145, 146) an bl'FFA on G.‘ stores Via the honey“ dem. enhancement ( endogenous lip 2.4.2. Free Fatty Acid, Long Chain Fatty Acyl-CoA, and Diacylglycerol Inhibition of FA oxidation by malonyl-CoA permits pancreatic B-cell accumulation of lipid signaling molecules, such as FFA, long-chain FA-CoA (LC-CoA), and DAG. Exocytosis of insulin granules is a complex process involving granule synthesis, transport to the plasma membrane, docking, priming, and membrane fusion and release of insulin (144). FFA, LC-CoA, and DAG enhance insulin release through a number of mechanisms. Intracellular FFA and LC-CoA levels are tightly regulated. Although de novo synthesized FAS can be rapidly converted to LC-CoA for incorporation into complex lipids, production of FFAs themselves could affect insulin release (99). Exposure to elevated glucose and exogenous FFA increases synthesis of intracellular lipid signaling molecules as well as activates islet B-cell G protein-coupled receptors, which enhances GSIS (145). GPR40 (or FFAl receptor) is highly expressed in rodent and human islets (145, 146) and was identified to bind exogenous FFAs. The effect of GPR40 activation by FFA on GSIS involves increased intracellular Ca2+ levels through release of ER Ca2+ stores via the Gorq-phospholipase C pathway (147). Knockdown of GPR40 in mice, however, demonstrated that its activation accounts for only half of the exogenous F FA enhancement of GSIS, supporting the role of FFA-induced increases in additional endogenous lipid signaling molecules (148). In addition to circulating FFA, B-cells have been shown to release FFA when treated with elevated glucose (149). In this way, FFA- mediated GPR40 activation could be a possible mechanism for amplification of GSIS by endogenously synthesized FA. 26 El€\ 3' Proteins dires for membram with the pi gnapwgamm (152). AS dl‘ amplifi'lng the of B-cells al: phospholipids DAG i glucose (108. GSIS through include proteir guanyl nucleol INCH proteir phosphorylatior both Ca2+-dept cells have addi (155-157). Rt. been Presented existence of c lll‘t“ detenmnav Specific PKC Elevated synthesis of LC-CoAs enhance GSIS by increasing the acylation of proteins directly involved in insulin granule exocytosis. Protein acylation is necessary for membrane targeting of specific proteins known to be involved in fusion of granules with the plasma membrane, such as synaptosomal-associated protein-25 and synaptogamin (150, 151). LC-CoAs were further shown to increase islet lipase activity (152). As discussed in the next section, this could provide an additional mechanism for amplifying the production of lipid signaling molecules. Additionally, glucose stimulation of B-cells also alters the specific species of LC-CoAs that are incorporated into phospholipids (110), which could cause membrane remodeling and effect granule fusion. DAG is also rapidly synthesized de novo in B-cells by acute exposure to elevated glucose (108, 110, 153). Generation of DAG by elevated glucose has been implicated in GSIS through activation of various DAG binding proteins. DAG binding proteins include protein kinase C isoforms (PKCs), protein kinase D (PKCu), chimaerins, Ras guanyl nucleotide-releasing proteins, mammalian homolog of caenorhabditis elegans UNC13 protein (Munc-13s), and DAG kinases (154). In B-cells, glucose stimulates the phosphorylation of many proteins, in part, through PKC (155), which occurs through both Ca2+-dependent (classical) and —independent (novel) PKC isoforms. Studies in B- cells have additionally demonstrated that glucose promotes the translocation of PKCs (155-157). Roles both for and against PKC activation in the mechanism of GSIS have been presented. This is in part due to the lack of isoform specific PKC inhibitors and the existence of compensatory mechanisms regulating protein phosphorylation that hinder the determination of which PKCs are directly involved in GSIS (158). Thus, a role for specific PKC isoforms in GSIS cannot be withdrawn. Enhanced activity of Munc-l3 27 potentiates insulin seer the role Mu 2.5. Lipolys In at from lipol} hydrolytic r glycerolipid glycerol-3-pl gene express. index of lipa glucose have activity of 3 enhanced lipo llPOIysis coulc click? capable I the fact that glt. llpases capable 2'5'1' Neutral 25".]. HUI-n“: potentiates normal GSIS in vitro, and islets from mice lacking Munc-13 had reduced insulin secretion and abnormal glucose tolerance (159, 160). This is in agreement with the role Munc-l3 binding to DAG and being involved in synaptic granule priming (161). 2.5. Lipolysis and Glycerolipid/Fatty Acid Cycling In addition to de novo synthesis, lipid-signaling molecules can also be generated from lipolysis of glycerolipids. Lipolysis describes the activity of lipase-catalyzed hydrolytic removal of FAS from a range of complex lipids. The final products of glycerolipid lipolysis are FF As and glycerol, the latter of which cannot be reactivated to glycerol-3-phosphate for lipid synthesis in B-cells due to the absence of glycerol kinase gene expression (162). Thus, measurement of extracellular glycerol content is used as an index of lipase activity. Interestingly, rat islets and B-cells stimulated with elevated glucose have increased glycerol release (99, 163, 164), which correlates with increased activity of a number of lipases (described below) (164-167). The combination of enhanced lipolysis and increased synthesis of glycerolipids suggested FFAs released by lipolysis could be rapidly reincorporated into glycerolipids, creating a glycerolipid/FA cycle capable of enhancing the generation of lipid signaling molecules (99). In light of the fact that glucose modulates lipase activity, a number of studies have identified various lipases capable of contributing to GSIS. 2.5.1. Neutral Glycerolipid Lipolysis 2.5.1.1. Hormone-Sensitive Lipase Glycerolipid/FA cycling in B-cells has emerged as a new metabolic pathway possibly involved in GSIS‘(99). Although it remains to be determined as to which pool of glycerolipids is most important, studies have drawn attention to neutral glycerolipids, 28 Specifically actiVlU' inC adiponumn‘ was the {IN (174, 175)- mice general responses (I ‘ mice. demon Additionall}'~ TAG (168, l' DAG was 001 maybe througl (178,179). In with a fl-Cell 5 phase of insul i (1781. Taken 1 exocytosis. but 2.5.1.2. Adipos Enzyma showed that th- (168,170, 177 lhan adiponutrgi Regulation of specifically TAG and cholesterol ester (CE). Lipases known to have TAG lipolysis activity include hormone-sensitive lipase (HSL), adipose tissue TAG lipase (ATGL), . adiponutrin, G82, and carboxylesterase 3, also termed TAG hydrolase (168-173). HSL was the first TAG lipase identified to be expressed in B-cells and regulated by glucose (174, 175). A role for HSL in GSIS has been unclear as independent lines of HSL null mice generated on different genetic backgrounds exhibited inconsistent insulin secretory responses (163, 176). Lipolysis was still present and activated by glucose in HSL null mice, demonstrating the presence of other lipases involved in B-cell function (163). Additionally, HSL has a higher substrate preference for DAG, MAG, and CE than for TAG (168, 177). Studies found the hydrolysis of neutral cholesterol esters rather than DAG was completely blocked in HSL null mice and that its effect on insulin secretion maybe through directly regulating membrane cholesterol levels and granule exocytosis (178, 179). In fact, HSL was found localized on insulin granule membranes (180). Mice with a B-cell specific deletion of HSL were found to have significantly reduced first- phase of insulin release, thus showing that HSL has a direct role in insulin exocytosis (178). Taken together, these studies show that HSL may participate in insulin granule exocytosis, but HSL is not the key TAG lipase linked to GSIS. 2.5.1.2. Adipose Triacylglyceride Lipase Enzymatic characterization of ATGL, adiponutrin, GS2, and carboxylesterase 3 showed that these lipases have a higher activity for TAG than DAG compared to HSL (168, 170, 172, 173). Recently, ATGL gene expression was identified to be much higher than adiponutrin, GSZ, and HSL in both rat islets and the INS832/ 13 B-cell line (165). Regulation of ATGL gene expression was found to be reduced in islets of fed versus 29 “:1 fasted rats, and Il exposed to long-1 gene expression decreased insuli: hormone glucagt displayed increa: insulin secretion however, there \t glycerol release maintain lipolyt glycerol release TAG pools. F' regulated by I molecules that 2.5.2.1’h05ph Glycei Old? noro 3r Ca} influx dll‘cct and phOSPhOlipar PhosphateS parallels an SECl’eIiOn E fasted rats, and this correlated with reduced ATGL gene expression when B-cells were exposed to long-term elevated concentrations of glucose (165). Knockdown of ATGL gene expression in B-cells reduced TAG lipase activity, increased TAG content, and decreased insulin secretion in response to glucose, exogenous FA, and the incretin hormone glucagon-like peptide-1 (165). Similarly, isolated islets from ATGL null mice displayed increased TAG content and decreased glucose- and exogenous FA-stimulated insulin secretion (165). In both B-cells and mouse islets with reduced ATGL expression, however, there was no change in total glycerol release. The absence of a change in total glycerol release demonstrates a compensatory mechanism that could be activated to maintain lipolytic activity. This is at odds with ATGL having a major role in total glycerol release. The authors propose that B-cells contain fuel—insensitive and —sensitive TAG pools. Fuel-sensitive TAG pools localized close to insulin granules could then be regulated by ATGL and other TAG/FA cycling enzymes and provide lipid signaling molecules that participate in insulin secretion. 2.5.2. Phospholipases C and D Glycerophospholipids represent a considerable pool for glycerolipid/FA cycling of de novo and exogenously derived FAs for participation in GSIS. As described above, Ca2+ influx upon exposure to elevated glucose signals for insulin release through both direct and indirect mechanisms. Of these mechanisms, influx of Ca2+ activates phospholipase C (PLC), which cleaves phosphatidylinositol to generate inositol phosphates and DAG (166, 181). Insulin secretion from glucose-stimulated B-cells parallels an increase in the release of inositol phosphates (166), which can amplify insulin secretion by activating the release of Ca2+ from intracellular stores (182-184). 30 Compared 10 ml pronounced and This reduced SCC islets by phamtak studies it is apt phosphatidylinos Phosphol choline and pho phosphatidic aci mammals, PLD PLD2 was local trafficking, endt The release of l promoting curtl “891 Interest phOSPhOrylatior phOSPhatase or membrane fuS i knockdown of lllSllllll granUlg phOSphatidic '4 o . Compared to rat islets, the second—phase of insulin release in mouse islets is less pronounced and coincides with lower levels PLC expression and activity (166, 185). This reduced second-phase in mouse islets could be elevated to levels comparable to rat islets by pharmacological activation of PLC or DAG binding proteins (166). From these studies it is apparent that active release of inositol and DAG by PLC lipolysis of phosphatidylinositol has a role in GSIS. Phospholipase D (PLD) catalyzes the hydrolysis of phosphatidylcholine to choline and phosphatidic acid. Subsequent dephosphorylation of phosphatidic acid by phosphatidic acid phosphatase can also generate DAG. Two isoforms of PLD exist in mammals, PLDl was found in the Golgi apparatus and on intracellular vesicles, while PLD2 was localized primarily to the plasma membrane (186, 187). Intracellular vesicle trafficking, endocytosis, and exocytosis are associated with increased PLD activity (188). The release of phosphatidic acid derived from PLD likely aides in membrane fusion by promoting curvature of the membrane due to the small negatively charged head-group (189). Interestingly, PKC is able to activate PLD by direct interaction rather than phosphorylation (186). Thus, it is feasible that generation of DAG from phosphatic acid phosphatase or PLC could induce translocation of PKC to activate PLD and facilitate membrane filSIOD. In B-cells, GSIS was elevated and reduced by over-expression and knockdown of PLD], respectively (167). In addition, PLDl was partially localized to insulin granules (167). These findings suggest PLD may have two roles, one to provide phosphatidic acid for glycerolipid/FA cycling and another to directly effect insulin granule exocytosis. 31 2.5.3. Phor Lip or sn-Z (Pl also contril and human specific act 1AA) com pr rodent islet: inhibition 01 and in rim) amplified GS expression tc Whether iPLa 2.5.3. Phospholipase A2 Lipolysis of glycerophospholipids by phospholipase A at either the sn-1 (PLAl) or sn-2 (PLA2) position generates FFA and lysophospholipids, the latter of which can also contribute to glycerolipid/F A cycling and act in signal transduction (190). In rodent and human islets, a Ca2+-independent ATP-sensitive PLA2 (iPLAZB) was identified with specific activity towards arachidonic acid at the sn-2 position (191). Arachidonic acid (AA) comprises approximately thirty percent of the total glycerophospholipid FA mass in rodent islets (192), and free AA accumulates in islets stimulated with glucose (193). Inhibition of potassium channels by ATP is amplified by increased cytosolic AA (194), and in vitro studies showed knockdown and over-expression of iPLA28 reduced and amplified GSIS, respectively (195, 196). In mice, however, failure of iPLAZB over- expression to amplify GSIS and inconsistencies in iPLA2l3 knockdown mice question whether iPLA2B is involved in normal glucose homeostasis (190, 197, 198). 32 TCA cycle ATP Glucose FFA GIy3P Mal-CoA o 1i ti ATP NADPH LSMIDAG O. . O .0 O O... * ...O 4 ‘ Insulin secretion Figure 1.6. Proposed model for the role of FA metabolism in glucose-stimulated insulin secretion. Glucose stimulation drives the generation of pyruvate (Pyr), which enters the TCA cycle to modulate ATP production and undergoes pyruvate cycling to generate NADPH. Mitochondrial cataplerosis and pyruvate cycling increase the level of malonyl-CoA, which blocks FA oxidation. De novo and exogenous FFAs are converted to LC-CoA and esterified to glycerol 3-phosphate (Gly3P) to enter into the glycerolipid/F A cylc (GL/F A cycle). Finally, the combination of GL/F A cycle-mediated generation of lipid signaling molecules (LSM), such as DAG, increased NADPH, and increased ATP/ADP ratio signal the B-cell to secreteinsulin. 33 3, Glucose 3.1. Gluco Tit dysfunctior maintain g1 of insulin responses tl‘ cells fail Glucotoxicit cells from C1 periods of } diminished ( ‘0 as glucor aCCOmpanied contribute to referred to a dependent up COmbination mechanisms recognlzed [h 3. Glucose and Fatty Acid Mediated Pancreatic fi-Cell Dysfunction 3.1. Glucotoxicity vs. Glucolipotoxicity in Type 2 Diabetes Type 2 diabetes (T2D) is characterized by insulin resistance and pancreatic B-cell dysfunction, due to both genetic and environmental factors, that results in the inability to maintain glucose homeostasis. In the early stages of the pathogenesis of T2D, the onset of insulin resistance causes glucose levels to rise and triggers B-cell compensatory responses that include increased synthesis and secretion ‘of insulin. T2D ensues when [3- cells fail to secrete sufficient amounts of insulin to maintain norrnoglycemia. Glucotoxicity and glucolipotoxicity have been proposed to underlie this progression of B- cells from compensation towards dysfunction and eventually failure (3, 199). Sustained periods of hyperglycemia are associated with adverse effects on the B-cell such as diminished GSIS, decreased insulin gene expression, and apoptosis, collectively referred to as glucotoxicity (4, 199). In obesity-associated T2D, hyperglycemia is often accompanied by hyperlipidemia and elevated levels of plasma FFAs, which also contribute to B-cell dysfunction (200-202). Damage due to chronic elevations in FF As, referred to as lipotoxicity (203), was found to require FA esterification and to be dependent upon elevated glucose (3). This concept of B-cell dysfunction from the combination of glucose and FAs is defined as glucolipotoxicity (3). The major mechanisms accounting for the effects of both glucotoxicity and glucolipotoxicity recognized thus far are reviewed in the following sections. 34 3.2. Mechal 3.2.1. Endol Hint quantities of capacity of ‘ proteins (.204 of the unfold integrity. anc transmembra stranded R} transcription autophosphor box binding activates gem eXpOl't from t PERK and A' Chain binding membrane an Wing (21 1-; fem01(eIF) 2 Proteins SUch and resistanci cleared, and 3.2. Mechanisms of B-Cell Failure 3.2.1. Endoplasmic Reticulum Stress Hyperglycemia signals B-cells to continuously synthesize and secrete large quantities of insulin. Over time, the rate of insulin translation exceeds the protein folding capacity of the B-cell endoplasmic reticulum (ER), causing accumulation of unfolded proteins (204). If sustained, buildup of unfolded proteins causes ER stress and activation of the unfolded protein response (UPR). The UPR acts to reduce ER stress, preserve ER integrity, and prevent cell death. Transduction of this response is mediated by the ER transmembrane proteins inositol requiring ER-to-nucleus signal kinase (IRE) 1, double- stranded RNA-activated kinase (PKR)-like ER kinase (PERK), and activating transcription factor (ATP) 6 (204). BR stress induces IREl dimerization and autophosphorylation to gain endoribonuclease activity and splice the mRNA encoding X- box binding protein (Xbp) 1 (204). Spliced Xbpl (Xbpls) protein transcriptionally activates genes involved in expansion of the ER, protein maturation, protein folding and export from the ER, degradation of misfolded proteins, and lipid metabolism (205-210). PERK and ATF6 remain inactive in the ER by binding with the ER chaperone Ig heavy chain binding protein (BiP, also known as GRP78 and HSPAS) on the luminal side of membrane and are activated as BiP detaches from the membrane to assist in protein folding (211-214). PERK phosphorylates and inactivates eukaryotic translation initiation factor (eIF) 2a, leading to decreased translation of most proteins except some specific proteins such as ATF4 (215, 216). ATF4 induces genes important for amino acid import and resistance to oxidative stress (217). Activated ATF6 is translocated to the Golgi, cleaved, and transported into the nucleus to initiate transcription of ER chaperone genes 35 such as Eli) and ATF 6 p cell failure a Rat i can induce I increased B expression it controls (221 activates L'P palmitate sig ATF4 and A‘ oleate. Acm islets as Well exPressing th mICmSCOP)’ o such as BiP, enhancing ER protein folding capacity (218, 219). Together, IREI, PERK, and ATF6 pathways act to preserve ER function, which if not maintained can lead to B- cell failure and ultimately cell death (204). Rat islets cultured in elevated glucose demonstrated that glucotoxic conditions can induce the UPR, exhibiting activation of IREl (by increased Xbpls) and ATF6 (by increased BiP expression) (220). This correlates with increased Xbpls and BiP expression in T2D human islets cultured in elevated glucose compared to non-diabetic controls (221). Exposure of B-cells to elevated concentrations of exogenous FFAs also activates UPR pathways. Multiple B-cell line models treated with the saturated FA palmitate significantly induces Xbpls, eIF20t phosphorylation, and protein levels of ATF4 and ATF6 (222-224), whereas only minimal ER stress is elicited by the MUFA oleate, Activation of all three UPR pathways by palmitate was demonstrated in human islets as well (222). These effects in a B-cell line could be partially reversed by over- expressing the ER chaperone BiP (224). In addition to UPR pathways, electron microscopy of B-cells treated with palmitate and pancreatic sections from T2D patients observed alterations in B-cell ER integrity reflected as distention of the ER (221, 223). Thus, the inability to maintain proper ER function in the B-cell could be a significant risk factor for the development of T2D. 3.2.2. Oxidative Stress Pancreatic B-cells have low levels of antioxidant enzymes compared with other tissues (225, 226), rendering fi-cells particularly susceptible to oxidative stress. Elevated production of reactive oxygen species during chronic hyperglycemia is detrimental to B- cells (4). Pathways through which ROS can be produced from elevated glucose include 36 oxidative phosphorylation, glyceraldehyde autoxidation to methylglyoxal and glycation, a-ketoaldehyde formation and glycosylation, DAG activation of PKCs, hexosamine metabolism, and sorbitol metabolism (227). In B-cell lines and isolated islets, approaches to reduce ROS production by use of antioxidants and over-expression of antioxidant enzymes protected from chronically elevated glucose-induced decreases in insulin gene expression, transcription factor binding to the insulin gene promoter, and GSIS (4, 228, 229). This demonstrates B-cell firnction is significantly affected by chronic hyperglycemia-induced oxidative stress. The involvement of oxidative stress in FA- mediated B-cell dysfunction, however, remains unclear, as evidence both for and against a role of FAs in oxidative stress have been presented (230, 231). 3.2.3. Malonyl-CoA Inhibition of Fatty Acid Oxidation and Lipid Accumulation Generation of malonyl-CoA via ACC activation during short-term glucose exposure is essential to normal B-cell GSIS due to the ability of malonyl-CoA to interact with CPT-1 and inhibit FA oxidation. Chronic hyperglycemia, however, continuously drives malonyl-CoA production and subsequent synthesis and storage of FAs into TAG, which has been linked to diminished B-cell GSIS (199, 232). The effects of chronic hyperglycemia on de novo lipogenesis, TAG accumulation, and loss of B-cell function was found to correlate with elevated nuclear SREBP-1c in diabetic islets (233, 234). Over—expression of a constitutively active nuclear SREBP-1c in islets and B-cell lines also resulted in increased lipogenesis and reduced GSIS (116, 235). This suggested that sustained glucose-stimulated lipogenesis and TAG accumulation via SREBP-1c activation facilitated diminished GSIS. 37 SREBP-1c has also been implicated in glucolipotoxicity during B-cell exposure to elevated levels of exogenous FF As. Treatment of islets with elevated palmitate increased lipogenic gene expression and the nuclear form of SREBP-1c, increased TAG content, and decreased GSIS (236). These effects were prevented by treatment with eicosapentaenoate (20:5,n-3), which is known to reduce SREBP-1c processing (236). More direct evidence of a relationship between malonyl-CoA and FA oxidation comes from examination of palmitate-induced B-cell death. Elevating FA oxidation through AMPK and PPARa activation protected B-cells from palmitate-induced cell death, whereas blocking CPT-l activity increased susceptibility to cell death from palmitate (237, 238). Together, these studies demonstrate that the intracellular capacity to modulate FA synthesis and oxidation could significantly predispose B-cells to the damaging effects of glucotoxicity and glucolipotoxicity. 3.2.4. Dysregulated Glycerolipid/Fatty Acid Cycling The role of glycerolipid/F A cycling in normal fi-cell function, particularly GSIS, relies on balanced synthesis and turnover of neutral lipid pools. To determine if excessive TAG synthesis was directly implicated in the loss of GSIS during chronic hyperglycemia, the final step of TAG synthesis in islets was increased by over-expression of DGATI (239). Elevated DGATl increased TAG accumulation and reduced GSIS, demonstrating that a loss of B-cell function may occur without a paralleled increase in TAG turnover (239). In line with this idea, knockdown of the TAG lipase ATGL reduced TAG turnover as well as GSIS (165). Conversely, excessive neutral lipid hydrolysis by over-expression of HSL also impairs GSIS (240), further supporting the importance of balanced lipid synthesis and turnover for normal B-cell function. In 38 addition to . TAGS than caused by s; cycling cont 3.2.5. Apopt Redu failure and a; the level of cells occupy clear, loss of 3p0ptosis. L .ATF-l-mediat (CEBP) ho, activation of, lSlets cultured and from hun dclction in comPOUem 01‘ Pathways, Suc Protects fmm bonuses pal a (.248, 249). > addition to glucotoxicity, exogenous saturated FAs are incorporated less efficiently into TAGS than MUFAs, which coincides with diminished GSIS and increased cell death caused by saturated F As (241). These studies show that dysregulation of glycerolipid/F A cycling contributes to (El-cell dysfunction. 3.2.5. Apoptosis-Mediated Loss of B-Cell Mass Reduced B—cell mass, or number of B-cells, that occurs naturally or due to B-cell failure and apoptosis is a critical factor in the development of T2D. In obese individuals, the level of fasting plasma glucose inversely correlates with the average percent of [3- cells occupying the whole pancreas (242). Although the mechanisms are not entirely clear, loss of B-cell mass has been attributed to both ER and mitochondrial induction of apoptosis. Unresolved ER stress can induce apoptosis via pathways that include the ATF4-mediated transcription of the proapoptotic genes CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP) and ATF3 (243, 244) and [RBI-mediated activation of c-Jun N-terminal kinase (JNK) (245, 246). CHOP induction is increased in islets cultured in hyperglycemic conditions, exposed to elevated palmitate concentrations, and from human T2D patients (220, 222, 224). Protection from B-cell death by CHOP deletion in multiple diabetes models further demonstrated CHOP to be a major component of ER stress-mediated apoptosis (247). Activation of JNK affects multiple pathways, such as CHOP induction, and inhibition of JNK activity in fi-cells partially protects from palmitate-induced apoptosis (222). In addition to the IREl and PERK pathways, palmitate-induced B-cell ER stress depleted Ca2+ from the ER (222), which can trigger mitochondrial release of cytochrome C and initiation of the caspase—9 cascade (248, 249). This cascade in part causes deoxyribonuclease-mediated DNA degradation 39 (250). at Mitochont (251). Ta. conditions apoptotic p (250), an effective marker for palmitate-induced (fl-cell apoptosis (223, 224). Mitochondrial release of cytochrome C has also been associated with generation of ROS (251). Taken as a whole, this illustrates that (fl-cell dysfunction from exposure to chronic conditions of glucotoxicity and glucolipotoxicity can lead to the induction of downstream apoptotic pathways and contribute to reduced B-cell mass. 40 Glucose Exogenous SFA (16:0) Malonyl-CoA \ gill. l/,\/ f LC-CoA (16:0-60A) ”3530A GLIFA ‘. cycling ER t Glycerolipid s ress - (UPR) I TAG MitoJER Oxidative stress - (ROS) l Diminished Glucose-Stimulated Insulin Secretion Decreased Insulin Gene Expression Loss of B-Cell Mass Figure 1.7. Mechanisms involved in B-cell dysfunction from glucolipotoxicity. Chronically elevated glucose drives malonyl-CoA synthesis, which blocks mitochondrial FA oxidation by inhibition of CPT-1. Next, LC-CoAs formed from de novo and exogenous saturated FA (SFA) are mostly driven towards glycerolipid storage as TAG. Accumulation of endogenous glucose metabolites and SFA-CoAs causes ER and oxidative stress to the B-cell, displaying activation of the unfolded protein response (UPR) and generation of reactive oxygen species (ROS). Sustained UPR activation and ROS generation lead to diminished glucose-stimulated insulin secretion, decreased insulin gene expression, and ultimately a loss of B-cell mass. 41 3.3. Evidence Prior Ii mechanisms to failure from g through enhan identified to p; 3.3.]. SREBP- ln anin expression of j that increased The link betm Of a Constituti increased T AC by studies Sho‘ secreting mor— (ZDF) rat, inc decreased GS isolated for l.‘ 1;) mime 90nd. 1; 3.3. Evidence and Mechanisms of B-cell Compensation Prior to the onset of T2D, B-cells are capable of activating various compensatory mechanisms to com with the increased demand for insulin secretion and to prevent B-cell failure from glucolipotoxicity. Of these mechanisms, regulation of FA metabolism through enhanced lipogenesis, glycerolipid/FA cycling, and MUF A synthesis have been identified to participate in B-cell compensation. 3.3.1. SREBP-1c and Liver X Receptors In animal models of T2D, diminished GSIS has been associated with increased expression of SREBP-1c and TAG accumulation in islets, and this led to the hypothesis that increased neutral lipid storage via SREBP- 1c activation was toxic to B-cells (233). The link between SREBP-1c activation and loss of GSIS was shown by over-expression of a constitutively active form of SREBP-1c, which forced lipogenic gene expression, increased TAG synthesis, and reduced GSIS (116). This idea, however, was contradicted by studies showing that the ability of islets to compensate for chronic hyperglycemia by secreting more insulin was dependent on SREBP-1c (234). In the Zucker diabetic fatty (ZDF) rat, increased islet SREBP-1c expression coincides with elevated lipogenesis and decreased GSIS (233). It was recently shown, however, that over-expression of a dominant negative SREBP-1c in ZDF rat islets prevented the increase in lipogenic gene expression and TAG content, but it did not correct for the diminished GSIS (252). In addition to SREBP-1c, activation of LXRs with the synthetic agonist T0901317 in isolated rat islets and fi-cell lines increased nuclear SREBP-1c accumulation, lipogenic gene expression, and TAG content as well as elevated both basal and GSIS under normal culture conditions (5, 118). Although elevated insulin secretion from LXR-activated (3- 42 cells was inh Furthermore. from B-cells suggest that e in response it 33.2. Glycer Dysre models to car FAS into gig-(- iats, obese 21 The ability 01 cell glucose- ; ml islets we CSIerif‘lCation inhibition of] 5€Cre10ry res; indicating Zr eXCess FFAs T2D ll] ObeSL 3.3.3. MOnO E1e\ L: \l’llll high ex cells was inhibited by knockdown of SREBP-1c (253), its mechanism is still unknown. Furthermore, it remains to be determined if LXR activation can enhance insulin secretion from B-cells cultured chronically in hyperglycemic conditions. Together, these studies suggest that enhanced de novo lipogenesis may have a critical role in B-cell compensation in response to glucotoxic and glucolipotoxic conditions. 3.3.2. Glycerolipid/Fatty Acid Cycling Dysregulation of glycerolipid/FA cycling in (i-cells has been shown in various models to cause reduced GSIS. This implies that balanced incorporation and turnover of FAs into glycerolipids is important for maintenance of B-cell function. Unlike the ZDF rats, obese Zucker fatty (ZF) rats exhibit insulin resistance but maintain norrnoglycemia. The ability of ZF rats to maintain glucose homeostasis was associated with enhanced (3- cell glucose- and FA-stimulated insulin secretion compared to lean control rats (104). ZF rat islets were identified to have increased glucose-responsive capacities for FFA esterification and lipolysis, evidence of enhanced glycerolipid/F A cycling (104). Inhibition of lipolysis by the general lipase inhibitor orlistat blocked the increased insulin secretory response (104). In addition, FA oxidation tended to be elevated as well (104), indicating ZF islets may have enhanced activation of pathways for detoxification of excess FFAs. This in vivo evidence demonstrates that enhancing both synthesis and turnover of glycerolipids in B-cells may significantly reduce susceptibility to developing T2D in obese individuals. 3.3.3. Monounsaturated Fatty Acid Synthesis Elevated TAGS often occur in obesity-associated T2D and has been correlated with high expression and activity of SCD] in adipose and liver (254). In rodent models 43 lacking SC D high fat- and SC 05 could One study. susceptible It associated vv suggesting St In agreement levels of SCI likely, hovvev for example. also exhibitec contribute to itself can p1 determined 1 raising the qut lacking SCDl expression, TAG levels remain low and these animals are protected from high fat- and high carbohydrate-diet induced obesity and T2D (48, 255), suggesting that SCDs could be potential pharmacological targets for the treatment of these conditions. One study, however, showed that deletion of SCDl in mice that are genetically susceptible to obesity and T2D resulted in an earlier onset of T2D (256). This effect was associated with the accumulation of saturated F As in islets and diminished GSIS (256), suggesting SCDl expression and activity is important for maintenance of fi-cell function. In agreement with this hypothesis, rat islets and B-cell lines with naturally occurring high levels of SCD] expression are protected from palmitate-induced cell death (6, 257). It is likely, however, that altered expression of other genes could also be involved as well. For example, the subpopulation of palmitate-resistant B-cells from the MIN-6 B-cell line also exhibited increased expression of CPT-1 and enhanced FA oxidation, which would contribute to the detoxification of palmitate (6). Whether over-expression of SCD by itself can protect (ft-cells from saturated FA-induced dysfunction remains to be determined. Intriguingly, the SCD2 isoform is highly expressed in rat islet B-cells (257), raising the question of whether SCD2 has a significant role in islet B-cell function. 44 4. Statemen Reg1 lucolipoto no to be depei mediated r: activation c bOIh basal glucose. 11 nova lipoge found that S}nthesis, a SCDS, the 1 Wm 5.1711 assays, I ex MUFA S,Vn1 e1Gilgation i Elongation, [ and lSlets fr the effects . lunction in r “mm/2951': Slg’llficaml; 4. Statement of Problem and Specific Aims Regulation of FA metabolism significantly affects pancreatic B-cell responses to glucolipotoxicity. The role of SREBP-1c in B—cell compensation versus failure appears to be dependent on the level of its activation. Using a synthetic LXR agonist, LXR- mediated regulation of SREBP-1c provides a way to examine the effects of moderate activation of lipogenesis on B-cell function. Our lab identified LXR activation to elevate both basal insulin secretion and GSIS from INS-l B-cells chronically cultured in high glucose. In light of this finding, I explored the effects of LXR activation on B-cell de nova lipogenesis and the mechanism of elevated insulin secretion. During this research I found that LXR-activated B-cclls have an enhanced capacity for de nova MUFA synthesis, and this coincided with increased expression of SCDl and SCD2. Unlike SCDs, the rate-limiting enzymes for MUF A synthesis, role of FA elongases in de nova MUFA synthesis is unknown. Thus, based on known substrate specificity elongation assays, I examined the effects of altered Elovl-5 and Elovl-6 expression on de nova MUFA synthesis. In contrast to the liver, less is known about FA desaturation and elongation in B-cells. To better understand the role of B-cell FA desaturation and elongation, I characterized FA desaturase and elongase gene expression in INS-1 B-cells and islets from the ZDF rat model of progressive B-cell failure. In addition, I examined the effects of altering SCD2 and Elovl-6 expression on B-cell FA metabolism and function in response to elevated levels of exogenous saturated FAS. I hypothesized that modulation of de nova lipogenesis and monounsaturated FA synthesis significantly effects pancreatic ,B-cell compensation in response to glucolipotoxicity. 45 The specifis Aim 1: To smthesis ar insulin relez Aim 2: To synthesis of Aim 3: TO I Ofprogressi involved in exogenous s The findin y!“ is.) \r H metaboli. The specific aims are: Aim 1: To characterize the effects of LXR-activation on de nova FA and neutral lipid synthesis and to determine which aspects of FA metabolism contribute to elevated basal insulin release and GSIS during chronic hyperglycemia. Aim 2: To define the roles of the FA elongases Elovl-5 and Elovl-6 in the de nova synthesis of MUFAs. Aim 3: To characterizes the expression of FA desaturase and elongase genes in a setting of progressive B-cell failure and to examine the effects of altering the expression of genes involved in MUFA synthesis on fi-cell FA metabolism and viability in response to exogenous saturated FAs. The findings from this research will contribute to the understanding of how regulation of FA metabolism effects B-cell compensation in response to glucolipotoxicity. 46 Chaptel 1. Mater Palmitic : [S-Snglt and D-[Lf acid-free (Indianap (Rank-aka nere phos caspase-9 l GADDISS l6lS-HRP; 1:3300 wer goat anti-mi Was from R( 2. Fatty acit Previously (; Chapter 2. Materials and Methods 1. Materials. T0901317 was purchased from Cayman Chemical (Ann Arbor, MI). Palmitic acid was from Nu-Chek Prep, Inc. (Elysian, MN). [1-14C]palmitic acid and D- [5-3H]glucose were from PerkinElmer Life Sciences (Boston, MA). [2-14C]acetic acid and D-[U-14C]glucose were from ICN Pharmaceuticals, Inc (Costa Mesa, CA). Fatty acid-free BSA for insulin secretion studies was from Roche Applied Science (Indianapolis, IN). Fatty acid-free BSA for FA treatments was from Celliance (Kankakee, IL). Primary antibodies used at a 1:1000 dilution, unless noted otherwise, were phospho-JNK (pSAPK/JNK; 9255), total JNK (SAPK/JNK; 9252), and cleaved caspase-9 (9507) from Cell Signaling Technology (Beverly, MA); SREBP-1 (IgG-2A4), GADD153/CHOP (SC-7351) and actin-HRP (horseradish peroxidase-conjugated; sc- 1615-HRP) at 1:3300 from Santa Cruz (Santa Cruz, CA). Secondary antibodies used at 123300 were goat anti—rabbit-HRP from Vector Laboratories, Inc. (Burlingame, CA) and goat anti-mouse-HRP from Bio-Rad Laboratories (Hercules, CA). Apoptosis ELISA kit was from Roche Diagnostics (Cell Death Detection ELISA; Indianapolis, IN). 2. Fatty acid preparation. Palmitic acid was bound to fatty acid free BSA as described previously (258). Briefly, stock solutions of 100 mM palmitic acid dissolved in 0.1 M sodium hydroxide and 5% fatty acid free BSA dissolved in RPMI-1640 without glucose were heated to 70°C and 55°C, respectively. The palmitic acid solution was then added drop-wise into the BSA solution to make a final stock of 5 mM palmitic acid/5% BSA. The solution was then kept at 55°C for 10 min, vortexed, brought to room temperature, and either used immediately or stored at —20°C for up to 3 three weeks. Aliquots of the 5 mM/5% palmitic acid/BSA stocks were then thawed at 55°C for 15 min, vortexed, and 47 brought to r concentratit' 3. Human Foundation of Miami. ax RPMI-16-10 and 0.5 pg n days in N2 r experiments. 22.1 mM glL same conditi. 4. Animals 3 animal care a from Charles 10 islet isolat. [3176721501 an from Charles a l2212 hr “.5 ZDF rats We! colleqed ffCu lnsulin Radi. fIOm panCre; brought to room temperature before diluting into culture media to give the final indicated concentrations of fatty acid and BSA (0.5%). 3. Human islets. Human islets were obtained from the Juvenile Diabetes Research Foundation Human Islet Distribution Program at the University of Minnesota, University of Miami, and Northwest Tissue Center, Seattle. Islets were maintained in N2 medium or RPMI-l640 plus 10% PBS containing 100 units/ml penicillin, 100 pg/ml streptomycin and 0.5 pg/ml fungizone. For gene expression experiments, islets were cultured for three days in N2 medium in the absence or presence of 10 pM T0901317. For lipid synthesis experiments, islets were cultured for 36 hrs in RPMI-l640 medium containing 11.1 or 22.1 mM glucose plus 5 uM T0901317 (see below). Then incubated for 12 hrs under the same conditions in the presence of 1 pCi [2-14C]acetic acid. 4. Animals and islet isolation. All animal procedures were approved by the institutional animal care and use committee at Michigan State University. Sprague-Dawley rats were from Charles River Laboratories and fed Harlan-Teklad laboratory chow (N o. 8640) prior to islet isolation between 8-10 weeks old. Male fa/fa Zucker Diabetic Fatty rats (ZDF- Leprfa/Crl and lean controls (fa/1’) received at 4 and 11 weeks of age were purchased from Charles River Laboratories and fed the Purina 5008 diet. All animals were kept on a 12:12 hr lightzdark cycle with food and water ad libitum. At 6 and 13 weeks of age, ZDF rats were weighed and blood glucose measured. Prior to islet isolation, blood was collected from the abdominal aorta and plasma insulin levels were measured using a Rat Insulin Radioimmunoassay (RIA) kit (Millipore, Billerica, MA). Islets were dissociated from pancreatic tissue by collagenase digestion and isolated by hand. 48 5.1554 “9 media Com: mercaploeti described (2 cm: and CU1 INS-l medi. fatty acid tr media conwi 6. siRNA ' Dhannacon. using the Ar butler (7 mM mM glucose) with modifiec or with l uhl protein analys palmitate, €l€l treatment wi 14Clpalmitie glucose and glucose. Cel “ll! 1 MG {‘ CA), alter til 5. INS-l cell culture. INS-1 cells were routinely cultured in IN S-l media (RPMI-1640 media containing 11.1 mM glucose, 10% F BS, 1 mM pyruvate, 10 mM Hepes, 50 uM 2- mercaptoethanol, 100 units penicillin/ml and 100 ug streptomycin/ml) as previously described (259). In all experiments, cells were seeded at a density of 0.2x106 cells per cm2 and cultured for 24 or 48 hrs in INS-l media. Cells were then cultured for 48 hrs in INS-1 media containing 4 or 16.7 mM glucose and vehicle or 10 uM T0901317. For fatty acid treatments, cells were cultured for the indicated times in a modified INS-1 media containing 0.5% FBS and palmitate complexed to BSA as described below. 6. siRNA treatment. Control, Elovl—5, Elovl-6 and SCD2 siRNA were from Dharmacon, Inc. (Lafayette, CO). siRNAs were introduced into cells by electroporation using the Amaxa Nucleofector (program D-026, Gaithersberg, MD) in electroporation buffer (7 mM ATP, 11.6 mM MgC12-6H20, 68 mM K2HPO4, 14 mM NaHCO3, and 2.2 mM glucose). Cells were cultured for 24 hrs in INS-l media and subsequently treated with modified INS-1 media containing increasing concentrations of palmitic acid without or with 1 uM G66976 for the indicated times. Cells were then harvested for mRNA and protein analysis, and apoptosis (see below). For FA and complex lipid analyses using palmitate, electroporated cells were cultured for 24 hrs in INS-1 media followed by 12 hr treatment with modified INS-l media plus 400 MM palmitic acid and luCi [1- 14C]palmitic acid. For de nova FA analysis, cells were cultured for 24 hrs in 5.5 mM glucose and subsequently cultured for 24 hrs in INS-1 media containing 11.1 mM glucose. Cells were then harvested for mRNA analysis or continued culturing overnight with 1 uCi [2-14C]-acetic acid (51 mCi/mmol, ICN Pharmaceuticals, Inc., Coasta Mesa, CA), after which lipids were extracted and analyzed for MC incorporation. 49 7. Aden construct was clan: using ATAGTC ATACTC recombin; Vector S) pShuttle-C ultracompc Adenoviru: Rapid Tite galactosida transductio; With 5 0r 1. glucose INS indicated lip 7. Adenovirus preparation. Elovl-5 and Elovl—6 cDNA were cloned and used for construction of recombinant adenoviruses as previously described (35). SCD2 cDNA was cloned into TOPO plasmids (Invitrogen) from reverse transcribed INS-1 cell mRNA using the following primers: sense 5’- ATAGTCGACATGCCGGCTCACATACTGCAAGAG-3 ’; antisense 5 ’- ATACTCGAGTCAGCCACTCTTGCAGCTCTCCTCCCC-3’ (Acc. No. AB032243). A recombinant adenovirus over-expressing SCD2 was constructed using the Adenoviral Vector System (Stratagene). In short, the coding regions of SCD2 were ligated into pShuttle-CMV, recombined with pAdEasy—l in BJ 5183 cells, propagated in XL10 Gold ultracompetent cells, and packaged into adenoviral particles in Ad—293 cells. Adenoviruses were further amplified and then titered in HEK293 cells using the Adena-X Rapid Titer kit (Clontech, Mountainview, CA). An adenovirus over-expressing [3- galactosidase was obtained from Dr Newgard, Duke University, North Carolina. For transduction of genes into INS-l cells, 90% confluent cell cultures were infected for 2 hrs with 5 or 10 pfu per cell and then cultured for an additional 24 hrs in 5.5 or 11.1 mM glucose INS-1 media to allow for gene expression. Afierwards, cells were treated for the indicated times in modified INS-1 media without or with increasing concentrations of palmitic acid. Cells were then harvested for protein analysis and apoptosis. For FA and complex lipid analyses using palmitate, cells were infected for 2 hrs, cultured for 24 hrs in INS-l media, and treated with 400 uM palmitic acid with 3 uCi [1-14C]palmitic acid for 12 hrs prior to lipid extraction. For de nova FA analysis, cells were cultured in INS-l media containing 11.1 mM glucose with 1 uCi [2-14C]-acetic acid for 24 hrs prior to lipid extraction. 50 8. RNA 2 mRNA in fluorogeni amounts c Results fo rodent isle” (Bio-Rad. combining primers (Ir qPCR Sup: 316000? q mRNAs we analyzed in Prorein L 3 3. (Tables 2_2 , C 8. RNA analysis. Total RNA was extracted using TRIZOL (Invitrogen). Levels of mRNA in human islets were determined by real-time qPCR using Light Upon Extension fluorogenic primers (Invitrogen, Table 2.1) as previously described (260). Relative amounts of mRNA were calculated using the comparative cycle threshold method. Results for human islets were normalized to the abundance of B—actin mRNA. For rodent islets and INS-1 cells, cDNA were synthesized using iScript cDNA Synthesis kit (Bio-Rad, Hercules, CA). Quantitative real—time (qRT) PCR was conducted by combining synthesized cDNA and various sets of gene-specific forward and reverse primers (Integrated DNA Technologies, Coralville, IA) with Platinum SYBR Green qPCR SuperMix-UDG (Invitrogen). qRT-PCR reactions were carried out using the Mx3000P quantitative PCR System (Stratagene, La Jolla, CA). The relative amounts of mRNAs were determined by the comparative cycle threshold method. All samples were analyzed in triplicate. Gene expression is reported relative to cyclophilin or ribosomal protein L32 (RPL32) mRNA levels. Primers for SYBR Green qRT-PCR are listed in (Tables 2.2 and 2.3). 51 Table 2 ______.—.————-—- Light I Name fi-actin SREBP ACC FAS ib-carbox internal Ct §6-carbox reactions. Table 2.1. Oligodeoxynucleotide sequences used for quantitative RT-PCR. Light Upon Extension Quantitative RT-PCR primers Name Species Direction Sequence (5’ to 3’) B—actin Human Forward CACGCCACCTTCTACAATGAGCTGCGG# Reverse GGTCATCTTCTCGCGGTTGG SREBP—1 Human Forward GACGGCCTCTGGAACCTCATCCGTC§ Reverse TAGCATCCACTCGCAGAGCA ACC Human Forward CACATGCTCCAAACCAGGCCATGTG§ Reverse GCCAGTCCACACGAAGACCA FAS Human Forward CACCTTAACCTGGTAGTGAGTGGGAAGGTG§ Reverse CTTTCCGGGTGGTCGAAGA #6-carboxy-4’,5’-dichloro-2’,7’-dimethoxy-fluorescein-labeled B-actin primer used as internal control for all real-time RT—PCR reactions. §6-carboxy-fluorescein—labeled forward PCR primers used for real-time RT-PCR reactions. 52 Table 2.2 SYBR G Cyclophi RPL32 SREBP-l ACCa FAS ABCAl ABC 01 CPT-l a CPT-2 VLCAD LC AD \ Table 2.2. Oligodeoxynucleotide sequences used for. quantitative RT-PCR. SYBR Green Quantitative RT—PCR primers Name Species Direction Sequence (5’ to 3’) Cyclophilin Rat Forward CTTCTTGCTGGTCTTGCCATTCCT Reverse TGGATGGCAAGCATGTGGTCTTTG RPL32 Rat Forward AAACTGGCGGAAACCCAGAG Reverse GCAATCTCAGCACAGTAAGATT SREBP- 1 Rat Forward GATTGCACATTTGAAGACATGCTT Reverse GGGTCCCAGGAAGGCTTCCAGAGA ACCOt Rat Forward CGATGTI‘CTGTTGGACAACGCCTT Reverse TCTCTGATCCACCTCACAGTTGAC FAS Rat Forward GTGCACCCCATTGAAGGTTCC Reverse GGTTTGGAATGCTGTCCAGGG ABCAl Rat Forward AGCAGTTTGTGGCCCTCTTGT Reverse TGAAGTTCCAGGTTGGGGTACTTG ABCGl Rat Forward ATGGAAGGTTGCCACAGCTTCTC Reverse AGTCATGGTCTTGGCCAGGTAGT CPT-la Rat Forward AGACCGTGAGGAACTCMACCCAT Reverse CACAACAATGTGCCTGCTGTCCTT CPT-2 Rat Forward TCCTGCATACCAGCAGATGAACCA Reverse ACAGTGGAGAAACTCTCGGGCATT VLCAD Rat Forward GTGGGAATGTTCAAAGGCCAGCTT Reverse AAGGAGTCATTCTTGGCAGGGTCA LCAD Rat Forward AATGGGAGAAAGCCGGAGAAGTGA Reverse GAAACCAGGGCCTGTGCAATTTGA 53 D60 Eknl-l EvaZ EMVL3 Eva4 EbVLS EmrL6 Ekwi7 Xmfls Xhflt ATF3 CHOP \ Table 2.3. Oligodeoxynucleotide sequences used for quantitative RT-PCR. SYBR Green Quantitative RT-PCR primers Name Species Direction Sequence (:to 3’) Insulin Rat Forward GCTTTTGTCAAACAGCACCTT Reverse CTCCAGTGCCAAGGTCTGAAG SCD] Rat Forward ACATTCAATCTCGGGAGAACA Reverse CCATGCAGTCGATGAAGAAC SCD2 Rat Forward ATGCCGGCTCACATACTG Reverse GACCAGTGTGATCCCGTACA D5D Rat Forward TGGAGAGCAACTGGTTTGTG Reverse GTTGAAGGCTGACTGGTGAA D6D Rat Forward TGTCCACAAGTTTGTCATTGG Reverse ACACGTGCAGGCTCTTTATG Elovl-1 Rat Forward CCCTACCTTTGGTGGAAGAA Reverse TCCAGATGAGGTGGATGATG Elovl-2 Rat Forward TTTGGCTGTCTCATCTTCCA Reverse GGGAAACCGTTCTTCACTTC Elovl-3 Rat Forward AATTCTGGTCCTGGGTCTTTC Reverse CCAAAGCTCGTAAACAGTAGCA Elovl-4 Rat Forward GAAGTGGATGAAAGACCGAGA Reverse GCGTTGTATGATCCCATGAA Elovl-5 Rat Forward ACAGCTTCATCCACGTCCTCATGT Reverse AGCTGGTCTGGATGATTGTCAGCA Elovl-6 Rat Forward CAACGGACCTGTCAGCAA Reverse GTGGTACCAGTGCAGGAAGA Elovl-7 Rat Forward TGGCGTTCAGCGATCTTAC Reverse GATGATGGTTTGTGGCAGAG Xbp 1 s Rat Forward GAGTCCGCAGCAGGTG Reverse GCGTCAGAATCCATGGGA Xbplt Rat Forward GAGCAGCAAGTGGTGGATTT Reverse TCTCAATCACAAGCCCATGA ATF3 Rat Forward GAAGGCACAAAGTCCCGCTTTCAA Reverse TTCAAATACCAGTCTCCACGGGCT CHOP Rat Forward AACTGTTGGCATCACCTCCTGTCT Reverse TCCTCAGCATGTGCACTGGAGATT 54 9. Western blot analysis. For analysis of SREBP-l protein, microsomes and nuclear extracts were isolated as previously described (261). Proteins (30-100 pg) were resolved by SDS-PAGE and transferred to nitrocellulose membranes as described previously (262). Proteins were detected with specific primary antibodies and the corresponding secondary antibodies and the bands were then visualized on film with SuperSignal West Pico and Dura chemiluminescent kits (Thermo Fisher Scientific, Rockford, IL). Protein bands were quantified by densitometry scanning. 10. Insulin secretion studies. IN S-l cells were preincubated twice for 30 min at 37°C in Krebs Ringer bicarbonate buffer (KRBB) (259) containing 2 mM glucose and subsequently incubated for 60 min in KRBB containing either 2 or 20 mM glucose. For studies using etomoxir (100 uM), orlistat (50 uM) or calphostin C (1 uM), agents were present throughout the incubation period in KRBB. Triacsin C (10 uM) was added 5 hrs prior to secretion studies. Verapamil (100 uM) was only present during the final 1 hr .incubation in KRBB. Insulin released into the media and insulin content from acid- ethanol extracted cells were determined by radioimmunoassay (Linco, St. Louis, MO). Total cell protein was determined by Lowry assay. 11. Glucose utilization studies. Glucose usage was measured using a modification of the method of Zawalich and Matschinsky (263) as previously described (259). Briefly, cells were incubated for 30 min at 37°C in KRBB containing either 2 or 16.7 mM glucose and [5-3H]glucose. Duplicate 50 ul samples were added to a tube containing 1 N HCI. The tubes were placed in vials containing 0.5 ml of H20, sealed, and incubated at 50°C for 18 hrs. Tubes were then removed from vials, scintillation cocktail was added, and 55 samples “Ci expressed as 12. Comple: cells were c vehicle or 1( containing 4 acid (51 mt extracted as lipid extracts (390 Quar. Phosphoimaé Anal) FA analysis 1 mM glUCOSe. was added I 0‘ KOH in 809 resusPended phase‘HPLC qUBntified b Toral F As quantified t:- For: lllCUbated ft samples were counted in a Beckman scintillation counter. Glucose utilization was expressed as picomoles of glucose metabolized per min per mg protein. 12. Complex lipid analysis and fatty acid profile. For de nova lipid synthesis, INS-l cells were cultured for 48 hrs in INS-l media containing 4 or 16.7 mM glucose and vehicle or 10 pM T0901317. During the last 12 hrs, cells were cultured in INS-l media containing 4 or 16.7 mM glucose, vehicle or 5 uM T0901317, and 1 uCi [2-14C]acetic acid (51 mCi/mmol). Cells from l4C-labeling studies were harvested and lipids extracted as previously described by Pawar et al. (264). For analysis of complex lipids, lipid extracts were separated by thin layer chromatography (TLC) as previously described (264). Quantification of l4C-labeled lipids was determined on a Molecular Dynamics Phosphoimager 820. Analysis of 14C-labeled FA profile was performed as previously described (35). FA analysis from glucose was determined by culturing INS-1 cells for 48 hrs in 4 or 16.7 mM glucose, and during the last 24 hrs 4 or 16.7 pl of [U-14C]glucose (260 mCi/mmol) was added to the media. Total lipid extracts from labeling studies were saponified (0.5 N KQH in 80% methanol, 1 hr at 50°C), neutralized, extracted in diethyl ether, dried, and resuspended in methanol and 0.1 mM BHT. FAs were then fractionated by reverse phase-HPLC using a J ’sphere ODS—H80 (YMC-Waters, Milford, MA) column and quantified by flow-through scintillation counting (IN/US Systems, Inc., Brandon, FL). Total FAS were quantified by evaporative light scatter, and unsaturated FA were quantified by UV absorption at 192 nm. For total FA synthesis from glucose during the insulin secretion study, cells were incubated for 1 hr with either 2 or 20 mM glucose plus 4 or 40 pCi of [U-14C]glucose 56 (260 mCi/mmol), respectively. Cells were extracted, extracts saponified, and 14C- labeled FA quantified as described above. Indexes of elongation and desaturation were determined using the 14C counts incorporated into each specific FA species and calculating the ratios of product(s) to substrate. Elongation of 16:0 and 16:1,n-7 was determined from the ratios of 18:0 plus 18:1,n-9 to 16:0 and 18:1,n-7 to 16:1,n-7, respectively. Desaturation of 16:0 and 18:0 was determined from the ratios of 16:1,n-7 plus 18:1,n-7 to 16:0 and 18:1,n-9 to 18:0, respectively. 13. Palmitate oxidation. INS-l cells were then incubated for 1 hr in KRBB containing 2 mM glucose, after which cells were incubated for 1 hr in KRBB containing 50 uM palmitate, 2 pCi/ml [1-14C]palmitic acid (56 mCi/mmol), and 2 or 20 mM glucose. Palmitate oxidation was determined by measuring [14C]C02 released into the medium using the method of Parkera et al. (265). 14. Statistical analysis. Islet studies are representative of 5 to 6 animals per group. All INS-l cell data represent 3 to 6 independent experiments performed in duplicate. Statistical significance was determined using one-way ANQVA followed by Bonferroni’s multiple comparison test for more than two groups or Student’s t-test for comparing two groups. P values < 0.05 were considered significant. 57 Chapter 3. Elevated Insulin Secretion From Liver X Receptor-Activated Pancreatic B-Cells Involves Increased de Nova Lipid Synthesis and Triacylglyceride Turnover Abstract Increased basal and loss of glucose-stimulated insulin secretion (GSIS) are hallmarks of B-cell dysfunction associated with type 2 diabetes. It has been proposed that elevated glucose promotes insulin secretory defects by activating sterol regulatory element binding protein-1c (SREBP-1c), lipogenic gene expression and neutral lipid storage. Activation of liver X receptors (LXR) also activates SREBP-1c and increases lipogenic gene expression and neutral lipid storage, but increases basal and GSIS. This study was designed to characterize the changes in de nova fatty acid (FA) and triacylglyceride (TAG) synthesis in LXR-activated B-cells and determine how these changes contribute to elevated basal and GSIS. Treatment of INS-l B-cells with a LXR agonist T0901317 and elevated glucose led to markedly increased nuclear localization of SREBP-1, lipogenic gene expression, de nova synthesis of monounsaturated fatty acids and TAG, and basal and GSIS. LXR-activated cells hadincreased FA oxidation and expression of genes involved in mitochondrial B-oxidation particularly camitine palmitoyltransferase-l. Increased basal insulin release from LXR-activated cells coincided with rapid turnover of newly synthesized TAG and required acyl-CoA synthesis and mitochondrial B-oxidation. GSIS from LXR-activated IN S-l cells required influx of extracellular calcium and lipolysis suggesting production of lipid-signaling 58 molecules from TAG. Inhibition of diacylglyceride (DAG)-binding proteins, but not classic isoforms of protein kinase C, attenuated GSIS from LXR-activated INS-l cells. In conclusion, LXR activation in B-cells exposed to elevated glucose concentrations increases de nova TAG synthesis; subsequent lipolysis produces free fatty acids and DAG, which are oxidized to increase basal insulin release and activate DAG—binding proteins to enhance GSIS, respectively. Introduction Type 2 diabetes mellitus occurs when pancreatic B-cells fail to secrete sufficient amounts of insulin necessary to overcome insulin resistance at peripheral tissues and to maintain glucose homeostasis. Loss of B-cell function in type 2 diabetes has been suggested to occur when B-cells are chronically exposed to elevated circulating glucose and free fatty acids (F FA) — a state defined as ‘glucolipotoxicity’ (3, 4). Sterol regulatory element binding protein 1c (SREBP-1c), a basic helix 100p helix transcription factor, plays a major role in inducing lipogenic gene expression in liver and adipose tissue (69, 266, 267), and thereby partitions glucose towards synthesis of lipid. The ability of elevated glucose to increase the nuclear form of SREBP-1c in B—cells (114, 115, 268) has been proposed to serve as a possible mechanism for glucolipotoxicity, and to explain the predominate role of elevated glucose in B-cell dysfunction (268). Consistent with this hypothesis, expression of a constitutively active nuclear form of SREBP-1c in islets or B- cell-lines increased lipogenic gene expression, triacylglyceride (TAG) synthesis and storage, and suppressed glucose-stimulated insulin secretion (GSIS) (115, 116, 235, 268, 59 Ell 269). Islet failure in Zucker Diabetic Fatty (ZDF) rats is also associated with increased expression of SREBP-1c and TAG accumulation (232, 233, 270). Although activation of SREBP-1c is an attractive mechanism to explain glucolipotoxicity, recent reports suggest that the role of SREPB-lc in fi-cell function is more complex. First, SREBP-1c activation and lipid synthesis are required for adaptive changes leading to hypersecretion of insulin from mouse islets exposed to elevated glucose concentrations (234). Second, inactivation of SREBP-1c in ZDF rat islets failed to restore GSIS suggesting that increased SREBP-1c and intracellular TAG are not the principal cause of B-cell secretory dysfunction (252). Third, activation of liver X receptors (LXR) in islets and B-cell-lines increased SREBP-1c, lipogenic gene expression, neutral lipid storage, and basal and GSIS (5, 253). LXRor (NR1H3) and LXRB (NR1H2), are nuclear receptors involved in transcriptional control of genes involved in cholesterol, fatty acid, and glucose metabolism (271). LXRor is primarily expressed in liver, kidney, intestine, and macrophages, whereas LXRB is ubiquitously expressed (78, 272). LXR is activated by oxysterols (273) and glucose (77). In macrophages, LXR regulates reverse cholesterol transport through increased expression of genes encoding ATP-binding cassette (ABC) cholesterol transporters ABCAI (274) and ABCGl (275). In liver, LXR controls transcription of genes involved in conversion of cholesterol into bile acids (273, 276) and excretion of biliary cholesterol (277). LXR also directly and indirectly, through increased SREBP-1c expression, activates lipogenic gene transcription including acyl CoA carboxylase (ACC), fatty acid synthase (FAS), and stearoyl CoA desaturase l (SCDI) and 2 (SCD2) (81, 278-280). 60 CK LXRa and LXRB are expressed in rodent and human pancreatic islets and B-cell lines (5, 281). Islets from LXRB knockout mice accumulated lipid, have reduced expression of cholesterol transporters, and reduced GSIS (117, 253). Treatment of islets and B-cell lines with the LXR agonist T0901317 increased expression of lipogenic genes and lipid accumulation (5, 118). Importantly, LXR agonists elevated basal and GSIS from islets and B-cell lines through a mechanism dependent upon increased SREBP—1c, and pyruvate carboxylase and ACCor activity (5, 253). These findings suggest that LXR activation promotes insulin release by stimulating anaplerotic and cataplerotic pathways, possibly to supply malonyl-CoA for de nova fatty acid and lipid synthesis. The principal aims of the study presented herein were to characterize the changes in de nova FA and TAG synthesis in LXR-activated B-cells and determine how these changes in lipid metabolism may contribute to elevated basal and GSIS. 61 Results 3.1. Effect of glucose and LXR activation on SREBP-1 and lipogenic gene expression. Culturing INS-1 cells in 16.7 mM glucose for 48 h increased protein levels of both the microsomal precursor (125 kDa) and nuclear active (65 kDa) forms of SREBP-1 (Figure 3.1A) (26]). Activation of LXR by the addition of T0901317 further increased microsomal precursor and nuclear forms of SREBP-1 for each glucose concentration tested. These results show that elevated glucose and direct activation of LXR by T0901317 can independently and synergistically activate SREBP-l synthesis, processing and nuclear localization in B-cells. Since LXR regulates only SREBP-1c and not SREBP-la (279), these changes in SREBP-l likely reflect increased SREBP-1c nuclear abundance. Activation of LXR in a number of tissues results in direct and indirect induction of genes involved in lipogenesis and cholesterol efflux. Culturing INS-1 cells in 16.7 mM glucose for 48 h led to a modest increase (1.4- to 2-fold) in mRNA levels of SREBP- ], FAS, SCDl and 2, and a large increase in ACCa (6.4-fold) (Figure 3.1B). LXR activation with T0901317 markedly increased the expression of SREBP-1, FAS, SCD], SCD2, ABCAl and ABCGl irrespective of the glucose concentration. T0901317 also led to a 2.3-fold increase in ACCa gene expression in cells culture in low glucose, but did not further increase ACCa expression in cells cultured in elevated glucose. Treatment of human islets with T0901317 (10 pM) for 72 h also led to a 6- to 13-fold increase in expression of SREBP-1 , FAS, and ACCa mRNA (Figure 3.1C). 62 Figure 3.1. Glucose and LXR activation increase SREBP-1 expression and nuclear localization, and lipogenic gene expression. IN S-l cells were cultured for 48 hrs in media containing 4 or 16.7 mM glucose i 10 uM T0901317. A. Microsomes and nuclear extracts were fractionated by SDS-PAGE and SREBP-1 immunoreactivity was detected by Western analysis. Results shown are representative of four independent experiments. B. Total RNA was isolated and analyzed for SREBP-1, ACC, FAS, SCDl, SCD2, ABCAl and ABCGl mRNA expression by real-time RT-PCR. Control genes cyclophilin and ribosomal protein L32 (RPL32) were unaffected by T0901317 or glucose (data not shown). Data are relative to cyclophilin and normalized to cells cultured in 4 mM glucose (mean i SEM, n = 4). C. Isolated human islets were cultured for three days in medium supplemented without or with 10 uM T0901317. Total RNA was isolated and mRNA expression levels were determined by real-time RT-PCR (mean i SEM, n = 3). 63 A Glucose (mM): 4 16.7 T0901317(pM): O 10 0 1O Microsomal (125 kDa) - Nuclear (65 kDa) - 25' E4 mM Glucose -4 mM Glucose + T0 E16] mM Glucose E11316] mM Glucose + TO .3 01 I Fold Response a" 3.3 SREBP-1ACC FAS SCD1 SCDZ ABCA1ABCG1 Figure 3.1. Continued i. S 1 a P d. B .I E C S 0 n R C A a S A F m I § U U H u). _ u u a a M 5 0 5 o 5 0 (UV 2 2 1 1 7 22.88. >3 :03265 20”. 13.. m 9 0 T C 65 3.2. Effect of glucose and LXR activation on lipogenesis. To assess de novo lipogenesis, INS-l cells were incubated for 36 hrs in 4 or 16.7 mM glucose :1: T0901317 and then incubated for 12 hrs under the same conditions in the presence of [2-14C]acetic acid. Total lipids were analyzed by TLC to measure 14C incorporation into polar lipids (phospho- and sphingolipids), FFA, TAG, cholesterol, and cholesterol esters (264). Figure 3.2A illustrates the fractional distribution of 14C in neutral, polar, and non-esterified lipid fractions. The percent of 14C in the polar lipid fraction ranged from 88 to 71% and was sensitive to both glucose and T0901317. For cells incubated in 4 mM glucose, 83.1 i 1.5% (N = 4) of 14C-labeled lipid were polar lipids and the fractional distribution was not altered by T0901317 (82.2 i 1.4%). Treatment of cells with 16.7 mM glucose shifted the fractional 14C distribution from polar lipids (77.6 a: 2.0%) to neutral lipids, and this was further shifted by T0901317 (71.1 :t 1.3%). In cells cultured in 4 mM glucose, the fractional distribution of 14C assimilation into cholesterol, CE, FFA, and TAG was 7.7 i 0.6%, 0.6 :t 0.1%, 1.1 i 0.2%, and 1.7 i0.3%, respectively, and this was unaltered by treatment with T0901317 (Figure 3.2A). Culturing cells in 16.7 mM glucose led to a 5.4- and 2.5-fold increase in 14C-labeled TAG (9.1 :l: 1.6%, p < 0.01) and CE (1.4 i 0.3%, p < 0.03), respectively. Addition of T0901317 led to an additional 2-fold increase in 14C-labeled TAG (16.7 :t 1.9%, p < 0.01) and a ~50% reduction in 14C-labeled cholesterol (4.0 :l: 0.4, p < 0.03) and CE (0.7 i 0.1, p < 0.04), respectively. Similar changes in fractional distribution of 14C-labeled polar lipids and TAG were observed in experiments using D-[U-14C]glucose (data not shown). Changes in 14C-labeled TAG were also proportional to changes in TAG mass 66 lira lion‘- I Ill den - _._fi,_»_‘ _. Lei 3C (data not shown). Treatment of human islets with T0901317 also increased 14C-labeled TAG and reduced 14C-labeled cholesterol (Figure 3.3A). Overall, these results demonstrate that activation of lipogenic genes in B-cells by elevated glucose and T0901317 markedly increase de novo lipid synthesis and partitioning of carbon from acetic acid or glucose into complex neutral lipids, particularly TAG. 3.3. Effect of LXR activation on fatty acid profile. Total lipids extracted from [2-14C]acetic acid labeled INS—l cells were saponified and the FA profile was determined by reverse-phase HPLC. When INS-1 cells were incubated in 4 mM glucose, 4.5%, 53.1%, 23.6%, 5.7%, 5.3%, and 7.8% of 14C-labeled FAs were myristic acid (14:0) palmitic acid (16:0), stearic acid (18:0), palmitoleic acid (16:1,n-7), vaccenic (18:1,n-7) and oleic acid (18:1,n-9), respectively (Figure 3.2B). Incubation of cells in 16.7 mM glucose only modestly affected the fractional distribution of 14C-labeled FAS and this was reflected by a small increase in 14C-labeled palmitoleic acid (16:1,n-7). In contrast, treatment of cells with T0901317 led to a large change in the fractional distribution of 14C-labeled FAs such that 14C-labeled palmitoleic (16:1,n-7), vaccenic (18;l,n-7) and oleic (18:1,n—9) acids increased by ~2-fold. T0901317 also led to a ~25% reduction in 14C-labeled palmitic (16:0) and stearic (18:0) acid. The T0901317- induced change in 14C-labeled FAs occurred with a commensurate increase in MUFA pool size and decrease in saturated FA pool size (data not shown). Treatment of isolated human islets with T0901317 also changed the fractional distribution of 14C-labeled F As with an increase in palmitoleic acid (16:1,n-7) synthesis and a reduction in stearic acid (18:0) production (Figure 3.3B). These results show that activation of LXR in B-cells increases de nova synthesis of MUFA. 67 figur' 1151 T19} Figure 3.2. Elevated glucose and T0901317 increase de nova lipid synthesis in INS-1 cells. INS-l cells were cultured for 48 h in 4 or 16.7 mM glucose :1: 10 uM T0901317. Cells were then incubated overnight under the same conditions in the presence of [2-14C]-acetic acid. A. Total lipids were extracted from the cells and separated on silica TLC plates with hexanezetherzacetic acid (90:30zl). Plates were dried, and radioactivity was detected and quantified on a Phosphoimager. Data are reported as percentage of total labeled lipid. Percent of labeled polar lipid is reported as a numeric value at the top of the graph. B. Total lipids were saponified and incorporation of 14C into fatty acids was determined by reverse—phase HPLC. Data are reported as percentage of total labeled fatty acid. Values are the mean i: SEM for four independent experiments. 68 A Iias 1“c-potar lipid: 83.1 30- N (II I 20- Percent of total labeled lipid a our Glucose (mM): 4 T0901317(pM): O B 110- 100- Percent labeled fatty acid Glucose (mM): 4 T0901317(pM): 0 82.2 77.6 71.1 I -Triacylglyceride ECholesterol -Cholesterol esters -Free fatty acids 4 16.7 16.7 10 0 10 notifier: x ,5. Sc“ : 2392: 4 16.7 16.7 10 0 10 69 Figure 3.3. isles were all 5 till conditions .. ;‘ . astronaut described : intestero. acids. \"ali Figure 3.3. T0901317 increase de nova lipid synthesis in human islets. Human islets were cultured for 36 hrs in medium containing 11.1 or 22.2 mM without or with 5 uM T0901317. Islets were then incubated for 12 hrs under the same conditions in the presence of l u Ci [2-14C]-acetic acid. Assimilation and distribution of 14C into complex lipids and fatty acids species was determined as described in Figure 3.2. A. Incorporation of 14C into triacylglyceride, cholesterol, cholesterol esters, and free fatty acids. B. Incorporation of 14C into specific fatty acids. Values are means, n = 2. 70 A L70 as ”C-polar lipid: s9", smswm__ts_z__wAgo _ l 50 QTriacylglyceride f? Cholesterol l Cholesterol ester @ Free fatty acids ZN i N 20- \\ 45 . Human islets 40‘ Percent of total labeled lipid 15 - 1o - 5 - o . WW” 7% . mm W Glucose (mM): 11.1 11.1 22.2 22.2 T0901317 (pM): 0 10 0 10 B 1731420 N1621 120 518:0 79 . Human lSletS I16:O and 18:1 8 100 .__,_ *“ ";:=,:2:::2:::s:;: >s 5225;123:335? """"""" 8 . T) 60 .Q S u 40 . C (D 2 20 Q) Q. o Glucose (mM): 11.1 11.1 22.2 22.2 T0901317 (uM): 0 10 0 10 71 3.4. Impact of LXR activation on basal and glucose-stimulated insulin secretion. IN S-l cells were cultured for 48 hrs in 4 or 16.7 mM glucose d: 10 uM T0901317, after which insulin release was measured in response to a 1 hr challenge with either 2 or 20 mM glucose. Acute treatment of INS-1 cells, previously cultured in 4 mM glucose rt T0901317, with 20 mM glucose led to an ~2-fold increase in GSIS (Figure 3.4A). In comparison, IN S-l cells cultured in 16.7 mM glucose for 48 hrs had reduced basal insulin release (2 mM glucose) and GSIS. In cells cultured in 16.7 mM glucose plus T0901317, insulin release was increased when cells were stimulated with 2 or 20 mM glucose and there appeared to be a partial recovery of GSIS. Although long-term exposure of IN S-l cells to elevated glucose reduces insulin expression (282), increased insulin release from cells cultured in 16.7 mM glucose plus T0901317 was not due to increased cellular insulin content (Figure 3.4B). Moreover, treatment of cells with T0901317 did not significantly alter glucose utilization at either 2 or 16.7 mM glucose (Figure 3.4C). Similar findings were also observed for glucose oxidation (data not shown). These data indicate that LXR-activated INS-1 cells cultured under an elevated glucose load have increased basal and stimulated insulin release, and this is independent of increased insulin content or changes in acute glucose metabolism. 72 Figure 3.4. INS-1 cells cultured in elevated glucose and T0901317 have increased insulin release. INS-1 cells were cultured for 48 h in 4 and 16.7 mM glucose i 10 uM T0901317. Cells were washed and preincubated at 370C in KRB buffer containing 2 mM glucose for 60 min. A. Acute insulin release was then determined by incubating cells for 60 min at 370C in KRB buffer containing 2 or 20 mM glucose. Data represent the mean i SEM for four independent experiments performed in triplicate. *, p < 0.03 when compared to cells cultured in 16.7 mM glucose. B. Intracellular insulin content from IN S-l cells that under went an acute glucose (2 mM) challenge as described above. Data represent the mean 5: SE. of four independent experiments. *, p < 0.05 when compared to cells cultured in 4 mM glucose. C. Glucose utilization was measured by incubating cells for 30 min at 370 C in KRB buffer containing either 2 or 16.7 mM glucose and [5-3H]-D-glucose. Conversion of [5-3H]-glucose to [3H]-H20 was determined. Data represent the mean :t SE. of five independent experiments. * and #, p < 0.04 or p < 0.001, respectively, when compared to cells cultured in 4 mM glucose. 73 25- .x .- N O U! D I I I Insulin Release (ng/ml) 0| 04 Glucose (mM): T0901317 (pM): ‘1 01 O I B Insulin Content (ng/well) 0- Glucose (mM): T0901317(uM): C 1100- 1000- 900- 800- 700- 600- 500‘ 400- 300' 200- 100- Glucose Utilization (pmol/mIn/mg) 0- Glucose (mM): T0901317 (uM): E2 mM Glucose -20 mM Glucose *' 4.016.7 16.7 0 10 0 10 4.0 4.0 16.7 16.7 0 10 0 10 D2 mM Glucose -16.7 mM Glucose 4.0 4.0 16.7 16.7 0 10 0 10 74 3.5. ' this aim ill] be n To 1 Gilli dot SC by in it 3.5. MUFA synthesis is not obligatory for elevated insulin release from LXR- activated INS-l cells. Exposure of human islets to elevated glucose and exogenous palmitate induces apoptosis and diminishes GSIS, whereas these effects are reversed by co-exposure to MUFAs (283). Thus, enhanced insulin secretion from LXR-activated INS-1 cells could be mediated by the increased conversion of de novo synthesized palmitate to MUFAs. To test this hypothesis, INS-1 cells were electroporated with SCD targeted siRNA and cultured for 48 hrs in 16.7 mM glucose with or without T0901317. SCD siRNA knocked down both SCD1 and SCD2 gene expression and markedly reduced the induction of SCD1 and SCD2 by T0901317 (Figure 35A). The expression of A6D was not effected by SCD siRNA. In cells cultured with [1-14C]-palmitate during the last 24 hrs, SCD knockdown significantly reduced the conversion of 14C-labeled palmitate into MUFAs (Figure 3.5B). After culture for 48 hrs in 16.7 mM glucose with or without T0901317, insulin release from INS-l cells with reduced SCD expression was slightly increased rather than decreased (Figure 3.5C). Nevertheless, this shows that elevated insulin release from LXR-activated INS-1 cells is not dependent on the induction of SCD gene expression. 75 Figure 3.5. Knockdown of SCD with siRNA does not inhibit insulin release from LXR-activated INS-1 cells. INS-1 cells were electrophorated in the presence of 100 nM control siRNA or siRNA against SCD1 and 2. Cells were cultured for 12 hrs in 11.1 mM glucose and then cultured for 48 hrs in 16.7 mM glucose with or without T0901317. After which mRNA levels, MUFA synthesis from 14C-labeled palmitate, and insulin release were assessed. A. SCD siRNA blocked the ability of T0901317 to increase SCD l and 2 mRNA levels. SCD siRNA, however, did not affect T0901317 induction of delta 6 desaturase (D6D) (n=3). B. Knockdown of SCD decreased the conversion of 14C-palmitate (16:0) to palmitoleate (16:1) or oleate (18:1) (n=3). C. Knockdown of SCD failed to decrease insulin release from LXR-activated cells cultured in 16.7 mM glucose (11 = 6). 76 EsiCTL -siCTL + T0 msiSCD -siSCD + T0 .72 o < >. 3: -18:1n9 “’ 80- u. @18fln7 E 60- -16:1n7 g E18:0 3 40- -16:0 “E 8 20- L o a. 0- T0901317(uM): o 10 0 10 siCTL siSCD t 60" E m E 45.. .T. D 2 mM Glucose g - 20 mM Glucose 8 30- cc 2 2 .E 15- 3 U) E 0' ’1 I siCTL siSCD siCTL siSCD 16.7 mM Gluc 16.7 mM Gluc + T0 77 3.6. Role of fatty acid oxidation in elevated insulin release. To investigate the role of FA oxidation in insulin release, secretion studies were performed in the presence of an inhibitor of long-chain acyl CoA synthetase (ACS), triacsin C, or an inhibitor of camitine palmitoyltransferase-l (CPT-1), etomoxir. Triacsin C prevents conversion of FFA to LC-CoA thereby indirectly inhibiting B-oxidation of FFA, whereas etomoxir directly inhibits the rate-limiting enzyme of B-oxidation. In INS- ] cells cultured for 48 hrs in 16.7 mM glucose, etomoxir (100 pM) had no effect on insulin release in response to a 1 hr challenge with 2 or 20 mM glucose (Figure 3.6A). In contrast, the enhanced basal insulin release (2 mM glucose) from INS-1 cells cultured in 16.7 mM glucose plus T0901317 was reduced 25% (p < 0.001, N = 6) by etomoxir. In LXR-activated cells cultured in 16.7 mM glucose, triacsin C reduced basal insulin release by 38% (p < 0.001, N = 6) (Figure 3.6A). These data suggest that enhanced basal insulin release from LXR-activated cells cultured in 16.7 mM glucose requires conversion of FFA to LC-CoA and increased B-oxidation. Consistent with this possibility, [14C]palmitate oxidation under basal glucose conditions (2 mM) was elevated ~3-fold in INS-1 cells cultured for 48 h in T0901317 (Figure 3.6B). Although palmitate oxidation was suppressed by elevated glucose (20 mM), [14C]palmitate oxidation remained elevated ~3.5-fold in LXR-activated INS-l cells. Oxidation of [14C]palmitate in LXR- activated cells was also inhibited ~65% by triacsin C and ~90% by etomoxir. Increased FA oxidation in T0901317-cultured INS-1 cells correlated with increased mRNA levels of genes involved in mitochondrial B-oxidation (Figure 3.6C) including CPT-1a, camitine palmitoyl transferase-2 and long chain acyl CoA dehydrogenase, but not very long chain acyl CoA dehydrogenase. These data strongly support the hypothesis that 78 LXR activation increases basal insulin release from INS-1 cells by a mechanism involving increased B-oxidation of fatty acids. 3.7. Role of triacylglyceride turnover in elevated insulin secretion. The relationship between enhanced insulin release and the turnover of TAG was examined using the general lipase inhibitor orlistat. Treatment of INS-1 cells cultured in 4 mM glucose with orlistat (5-50 uM) increased basal insulin secretion (data not shown). The mechanism accounting for the elevated basal insulin release is unknown, but may be related to metabolic stress associated with low glucose and the inability to turnover lipid pools. Consistent with this, orlistat (50 uM) had no effect on basal insulin release from INS-1 cells initially cultured in 16.7 mM glucose 5: T0901317 (Figure 3.7A). Orlistat, however, completely blocked insulin release in response to 20 mM glucose. These results suggested that enhanced GSIS from LXR-activated INS-1 cells cultured in 16.7 mM glucose was associated with turnover of TAG. To examine TAG turnover, INS-l cells were cultured in 16.7 mM glucose and T0901317 for 48 hrs, and during the last 6 hrs were labeled with [2-14C]acetic acid. After labeling, cells were subjected to conditions mimicking an acute insulin release assay in the presence of orlistat and turnover of labeled lipid was measured. Incubation of cells for 2 hrs in 2 mM glucose led to an ~65% decrease in TAG and DAG labeling, and an ~50% decrease in FFA labeling (Figure 3.7B). Treatment of cells for 1hr in 2 mM glucose followed by 1 hr in 20 mM glucose tended to slow the turnover of TAG and DAG. Orlistat completely blocked the turnover TAG and led to a precipitous fall in labeled F FA, possibly due to FA oxidation of cellular FFA. Orlistat has also been shown to inhibit FAS (284), suggesting that orlistat may block GSIS from LXR-activated INS-l cells by inhibiting de nova lipid 79 synthesis. To test this possibility, INS-1 cells cultured for 48 h in 16.7 mM glucose :1: T0901317 were subjected to an acute insulin release assay in the presence of 2 or 20 mM glucose containing [U-14C]glucose and incorporation of 14C into FA from methanol- soluble lipids was determined. Incubation of control and LXR-activated cells for 1 h with 20 mM glucose markedly increased 14C-labeled FA and this was further increased by orlistat (Figure 3.7C), suggesting orlistat’s action on GSIS is independent of inhibition of de nova lipogenesis. These data suggest that a byproduct of TAG turnover such as DAG or FFA may participate in enhanced GSIS from LXR-activated INS—1 cells. To investigate a potential role for DAG, acute insulin release studies were performed in the presence of calphostin C, an inhibitor of protein kinase C (PKC) that competitively interferes with DAG and phorbol ester binding. GSIS from LXR-activated INS-1 cells was attenuated by calphostin C (1 uM) (Figure 3.8), but not by other PKC inhibitors - GO6976 or GO6983 (data not shown). Because calphostin C and orlistat only affected GSIS, the role for influx of calcium through the L-type voltage-gated calcium channel (L-VGCC) was tested. The L-VGCC inhibitor verapamil (100 uM) completely inhibited GSIS from LXR-activated INS-1 cells without affecting basal insulin release. 80 Figure 3.6. Elevated basal insulin release from LXR-activated INS-l cells involves increased fatty acid oxidation. INS-l cells were incubated for 48 h in 16.7 mM glucose i T0901317 (10 uM). A. Insulin release (60 min) in response to 2 or 20 mM glucose was then assessed in the presence or absence of etomoxir (100 uM) or triacsin C (10 uM). Values are mean :h SEM for six independent experiments. *, p < 0.001 when etomoxir- or triacsin C-treated cells are compared to control cells. B. FA oxidation was determined by measuring [14C]C02 production from cells incubated for l h in 2 mM glucose followed by a l h incubation with palmitic acid (50 uM), [14C]palmitic acid, and 2 or 20 mM glucose. Data are mean :t SEM for 3 independent experiments. *, p < 0.01 when cells cultured in 16.7 mM glucose plus T0901317 are compared to cells cultured in 16.7 mM glucose. #, p < 0.001 when triacsin C- or etomoxir-treated cells are compared to cells cultured in 16.7 mM glucose plus T0901317. C. Total RNA was extracted and mRNA levels for camitine palmitoyl transferase-let (CPTl), camitine palmitoyl transferase-2 (CPT2), very long chain acyl CoA dehydrogenase (VLAD) and long chain acyl CoA dehydrogenase (LCAD) were determined by real time RT-PCR. Data are mean i SEM for 3 independent experiments. *, p < 0.02 when cells cultured in 16.7 mM glucose are compared to cells cultured in 16.7 mM glucose plus T0901317. 81 120- 110-l 100- 90- 80" 70- 60-I 50- 40' 30- 20- 10- A Insulin release (ng/mg/hr O l 1000- Palmitate Oxidation (cpmlmglhr) w m o O o I 0 Fold Response ‘r’ ' 'k * D2 mM Glucose - -20 mM Glucose Control TC Eto Control TC Eto 16.7 mM Gluc 16.7 mM Gluc + TO B 2 mM Glucose * - 20 mM Glucose f" # # Control Control Triac'sin C Etonitoxir 16.7 mM Gluc 16.7 mM Gluc + T0 D16] mM Glucose * -16.7 mM Glucose + T0 CPT1 CPT2 VLAD LCAD 82 Figure 3.7. TAG turnover is required for enhanced glucose-stimulated insulin release from LXR-activated INS-l cells. A. Impact of orlistat on insulin release. INS-1 cells were incubated for 48 h in 16.7 mM glucose :t T0901317 (10 pM). Insulin release (60 min) in response to 2 or 20 mM glucose was then assessed in the presence or absence of orlistat (50 uM). Values are mean :1: SEM for six independent experiments. *, p < 0.001 when orlistat-treated cells are compared to control cells. B. Impact of orlistat on turnover of de nova-derived TAG, DAG and FFA. INS-l cells were cultured for 48 h in 16.7 mM glucose :1: T0901317 (10 pM). During the last 6 h cells were incubated with [2-14C]-acetic acid (t=0), after which cells were subjected to conditions for an acute insulin release study: 1 h incubation in 2 mM glucose followed by an 1 h incubation in 2 or 20 mM glucose. Total lipids were extracted and analyzed as described in Figure 3A. Values are mean i SEM for 3 independent experiments. Data are presented relative to 14C-labeling at t=0. Phosphoimager intensity values at t=0 for TAG are 482,621 i 59,573, for DAG are 39,461 i 4,451 and for FFA are 37,267 :1: 3,691. C. Impact of orlistat (50 pM) on de nova FA synthesis from glucose. INS-1 cells were incubated for 48 h in 16.7 mM glucose :1: T0901317, and subjected to a insulin release assay with 2 or 20 mM glucose containing 4 or 40 uCi of [U-14C]glucose, respectively. 14C-labeled FA were quantified as described in the Experimental Procedures. Values are mean i SEM of 3 independent experiments. *, p < 0.001 orlistat-treated cells are compared to control cells. 83 Insulin release (ng/mglhr) > Relative labeling Total FA Counts/pg protein o * 90- E2 mM Glucose 80' -20 mM Glucose 70- * 60- 50- 40- 30- 20- 10- o- Control Orli'stat Coritrol Orli'stat 16.7 mM Gluc 16.7 mM Gluc + T0 Et=0 -2 mM Glucose 520 mM Glucose 1_4- IIIIDZ mM Gluc + Orlistat 1.2_ -20 mM Gluc + Orlistat W TAG DAG1 ,2 FFA 1 0000- 8000- E 2 mM Glucose - 20 mM Glucose 6000- 4000- 2000- 0- Veh Veh Orlistat 16.7 mM Gluc 16.7 mM Gluc + T0 84 3(- 3(- 1: 70'! % E2 mM Glucose g 60' -20 mM Glucose g 50- a) 40- ll) 8 30- E ‘- 20- .E 3 10- U) E 0' l l I l Control Verap Cal C Control Verap Cal C 16.7 mM Gluc 16.7 mM Gluc + T0 Figure 3.8. Enhanced glucose-stimulated insulin release from LXR-activated INS-1 cells is attenuated by verapamil or calphostin C. Insulin release (60 min) in response to 2 or 20 mM glucose was then assessed in the presence or absence of verapamil (100 uM) or calphostin C (1 uM). Values are mean i SEM for six independent experiments. *, p < 0.01 when verapamil- or calphostin C-treated cells are compared to control cells. 85 Discussion Culturing INS-1 cells in elevated glucose led to increased nuclear SREBP-1c, lipogenic gene expression, TAG synthesis, and loss of GSIS. These findings are consistent with reports that SREBP-lo activation, either by elevated glucose or over- expression, in B-cell lines or islets increased lipogenic gene expression, TAG synthesis and decreased GSIS (114-116, 139, 235, 268, 269, 285). Compared to INS-l cells cultured in elevated glucose, LXR-activated INS-1 cells had significantly elevated microsomal and nuclear forms of SREBP-1c and lipogenic gene expression particularly SREBP-1, FAS, SCD1 and SCD2. These findings are consistent with the role of LXR in regulating SREBP-1 gene transcription, and LXR and SREBP-lo in regulating FAS, SCD1 and SCD2 gene transcription (21, 286). Of the lipogenic genes examined only ACCa mRNA levels were more strongly induced by elevated glucose than by LXR activation. Glucose has been reported to bind and activate LXR (77), but this does not appear to play a prominent role in INS-l cells because elevated glucose did not induce expression of LXR target genes including ABCA1 and ABCGl. As expected, increased lipogenic gene expression in LXR-activated INS-1 cells cultured in elevated glucose markedly increased de nova neutral lipid synthesis. Because LXR activation only affected lipid synthesis and insulin secretion in cells cultured in elevated glucose suggests that the two events are linked. This is likely an adaptive effect because it has recently been shown that metabolic flux through lipogenic pathways is not required for normal GSIS (287, 288). Hypersecretion of insulin, however, has been shown to involve the induction of SREBP-lo and enhanced lipid synthesis in mouse islets cultured in elevated glucose (234). 86 Fatty acid D9 desaturases (SCD1 and SCD2) function as the rate-limiting step for MUFA synthesis and play an integral role in neutral lipid (TAG and CE) synthesis (21). In agreement with increased SCD1 and SCD2 mRNA levels, LXR-activated INS-1 cells exhibited a 2-fold increase in de nova derived MUFA (16:1,n-7, 18:1,n-7, 18:1,n-9) and a commensurate drop in synthesis of saturated FA (16:0, 18:0). The increase MUFA synthesis in LXR-activated INS-1 cells cultured in elevated glucose corresponded with increased MUFA mass (data not shown). Chronic exposure of islets and B-cell lines to oleic (18:1,n-9) or vaccenic (18:1,n-7) acid have been reported to increase basal insulin release (289-291), suggesting that increased MUFA synthesis in LXR-activated cells might be directly involved in basal insulin release. To test this possibility, siRNA targeting SCD1 and SCD2 were introduced into LXR-activated INS-1 cells. SCD1/2 siRNA effectively decreased SCD1 and SCD2 mRNA levels and MUFA synthesis, but did not lower insulin release from LXR-activated INS-l cells. These data indicate that increased de nova MUF A synthesis is not an obligatory step for enhanced insulin release from LXR-activated INS-l cells, but likely facilitates neutral lipid synthesis. Enhanced basal insulin release from LXR-activated INS-1 cells was attenuated by triacsin C and etomoxir, indicating a role for increased acyl-CoA formation and FA oxidation. Under our experimental paradigm for insulin release studies, INS-1 cells are first preincubated for 1 hour in 2 mM glucose followed by incubation for 1 hour in either 2 or 20 mM glucose. During this timeframe, newly synthesized TAG is rapidly turned over (Figure 3.7) and likely serves as the source of the F FA for acyl-CoA formation and oxidation. INS-1 cells cultured in elevated glucose also have increased de novo synthesized TAG, but do not have elevated basal insulin release. This suggests that INS- 87 1 cells cultured in elevated glucose either do not synthesize sufficient quantities of TAG to sustain increased basal insulin release or that LXR activation stimulates additional pathways associated with lipid metabolism. Consistent with the later, LXR activation was shown here to increase FA oxidation and this correlated with increased expression of genes involved in mitochondrial B-oxidation particularly CPT-lat. Recently, Colin et al. showed that activation of LXR with synthetic agonists induced PPARCX and subsequently its target CPT-l in the intestine, but not the liver (292). This suggests that LXR agonists may also induce CPT-l through PPARa in B-cells. Alternatively, LXR-activation of INS-1 cells may increase de nova synthesized FA to levels sufficient to induce CPT-lat. This possibility is supported by the observation that long-term exposure of IN S-l cells to long-chain FA increases CPT-1 gene expression and FA oxidation (290, 293). Our findings also raise the possibility that LXR activation can protect the B-cell from glucose toxicity by shuttling glucose toward FA, which can be oxidized immediately or after release from TAG. Lipolysis of intracellular TAG and the subsequent generation of lipid signaling molecules including FFA, acyl-CoA and DAG have been proposed to mediate GSIS (reviewed in (99)). TAG turnover produces a FFA and a predominantly less biologically active sn2,3-DAG species, which can be further broken down to monoacylglyceride, glycerol and FFA (294-296). These later products, along with de nova derived FA, can be reincorporated into biologically active snl,2-DAG species through a glycerolipid/F F A cycle (297). Based on this, we hypothesize that enhanced GSIS from LXR-activated INS-1 cells results from elevated lipolysis and formation of lipid products that can directly serve as signaling molecules (e.g. FFA) or used as substrates for the 88 glycerolipid/FFA cycle to generate snl,2-DAG. Consistent with this hypothesis, the ’ general lipase inhibitor orlistat blocked turnover of de nova derived TAG and GSIS in LXR-activated INS-1 cells, but did not block de nova synthesis of FA (Figure 3.7). Mulder et al. have also proposed that orlistat attenuates GSIS by blocking the formation of an acylglyceride-coupling factor (164). If the glycerolipid/FFA cycle is involved in enhanced GSIS from LXR-activated INS-1 cells, one would predict that inhibition of acyl-CoA formation with triacsin C would have also blocked GSIS, which did not occur (Figure 3.6). This might be due to the inability of triacsin C to inhibit all ACS isoforms (298) and that triacsin C is more efficacious at inhibiting FA oxidation than lipid synthesis in B-cells (299). It remains a possibility that enhanced GSIS from LXR- activated INS-1 cells might also involve turnover of phospholipids and direct production of snl,2-DAG (190). Polar lipid turnover, however, was much slower than TAG turnover in INS-l cells and not effectively blocked by orlistat (data not shown). DAG generated from the glycerol/FF A cycle might serve as the coupling factor to enhance GSIS from LXR-activated IN S-l cells. Classic (at, [31, 011, y) and novel (5, e, r], 0) isoforms of PKC are activated by DAG in Ca2+-dependent and —independent manners, respectively. Pharmacologic inhibition of many of these PKC isoforms with GO6976 (inhibits PKCa, [31) and GO6983 (inhibits PKCa, [3, y, 6, 1;), however, did not attenuate GSIS from LXR-activated INS-1 cells (data not shown). Calphostin C, which competitively blocks DAG-binding sites on classic and novel PKC isoforms, PKD (PKCu) and DAG-binding proteins, significantly attenuated GSIS from LXR-activated INS-1 cells (Figure 3.8). Blockade of the influx of extracellular Ca2+ with the L-VGCC inhibitor also completely abrogated GSIS from LXR-activated INS-l cells. Taken as a 89 whole, these data suggest that enhanced GSIS from LXR-activated IN S-l cells does not involve classic or novel PKC isoforms, but involves activation of a DAG-binding protein that is calcium-dependent or mediates biochemical events upstream from the influx of calcium. There are a number of families of DAG-binding proteins that could be involved including PKD (PKCu), chimaerins, RasGRPs, MUNCl3s or DAG kinases (reviewed in (154)). Further experimentation is necessary to determine the exact DAG-binding protein(s) involved. Straub and Sharp have proposed a similar mechanism to explain how FA depletion of rat islets caused large increases in GSIS (300). In their model, FA depletion is proposed to cause lipid remodeling or increase breakdown of intracellular TAG, which increases DAG production, activates a DAG-binding protein and augments GSIS. Enhanced insulin release from LXR-activated B-cells has been reported to be associated with increased mRNA levels of de-l, insulin and GLUT2 (253) suggesting a role for LXR or SREBP-1c in augmenting B-cell phenotype and glucose sensing. SREBP-l is also required for elevated glucose to increase mRNA levels of de-l and genes involved in glucose sensing including GLUT2 and glucokinase (234). Similar changes in gene expression may play a role in enhanced insulin release from LXR- activated INS-1 cells. Nevertheless, this seems unlikely because glucose utilization and insulin content were not significantly increased in LXR-activated INS-1 cells. In conclusion, our study shows that LXR activation of INS-1 B-cells exposed to elevated glucose increases TAG synthesis; and subsequent TAG turnover can lead to the production of lipid signaling molecules resulting in elevated insulin release. Similar 90 mechanisms may account for the ability of SREBP-1c to establish hypersecretion of insulin in some models of hyperglycemia. 91 Chapter 4. Stearoyl-CoA Desaturase Modulates Palmitate-Induced Endoplasmic Reticulum Stress and Apoptosis in Pancreatic fi-Cells Abstract Chronic elevations in exogenous free fatty acids (FF As) have been implicated in the pathogenesis of B-cell failure and the development of type 2 diabetes. The effects of exogenous FF A, particularly saturated fatty acids (FAS), on B-cells include endoplasmic reticulum (ER) stress and downstream induction of apoptosis. Regulation of monounsaturated FA (MUFA) synthesis through altered FA desaturase and elongase gene expression may serve to protect B-cells from exogenous saturated FAS. In the Zucker diabetic fatty (ZDF) rat model of progressive [El-cell failure, islets from 6-week old pre- diabetic ZDF rats showed a 1.5- to 2.3-fold induction in the stearoyl-CoA desaturases (SCD) l and 2 mRNA, respectively, compared to control rats. At 13 weeks of age, ZDF rats were hyperglycemic and exhibited decreased plasma insulin levels, an indicator of B- cell dysfunction. This was associated with markedly decreased mRNA levels of insulin, SCD1, SCD2 and Elovl-6, which elongates 16:0 to 18:0 and 16:1,n-7 to 18:1,n-7. These findings suggested enhanced expression of SCD1/2 and other FA modifying genes may protect B-cells from damage caused by exogenous saturated FAs. Next, siRNAs and adenoviral constructs were used to investigate the role of altered SCD and Elovl-6 expression in IN S-l fi-cells exposed to exogenous palmitate. Knockdown of SCD gene expression decreased conversion of palmitate to MUFA and increased the susceptibility to palmitate-induced ER stress, as measured by splicing of Xbpl, induction of ATF 3 and 92 CHOP, and JNK phosphorylation. Palmitate-induced apoptosis was also increased by SCD knockdown, as shown by elevated caspase-9 cleavage and DNA fragmentation. Over-expression of SCD2 increased synthesis of n-7 MUFAs and markedly reduced the ER stress and apoptosis induced by palmitate. Elovl-6 knockdown decreased palmitate elongation and tended to reduce palmitate toxicity, whereas Elovl-6 over-expression increased palmitate elongation to stearate (18:0) and increased susceptibility to palmitate- induced JNK phosphorylation and apoptosis. In addition, elevated ER stress in INS-1 cells with decreased SCD expression involved reduced palmitate incorportation into TAG and activation of Ca2+-dependent protein kinase Cs (PKCs). These findings demonstrate that altered expression and activity of SCD2 and Elovl-6 modulate the susceptibility of B- cells to the toxic effect of saturated F As. Introduction Type 2 diabetes arises from an inability of pancreatic B-cells to compensate for insulin resistance in peripheral tissues. The progressive loss of B-cell compensation is likely due to reduced insulin secretory capacity or B-cell mass (301-305). Elevated levels of plasma non-esterified free fatty acid (FF A), a risk factor for insulin resistance and type 2 diabetes (201, 202), have been associated with the pathogenesis of B-cell dysfunction (4, 203). The response of fi—cells to long-term elevations in fatty acids (FAs), however, is largely dependent on the FA composition. Saturated FAS, such as palmitate (16:0), cause diminished insulin secretion and insulin gene expression and the induction of apoptosis through multiple processes, including generation of ceramides, reactive oxygen species, and endoplasmic reticulum (ER) stress (223, 237, 306-308). Monounsaturated PAS 93 (MUFAs), such as palmioleate (16:1,n-7) and oleate (18:1,n-9), and the polyunsaturated FA (PUFA) eicosapentaenoate (20:5,n-3) can protect B-cells from apoptosis and insulin secretory defects induced by saturated FAs (236, 283, 308). In addition to exogenous FA structure, evidence has demonstrated that the intracellularcapacity to modulate FA fate has an important role in B-cells. Alterations in FA metabolism critically affect the response of B-cells to exogenous FAs, particularly the lipotoxicity of saturated FAs. Approaches used to enhance FA oxidation and triacylglyceride (TAG) storage have demonstrated significant alterations in the effects of exogenous saturated FAs on B-cell function (309-311). Studies have also shown that regulation of FA structure may participate in modulating the effects of FAS on B-cellsi Subpopulations of MIN-6 and rat B-cells identified to be resistant to palmitate-induced apoptosis were associated with increased expression of the FA delta-9 desaturase, stearoyl-CoA desaturase (SCD) 1, and hence increased conversion of palmitate to MUFAs (6, 257). The level of SCD1 gene expression has also been correlated with the susceptibility of B-cell lines and islets to ER stress in vitra and with the severity of diabetes in viva in a mouse model of obesity (256, 312). FA desaturase and elongase enzymes modify fatty acids by adding a cis-double bond or two-carbons to a fatty acyl-CoA, respectively. These activities are essential for a variety of cellular functions, including maintenance of membrane FA composition and generation of signaling molecules. The desaturase subtypes in mammals include delta 5 desaturase (ASD), delta 6 desaturase (A6D), and SCD. Isoforms of SCD include four in mouse (SCD1-4) (1 1-14), two in rat (SCD1 and 2) (15), three in hamster (SCD1—3) (16), and three in human (SCD1, 2, and 5) (17-19). FA elongase (Elovl) subtypes range from 94 Elovl-l to 7 in mouse, rat, and human (www.cnsembl.org). Synthesis of PUFAs from essential dietary FAs occurs through the desaturases A5D and A6D and the elongases Elovl-2 and Elovl-5 (7, 31, 32). SCD, the rate-limiting enzyme in C16 and C18 MUFA synthesis, and Elovl-6 synthesize the MUFAs oleate, palmitoleate, and vaccenate (18:1,n- 7) (20, 31). Elovl-5 can also elongate palmitoleate to vaccenate (32). Elovl-1, -3, and -4 elongate a broad array of very long chain FAs (>C20) and are involved in sphingolipid synthesis, brown adipose and skin barrier function, and retinal degeneration, respectively (25-30). Unique roles for these enzymes in pancreatic B-cells, however, remain to be defined. In this study, we first characterized FA desaturase and elongase gene expression in rat islets and IN S-l B-cells. Next, using the Zucker diabetic fatty (ZDF) rat model, FA desaturase and elongase genes were identified to be differentially expressed between pre— diabetic and diabetic ZDF rat islets. Specifically, SCD1 and SCD2 were increased in pre- diabetic islets and, along with Elovl-6, reduced in diabetic islets. Thus, we hypothesize that regulation of genes required for MUFA synthesis may significantly affect B-cell compensation and failure in the pathogenesis of T2D. The results demonstrate that altered expression of SCD2 and Elovl-6 in INS-1 cells modulate the effects of exogenous palmitate on ER stress and apoptosis. In addition, enhanced ER stress in the absence of SCD1 and SCD2 expression may involve altered FA partitioning into neutral lipids and activation of protein kinase C (PKC). 95 Results 4.1. Rat islet and INS-1 cell FA elongase and desaturase gene expression profiles. To determine which FA elongase and desaturase genes are expressed in B-cells, elongase and desaturase mRNA levels were characterized in rat islets and INS-l cells under non-stimulatory glucose conditions. In rat islets and INS-1 B-cells, mRNA expression was detected for the FA elongases Elovl-l, Elovl-2, Elovl-4, Elovl-5, Elovl-6 and Elovl-7, and the FA desaturases SCD1, SCD2, A5D and A6D (Figure 4.1A-D). There was no difference between A5D and A6D mRNA levels in either rat islets or INS-1 cells. For SCD mRNA levels, however, the expression of SCD2 was greater than 4-fold higher than SCD1 in both rat islets and INS-l cells. This suggests that SCD2 may have a larger role in B-cells, whereas SCD1 is highly expressed in tissues with large lipid storage capacities such as adipose and liver. 96 Figure 4.1. Fatty acid elongase and desaturase gene expression in rat islets and INS-l cells. Total RNA was isolated from Sprague-Dawley rat islets (A and B) and INS-l cells (C and D) and analyzed for Elovl-1 to 7, SCD1, SCD2, D5D, and D6D mRNA expression by real-time RT-PCR. Gene expression is reported relative to RPL32 and represents mean :t SEM for three independent experiments. 97 °-°5' Rat Islets 0.04- - 0.03-1 0.02- ' 0.01- Relative Expressron (mRNA) > Elovl-1 Elovl-2 Elovl-3 Elovl-4 Elovl-5 Elovl-6 Elovl-7 0.077 Rat Islets P O a: I 0.05- ' 0.04- 0.03- 0.02- 0.01- Relative Expressron (mRNA) w 0.00- SCD1 SCD2 05D Figure 4.1. Continued C All-04' INS-1 Cells .0 0 w I 0.02! 0.01- Relative Expression (mRNA 0 Elovl-1 Elovl-2 Elovl-3 Elovl-4 Elovl-5 Elovl-6 Elovl-7 0-20' INS-1 Cells 9 .3 01 I 0.10- 0.05- Relative Expression (mRNA) U 0.00- SCD1 SCDZ 4.2. FA elongase and desaturase gene expression in ZDF rat islets. Before 10 weeks of age ZDF rats are pre-diabetic, displaying obesity and insulin resistance while maintaining normal glucose levels (313). Here, 6-week-old ZDF rat blood glucose levels were normal and plasma insulin levels were elevated (Supplementary Table 2), which coincided with increased islet insulin mRNA expression (Figure 4.2A and B). ZDF islet expression of Elovl-5, SCD1, and A6D were modestly increased from 1.4- to 1.6-fold compared to control rats at 6 weeks. Expression of SCD2, however, was increased fitrther to 2.3-fold over control. After 13 weeks of age, ZDF rats were hyperglycemic and both plasma insulin and insulin mRNA levels were diminished, possibly indicating islet failure. Except for A5D and A6D, which decreased with age in both control and ZDF rat islets, a large 60% decrease in expression was found for Elovl-6 and SCD2 in ZDF islets from 6 to 13 weeks. These results suggest the expression of enzymes involved in monounsaturated fatty acid synthesis may have an important role in islet function. 100 Table 4.1 Physiolggical parameters of 6 and 13 week old Control (fal?) and ZDF rats Control (fa/?) ZDF (Leprfa/Crl) 6 week 13 week 6 week 13 week Body weight (g) 173.8 330.4 179.3 353.5 +/- 9.0 +/- 5.8 , ** +/- 4.0 +/- 11.1 , ** Insulin (ng/ml) 2.0 3.6 9.2 2.6 +/- 0.3 +/- 1.1 +/- 1.4 , "‘ +/- 0.5 , ** Glucose (mg/d1) 83.0 80.6 117.0 316.7 +/- 3.5 +/— 4.0 +/- 5.0 , * +/- 25 , * , *"‘ Data are mean +/- SEM. Glucose values are fed blood glucose. *, p < 0.006 compared to Zucker control age matched. **, p < 0.008 compared to 6 week old with same phenotype. 101 Figurt ll W in in?) nll\' MCI Figure 4.2. Differential expression of fatty acid elongase and desaturase genes in pre-diabetic and diabetic ZDF rat islets. A and B. Total RNA was isolated from control (fa/1’) and ZDF (Leprfa/Crl) rat islets at 6 and 13 weeks of age and analyzed for insulin, Elovl-2, Elovl-5, Elovl-6, SCD1, SCD2, D5D, and D6D mRNA expression by real-time RT-PCR. Data are reported relative to cyclophilin and represent mean i SEM for five or six animals per group. *, p < 0.04 and #, p < 0.03 when compared to 6 week control and ZDF islets, respectively. **, p < 0.04 when compared to 13 week control islets. 102 > E 6 week 1000- 0.014 A -13 week i < 0.012- E 800- * E 0.0104 * 8 * '13 60°" 0.008- aa * a X 400- 0.006' ** ”J # d) 3 ,,,. 0.004- 5 200- # <0 0.002- # ** 05 ** I 0- Control ZDF Control ZDF Control ZDF Control ZDF Insulin Elovl-2 Elovl-5 Elovl-6 0.06- * A 0.05- g I g 0.04- * S g 0.03- 2 # g- 0.02- m * 113 .g 0.01- * # # 2 # * ** *7: d) I! 0.00- - Control ZDF Control ZDF Control ZDF Control ZDF SCDl SCD2 D5D DGD 4.3. Knockdown of SCD and Elovl-6 gene expression modulate MUFA synthesis. To determine if SCD and Elovl-6 expression alter FA metabolism in B-cells, siRNAs were introduced into INS-l cells and examined for effects on palmitate metabolism. In cells cultured for 36 hrs in INS-l media, SCD siRNA effectively reduced both SCD1 and SCD2 mRNA levels compared to control (siCTL), but resulted in increased Elovl-6 mRNA (Figure 4.3A). Elovl-6 siRNA reduced Elovl-6 mRNA but did not affect SCD1 or SCD2. As a control, neither SCD nor Elovl-6 siRNAs affected Elovl- 5 mRNA levels. Effects of siRNAs on FA metabolism were determined by culturing INS-l cells for 12 hrs in 400 uM palmitate plus [1-14C]palmitic acid. Total lipids were extracted, saponified, and the FA profile analyzed by reverse-phase HPLC. In IN S-l cells with control siRNA, 14C-labeled palmitate was distributed by 83.9, 6.7, 6.4, 1.0, and 1.9% into palmitic acid (16:0), stearatic acid (18:0), palmitoleic acid (16:1,n-7), vaccenic acid (18:1,n-7), and oleic acid (18:1,n-9), respectively (Figure 4.3B). SCD siRNA significantly reduced the ability to convert palmitate into MUFA, as the fractional distribution of 14C-labeled 16:1,n-7, 18:1,n-7, and 18:1,n-9 decreased between 66 and 97%. This resulted in increased accumulation of 14C-labeled palmitic acid and the ratio of saturated FAs to MUFAs (data not shown). Elovl-6 siRN A caused a marked decrease in palmitate elongation, as shown by an approximate 50% reduction in the fractional distribution of 14C-labeled 18:0 and 18:1,n-9. This led to a 1.5-fold increase in labeled 16:1,n-7. These results show that knockdown of SCDs or Elovl-6 effectively decreases the respective desaturation and elongation of palmitate and alters MUF A synthesis. 104 Figure 4.3. Modulation of SCD and Elovl-6 gene expression alters synthesis of specific MUFAs species derived from exogenous palmitate. A. Effect of SCD2 and Elovl-6 siRNAs an mRNA levels. INS—1 cells were electroporated with siRNAs for control (siCTL), SCD (siSCD) or Elovl-6 (siElovl-6) and cultured for 36 hrs in INS-1 media. Levels for Elovl-5, Elovl-6, SCD1 and SCD2 mRNA were normalized to RPL32 mRNA and reported as fold expression relative to siCTL .treated cells. Data are mean :1: SEM for three independent experiments. *, p < 0.02 when compared to siCTL cells. B. Effect of SCD and Elovl-6 siRNAs on conversion of exogenous palmitate to MUFA. INS-1 cells electroporated with siRNAs for siCTL, siSCD, or siElovl-6 were treated with INS-l media containing 400 uM palmitate and [1-14C]-palmitic acid for 12 hrs. Total lipids were extracted, saponified, and incorporation of 14C into FAs was determined by reverse-phase HPLC. Data represent the percent of labeled FA. Values are the mean 3: SEM for three independent experiments. 105 Fold Response EElovl-S 25., -Elovl-6 ESCD1 2.0_ -SCD2 * 0.5- 0.0- 110- .3 O O l (O O l Percent Palmitate Incorporation 01 ? siCTL /////}}////1 / '.' L -18:1n9 —‘ mwnm -16:1n7 E18zo -16:0 siSCD siElovl-6 106 4.4. Susceptibility to palmitate-induced ER stress is increased by SCD knockdown. Loss of B-cell function upon exposure to exogenous FFAs, particularly saturated FAs, involves activation of the ER stress response pathways inositol requiring ER to nucleus signal kinase (IRE)1, PKR-like ER kinase (PERK), and, to a lesser extent, activating transcription factor (ATF)6 (204). In ZDF islets, the response to exogenous FFAs could be affected by altered SCD and Elovl-6 gene expression, described in Figure 4.2. To test whether reduced SCD or Elovl-6 expression affects the [El-cell response to exogenous FFAs, INS-1 cells treated siRNAs were cultured for 9 hrs with 0, 200, or 400 uM palmitate and examined for the induction of ER stress. In control cells, only 400 uM palmitate induced a 2.3- to 4-fold increase in splicing of X-box binding protein 1 (Xbpls) and mRNA levels of ATF3 and CHOP, markers of IREl and PERK activation, respectively (Figure 4.4A). Palmitate induction of CHOP mRNA levels corresponded with a 5.5-fold increase in CHOP protein (Figure 4.4B). In IN S-l cells treated with 200 MM palmitate, decreased SCD expression significantly increased the sensitivity to Xbpl splicing and induction of ATF3 and CHOP mRNA by approximately 2-fold. Compared to control cells, induction of ATF3 mRNA, CHOP mRNA, and CHOP protein by 400 pM palmitate was increased further by SCD siRNA. In contrast, decreased Elovl-6 expression tended to reduce Xbpl splicing and CHOP protein levels induced by palmitate at 400 uM, but it was not significant. IREl also mediates phosphorylation of Jun N-terminal kinase (JNK) (245), which can lead to enhanced CHOP expression and apoptosis (204). In B-cells treated with F FAs, INK was phosphorylated prior to the induction of CHOP expression (314). INS-l cells treated for 6 hrs with 400 uM palmitate displayed increased JNK phosphorylation, 107 and this was markedly increased 2-fold by decreased SCD expression (Figure 4.4C). These findings demonstrate that susceptibility to palmitate-induced ER stress is enhanced in [ii-cells with a reduced capacity to synthesize MUFAs. 4.5. SCD knockdown impacts susceptibility to palmitate-induced apoptosis. INS-1 cells with decreased SCD and Elovl-6 expression were then examined to determine whether early changes in sensitivity to ER stress correlated with the induction of apoptosis. In response to increasing concentrations of palmitate, control cells treated for 24 hrs with 300 and 400 uM palmitate had increased caspase-9 cleavage and DNA fragmentation, markers of apoptosis (Figure 4.5A and B). The susceptibility to both caspase-9 cleavage and DNA fragmentation in cells with decreased SCD expression were significantly increased at 200 uM palmitate. Compared to control cells, the induction of apoptosis was further increased at 300 and 400 uM palmitate by SCD knockdown, whereas knockdown of Elovl-6 did not affect either apoptotic marker. Thus, this data confirms a recent report that palmitate induced lipotoxicity is increased by simultaneous knockdown of both SCD1 and SCD2 (257). 108 Figure 4.4. Sensitivity to palmitate-induced ER stress is increased by SCD knockdown. INS-1 cells electroporated with siRNAs for control (siCTL, C), SCD (siSCD, S), or Elovl-6 (siElovl-6, B) were treated for 9 hrs with modified INS-l media containing increasing palmitate concentrations. A. Effect of knockdown of SCD and Elovl-6 on levels of spliced Xbpl (Xbpls), Xbpl total (Xbplt), ATF3 and CHOP mRNA. Data are mean i SEM for three independent experiments and are expressed relative to siCTL cells. * and #, p < 0.02 when compared to 0 uM siCTL cells and siCTL cells at the same palmitate concentration, respectively. B. Effect of SCD or Elovl-6 siRNA on CHOP protein levels. Whole cell protein extracts were fractionated by SDS-PAGE and CHOP protein was analyzed by Western blotting. Data are mean :2 SEM for four independent experiments. * and #, p < 0.03 when compared to 0 pM siCTL cells and siCTL cells at 400 uM palmitate, respectively. C. Effect of SCD and Elovl-6 siRNA on JNK phosphorylation. INS-1 cells electroporated with siRNAs for control (siCTL, C), SCD (siSCD, S) or Elovl-6 (siElovl-6, E) were treated for 6 hrs without or with 400 uM palmitate. Phosphorylated and total JNK (pJNK and JNKt) were analyzed by Western blotting. Data are mean d: SEM for three independent experiments. * and #, p < 0.03 when compared to 0 uM siCTL cells and siCTL cells at 400 uM palmitate, respectively. 109 A 4' EsiCTL A * -siSCD E 3. * -siElovl-6 ‘5 \‘ o x U \ 2 2. § a s < x z 1. \ 0! \ E x x .\ 0- 16:0 (11M): o 200 400 o 200 400 Xbp1s Xbp1t 10' :lsicrL _siSCD 8' # .siElovl-G l/I/I/I/I/I/I/A =31: 110 Figure 4.4. Continued 3 16:0 (um: o 200 400 c s E c s E c s E CHOPl —- Actin l—-—'—--————l 18- . ESICTL # -siSCD 14' -siElovl-6 .3 a) I _l A O N I I h a: I I 2- CHOP Protein (Fold Control) CO 0 16:0 (uM : C 16:0(uM): o 400 s E c s E pJNKl - ~ 2 l JNK: |.—. :::: 7:: --- 35- |:|sicrL -siSCD €30“ -siElovl-6 # *5 25- 0 20.. pJNK (Fold e a 0| I 111 Figure 4.5. Suceptibility to palmitate-induced apoptosis is increased by SCD knockdown. IN S-l cells electroporated with siRNAs for control (siCTL, C), SCD (siSCD, S), or Elovl-6 (siElovl-6, B) were treated for 24 hrs with increasing palmitate concentrations. A. Effect of SCD and Elovl-6 siRNA on caspase-9 cleavage. Cleaved caspase-9 proteins were analyzed by Western blotting. Results shown are representative of six independent experiments. B. Effect of SCD and Elovl-6 siRNA on DNA fragmentation as determined by ELISA. Data represent fold induction and are mean :1: SEM for six independent experiments. *, p < 0.02 when compared to 0 pM Luc. #, p < 0.04 when compared to siCTL at the same palmitate concentration. 112 16:0 (11M): 0 200 300 400 Cleaved 38kDa " Caspase-Q 17kDa __. __.......— Actinl ‘-—--—-——~—_____.._._] 16:0 (pm: 0 200 300 400 C E C E C E C E Cleaved 38kDa Caspase-9 17kDa __ Actin I———-——-————-—-~———J 25 :lsicrL # : -siSCD .2 E 2° -siElovl-6 3i .9 = *5 d) E 3 U) C N _ LT. E O :1, e D 0 16:0 (11M): 0 200 300 400 113 4.6. Over-expression of SCD2 and Elovl-6 differentially modulate MUFA synthesis and markers of ER stress. Naturally occurring elevated expression of SCD1 and SCD2 in rat islets and [3- cells coincides with reduced palmitate-mediated lipotoxicity, presumably due to SCD1 activity (6, 238). In rat islets and B-cells, however, the SCD2 isoform is expressed much higher than SCD1 (Figure 4.1 and 4.2) (257). To test whether enhanced SCD2 and Elovl- 6 gene expression alone affect B-cell FA metabolism and ER stress, adenoviral constructs were used to over-express the respective genes in INS-1 cells treated with palmitate. In cells over-expressing luciferase (Luc) and cultured for 12 hrs in 400 uM palmitate plus [1-14C]palmitic acid, 14C-labeled palmitate was distributed by 71.5, 10.6, 10, 2.5, and 5.3% into palmitic acid (16:0), stearic acid (18:0), palmitoleic acid (16:1,n-7), vaccenic acid (18:1,n-7), and oleic acid (18:1,n-9), respectively (Figure 4.6A). Over-expression of SCD2 significantly increased palmitate conversion to MUFAs, as the fractional distribution of 14C-labeled 16:1,n-7 and 18:1,n-7 was increased 1.3- and 2.1-fold, respectively. This resulted in a marked increase in the accumulation of n-7 rather than n- 9 MUFAs (data not shown). Elovl-6 over-expression, however, significantly increased palmitate elongation products, as the distribution of 14C-labeled 18:0 and 18:1,n-9 were increased 2.3- and 1.8-fold, respectively. These results demonstrate that in [fl-cells over- expression of SCD2 preferentially drives synthesis of n-7 MUFAs, whereas over- expression of Elovl-6 drives synthesis of stearate (18:0) and oleate (18:1,n-9). Next, IN S-l cells over-expressing either SCD2 or Elovl-6 were treated with 400 uM palmitate and examined for activation of ER stress. In contrast to SCD knockdown, over-expression of SCD2 resulted in a marked 56% reduction in CHOP protein levels 114 after 9 hrs of palmitate treatment compared to control cells (Figure 4.68). Palmitate— induced JNK phosphorylation at 6 hrs was also reduced 40% by SCD2 over-expression (Figure 4.6C). Although Elovl-6 over-expression did not alter CHOP protein, it resulted in a significant 1.6-fold increase in JNK phosphorylation in palmitate treated cells compared to control cells. Together, this shows that enhanced SCD2-mediated synthesis of n-7‘ MUFAs protects from ER stress induced by exogenous palmitate, whereas ER stress is potentiated by enhanced Elovl-6 expression. 4.7. Effects of SCD2 and Elovl-6 over-expression on palmitate-induced apoptosis. INS-l cells with elevated SCD2 and Elovl-6 gene expression were then treated for 24 hrs with increasing concentrations of palmitate and monitored for apoptosis. Control cells over-expressing luciferase exhibited significant capase-9 cleavage at 400 uM palmitate and a dose-dependent increase in DNA fragmentation at 200, 300, and 400 uM palmitate (Figure 4.7A and B). Over-expression of SCD2 showed a marked reduction in cleaved caspase-9 and a 30-60% decrease in DNA fragmentation at each FA concentration tested compared to control cells. Elovl-6 over-expression increased both apoptotic markers at intermediate levels of palmitate but not at the 400 uM level. These results provide the first direct evidence that enhanced SCD2 and Elovl-6 expression modulate lipotoxicity in B-cells. 115 Figure 4.6. SCD2 and Elovl-6 over-expression on MUFA synthesis and palmitate-induced ER stress. A. Effect of SCD2 and Elovl-6 over-expression on MUFA synthesis. INS-1 cells treated with Ad-CMV-Luciferase (Luc), Ad—CMV- SCD2 (SCD2), or Ad-CMV-Elovl-6 (Elovl-6) were treated with INS-1 media containing 400 uM palmitate and [l-l4C]-palmitic acid for 12 hrs. Total lipids were extracted, saponified, and incorporation of 14C into F As was determined by reverse- phase HPLC. Values are the mean 3: SEM for three independent experiments. B. Effect of SCD2 and Elovl-6 over-expression on CHOP protein levels. INS-l cells treated with Ad-CMV-Luciferase (Luc, L), Ad-CMV-SCDZ (SCD2, S) or Ad-CMV- Elovl-6 (Elovl-6, B) were treated for 9 hrs with INS-l media containing increasing palmitate concentrations, after which CHOP protein was analyzed by Western blotting. Data are mean :t SEM for six independent experiments. * and #, p < 0.02 when compared to control (Luc) cells treated with 0 or 400 uM palmitate, respectively. C. Effect of SCD2 and Elovl-6 over-expression on JNK phosphorylation. INS-1 cells treated with Ad-CMV-Luciferase (Luc, L), Ad-CMV- SCD2 (SCD2, S) or Ad-CMV-Elovl-6 (Elovl-6, E) were treated for 6 hrs without or with 400 uM palmitate. Phosphorylated and total JNK (pJNK and JNKt) were analyzed by Western blotting. Data are mean i SEM for three independent experiments. * and #, p < 0.03 when compared to control (Luc) cells treated with 0 or 400 uM palmitate, respectively. 116 C '3 100‘ .\\\\\ g 90. iiiiiiiiiiiiiiiiiii :§:§:5: Etta-1821119 g ao-l mwnnr E 70' -16:1n7 60- g 50_ Emma g 40- -16:0 (U 9:. 30- § 20- ; 10- a. 0_ Luc SCD2 Elovl-6 B 16:0(uM): o 400 L S E L S E CHOP [ -- -—1 25' ELuc -SCD2 N O - Elovl-6 * _I U! I .L O I 0| I CHOP Protein (Fold Control) 0- 16:0 (pM): 0 400 117 Figure 4.6. Continued C 16:0 (M): o 400 o 400 Marl—"9‘1 FLLLh JNK! :7r5—l [é-‘TIZZ'I 8" ELUC 8' ELuc "9‘ 7' -SCD2 7- -Elovl-6 *5 6- 6- # o * o 5' 5- E 4' 4' * o E; 3|! # 3- x - I z 2 2 31- 1- 0- 0- 16:0(uM): o 400 o 400 118 Figure 4.7. SCD2 and Elovl-6 over-expression on palmitate-induced apoptosis. INS-1 cells treated with Ad-CMV-Luciferase (Luc, L), Ad-CMV-SCD2 (SCD2, S) or Ad-CMV—Elovl-6 (Elovl-6, E) were treated for 24 hrs with increasing palmitate concentrations. A. Effect of SCD2 and Elovl-6 over-expression on caspase-9 cleavage. Cleaved caspase-9 proteins were analyzed by Western blotting. Results shown are representative of six independent experiments. B. Effect of SCD2 and Elovl-6 over-expression on DNA fragmentation as determined by ELISA. Data represent fold induction and are mean d: SEM for six independent experiments. *, p < 0.03 when compared to 0 uM Luc. #, p < 0.03 and 5, p < 0.05, when compared to Luc at the same palmitate concentration. 119 A 16:0 (uM): 0 200 300 400 L S L S L S L S Cleaved 33"“ _. Caspase-9 17kDa __’ Actin [ 16:0 (M): 0 200 300 400 L E L E L E L E Cleaved 38kDa Caspase-9 17kDa , Actin I —— 20- B ELuc : -SCD2 .g E‘ 15- -Elovl-6 * .‘E .2 c H E g 0 # mg 10" E — . a ‘2‘ L‘- 5- D 0- 16:0(uM): o 200 300 400 120 4.8. Elevated CHOP expression by SCD knockdown coincides with increased diacylglycerol formation and involves Ca2+-dependent PKC activation. Palmitate-induced lipotoxicity has been proposed to involve reduced incorporation into TAG and cholesterol ester (CE) compared with MUFAs (6, 315). To determine if modulation of palmitate toxicity in INS-1 cells involved changes in neutral lipid synthesis, cells treated for 12 hrs with 400 uM palmitate plus [1-14C]palmitic acid were analyzed for 14C-labeled palmitate incorporation into complex lipids. Compared to control cells, INS-1 cells treated with SCD siRNA had a 25% reduction in palmitate incorporation into TAG, resulting in a significant 2.4-fold increased accumulation of DAG (Figure 4.8A). In cells over-expressing SCD2 complex lipid synthesis was not significantly altered (data not shown), suggesting protection from palmitate toxicity by enhanced SCD2 expression may not involve changes in neutral lipid synthesis. Elovl-6 siRNA did not affect TAG levels but reduced DAG levels by 23%, whereas complex lipid synthesis was not affected by over-expression of Elovl-6 (data not shown). ER stress mediated by exogenous palmitate has been associated with release of ER Ca2+ into the cytoplasm (222). The combination of increased ER Ca2+ release with accumulation of DAG could cause B-cell dysfunction through sustained Ca2+-dependent protein kinase C (PKC) activation. To test this possibility, INS-1 cells treated with control or SCD siRNA were treated for 9 hrs with 400 uM palmitate and without or with the Ca2+-dependent PKC inhibitor G66976. In SCD knockdown cells, inhibition of Ca2+-dependent PKCs resulted in a significant 41% reduction of CHOP protein levels (Figure 4.88). Taken together, increased susceptibility to palmitate-induced B-cell 121 dysfunction by SCD knockdown involves increased DAG accumulation and PKC activation. 122 Figure 4.8. Effect of SCD knockdown on palmitate-induced ER stress involves diacylglycerol accumulation and activation of Ca2+-dependent PKCs. A. INS-1 cells electroporated with siRNAs for control (siCTL), SCD (siSCD), or Elovl-6 (siElovl-6) were treated with modified INS-l media containing 400 uM palmitate and [l-l4C]-palmitic acid for 12 hrs. Total lipids were extracted, fractionated by TLC and 14C-labeled palmitate incorporation into complex lipids was determined by densitometry. Data represent fold change and are mean i SEM for three independent experiments. *, p <0.01 when compared to siCTL. B. INS-1 cells electroporated with siRNAs for control (siCTL, C) or SCD (siSCD, S) were treated for 9 hrs with 400 uM palmitate and without or with 1 uM G66976. Whole cell protein extracts were fractionated by SDS-PAGE and CHOP protein was analyzed by Western blotting. Data are mean i SEM for three independent experiments. *, p < 0.02 when compared to siCTL cells. 123 > 2_5. * ESiCTL c V -siSCD % ’5 2_o. § -siElovl-6 2: Q g E 1.0- \ :2 2 * § :8“ Er 0.5! § § TAG DAG 1,2 FFA CE Vehicle 666976 16: 0 (pm: 0 400 400 cscoscs CHOPI —] ActinI-——v—-—~—-—-——-] 50] |:|siCTL |||ysco N w A c o o l l I 3(- HOP Protein (Fold Control) 8 U 0- 16:0 (uM): 0 400 0 400 Vehicle 666976 124 Discussion Chronic elevations in F F As are associated with loss of fi-cell function and the risk of developing type 2 diabetes (4, 203). Modulation of intracellular FA metabolism in [3- cells is essential for preventing the toxic effects of F F As and maintaining proper function. This study examined whether alterations in FA desaturase and elongase gene expression contribute to B-cell compensation and failure in response to lipotoxicity. Rat islets and INS-1 B-cells were found to express the desaturases SCD1, SCD2, ASD, and A6D, and the elongases Elovl-1, Elovl-2, and Elovls-4 to -7. In contrast to liver and adipose tissue, SCD2 is the predominant SCD isoform expressed in rat islets and B-cells (Fig 1) (257), and to date, the importance of SCD2 in B—cell MUFA synthesis has not been addressed. In addition, altered expression of SCDs and Elovl-6 in pre-diabetic and diabetic ZDF rat islets emphasizes that the capacity to synthesize MUFAs could significantly affect function, including the susceptibility of B-cells to lipotoxicity. ZDF rats exhibit gradually increased plasma FFAs levels prior to the onset of overt diabetes (313). Thus, alterations in B-cell FA metabolism likely affect the response of ZDF islets to exogenous FFAs. Pre-diabetic ZDF rats at 6 weeks of age maintained euglycemia but were hyperinsulinemic, which correlated with increased islet insulin gene expression. Islets from pre-diabetic ZDF rats expressed significantly higher levels of SCD1, A6D, Elovl-S, and particularly SCD2, than control rats. This is consistent with studies showing elevated expression of SCD1, A6D, and Elovl-5 in livers of ZDF rats, Zucker fatty rats, and insulin resistant ob/ob mice (316-318). Increased Elovl-5 expression may serve as a negative feedback mechanism due to the ability of PUF As to inhibit transcriptional activity of the sterol regulatory element binding protein-lo, a 125 regulator of lipogenesis (319, 320). More importantly, increased SCD gene expression in pre-diabetic ZDF rat islets may have a protective role by increasing the conversion of lipotoxic saturated FAs into MUFAs as observed in Zucker fatty rat islets (257). SCD and A6D gene expression is induced by insulin in liver (318, 321). Thus, elevated plasma insulin levels in ZDF pre-diabetic rats and Zucker fatty rats likely contribute to increased islet desaturase gene expression (Fig. 2) (257). As both control and ZDF rat islets aged, ASD and A6D gene expression decreased significantly, consistent with decreased PUFA desaturase activity found during aging in other tissues (322, 323). Hyperglycemic, diabetic ZDF rats at 13 weeks had decreased plasma insulin levels that coincided with reduced islet insulin gene expression, indicating islet failure. These islets also had reduced expression of SCD1, SCD2, and Elovl-6. Decreased SCD expression could be a result of islet failure due to reduced plasma insulin levels and a lack of islet insulin signaling. Loss of SCD expression and activity could contribute to islet failure due to increased accumulation of saturated FAS. Reduced Elovl-6 expression may have a protective role by decreasing elongation of palmitate to stearate and allowing it to be immediately desaturated to palmitoleate, a less lipotoxic FA. Altered SCD and Elovl-6 gene expression in pre-diabetic and diabetic ZDF rat islets raised the possibility that changes in MUFA synthesis may modulate B-cell function. To examine the roles of SCDs and Elovl-6 in B-cells, these genes were knocked down and over-expressed in INS-1 cells and subsequently treated with elevated levels of palmitate. SCDs primarily desaturate the saturated FAs palmitate (16:0) and stearate (18:0). Reduced SCD1 and SCD2 expression in INS-1 cells exposed to exogenous palmitate significantly decreased total MUFA synthesis and increased the 126 ratio of saturated FA to MUFAs. Over-expression of SCD2, however, selectively increased n-7 MUFAs palmitoleate (16:1,n-7) and vaccenate (18:1,n-7), not oleate (18:1,n—9). This is at odds with increased oleate in cells over-expressing SCD1 (315), and raises the possibility that SCD2 preferentially desaturates palmitate over stearate. Elovl-6 elongates both palmitate and palmitoleate. Reduced Elovl-6 expression in INS-1 cells decreased palmitate elongation and increased palmitoleate accumulation. Over- expression of Elovl-6 strongly drove elongation of palmitate but not palmitoleate, resulting in enhanced synthesis of stearate and oleate. Taken together, conversion of exogenous palmitate into specific MUFAs, n-7 or n-9, in INS-1 cells is dependent on the level of expression and activity of SCD2 and Elovl-6. ER stress results in activation of the unfolded protein response pathways IREl, PERK and ATF6 (204). IRE] induces genes important for ER expansion and reducing protein load by splicing and, in turn, activating the transcriptional activator Xbpl (206). PERK phosphorylates eukaryotic translation initiation factor 2a (eIF20t) to inhibit general protein translation while enhancing others such as ATF4 (244). Sustained PERK-ATF4 activation induces the pro-apoptotic genes ATF3 and CHOP (244, 324). Active ATF6 induces ER protein chaperones to aid in protein folding (219). In B-cells, exogenous saturated F As largely activate the IRE] and PERK pathways, increasing Xbpl splicing, eIF2a phosphorylation, ATF4 protein, and mRNA and protein levels of ATP 3 and CHOP (222, 223). Here, IN S-l cells treated with palmitate exhibited increased Xbpl splicing, ATP 3 and CHOP mRNAs, and CHOP protein. Decreased SCD gene expression significantly increased the sensitivity to palmitate induction of each of these ER stress markers, whereas decreased Elovl-6 tended to reduce Xbpl splicing and CHOP protein. 127 Over-expression of SCD2 reduced palmitate induction of CHOP protein, demonstrating for the first time that SCD2 can modulate ER stress. Liver X receptor (LXR)-activated [3- cells display increased SCD1/2 gene expression and protection from lipotoxicity (257, 325). LXR activation, however, did not alter ER stress, suggesting other mechanisms were involved (257). CHOP is also mediated through IRE] activation of the cJun/cFos pathway by phosphorylation of JNK (204). INS-1 cells exposed to elevated palmitate had increased JNK phosphorylation, and this was significantly enhanced and reduced by knockdown and over-expression of SCDs, respectively. Decreased Elovl-6 expression tended to lower JNK phosphorylation by elevated palmitate, whereas over-expression of Elovl-6 increased palmitate-induced JNK phosphorylation. Thus, altered stearate production could affect ER stress. Overall, these findings directly demonstrate that enhanced MUFA synthesis, particularly through SCD2, reduces the susceptibility to palmitate-induced ER stress. Accumulation of endogenous palmitate causes the release of ER Ca2+ stores, which could activate the intrinsic apoptosis pathway (204, 222). Increased ER stress in INS-1 cells with decreased SCD expression coincided with increased sensitivity to palmitate-induced caspase-9 cleavage and DNA fragmentation, hallmarks of apoptosis. This confirms that SCD knockdown increases susceptibility to B-cell dysfunction (256, 257). Consistent with a role of SCD to protect B-cells from palmitate-induced ER stress, SCD2 over-expression significantly reduced both markers of apoptosis. Decreased Elovl-6 expression did not significantly affect palmitate-induced apoptosis, whereas it was increased by over-expression of Elovl-6. The minimal effect of reduced Elovl-6 on ER stress and apoptosis could be due to the absence of a simultaneous increase in SCD 128 gene expression. Thus, Elovl-6 knockdown increases palmitoleate synthesis, but SCD2 over-expression drives it further and additionally increases total MUFA synthesis. Protection from lipotoxicity in cells with elevated expression of SCD] has been proposed to involve enhanced palmitate incorporation into neutral lipids (6, 315). Surprisingly, SCD2 over-expression did not enhance storage of exogenous palmitate into TAG or cholesterol ester (data not shown). This absence of an effect of SCD2 on neutral lipid synthesis could be due to enhanced glycerolipid/FA cycling or FA oxidation, thus the role of SCD2 in neutral lipid synthesis is under investigation. INS—1 cells with decreased SCD1 and SCD2 expression had lower palmitate incorporation into TAG and CE but a buildup of DAG, consistent with SCD1 involvement in TAG and CE synthesis (21, 315). Palmitoyl-CoA has been shown to activate PKCs (Corkey 2000). Accumulation of DAG and palmitoyl-CoA combined with release of ER Ca2+ could cause sustained activation of Ca2+-dependent PKCs, which may result in increased B-cell dysfunction such as ER stress. In support of this possibility, treatment with the Ca2+- dependent PKC inhibitor G66976 reduced the effect of palmitate on CHOP protein levels in INS-1 cells with decreased SCD expression. In conclusion, we demonstrate that altered SCD and Elovl-6 expression in INS-1 cells modulates MUFA synthesis and susceptibility to palmitate-induced B-cell lipotoxicity. These findings emphasize that regulation of SCDs and Elovl-6 may significantly contribute to the preservation or loss of B-cell function and the development of type 2 diabetes. 129 Chapter 5. Role of Fatty Acid Elongases in Determination of De Novo Synthesized Monounsaturated Fatty Acid Species Abstract Enhanced production of monounsaturated fatty acids (FAS) derived from carbohydrate-enriched diets has been implicated in the development of obesity and insulin resistance. The FA elongases Elovl-5 and Elovl-6 are regulated by changes in nutrient and hormone status and have been shown using intact yeast and mammalian microsome fractions to be involved in the synthesis of monounsaturated FAs. Herein, targeted knockdown and over-expression of Elovl-5 or Elovl-6 was used to determine their roles for de novo synthesis of specific species of monounsaturated FA in mammalian cells. Treatment of INS-1 cells with elevated glucose increased de novo FA synthesis and reduced the ratio of saturated to monounsaturated FAs. Elovl-5 knockdown decreased elongation of 16:1,n—7, whereas Elovl-5 over-expression increased synthesis of 18:1,n-7 but was dependent on stearoyl-CoA desaturase driven substrate availability of 16:1,n-7. Knockdown of Elovl-6 decreased elongation of both 16:0 and 16:1,n-7, resulting in accumulation of 16:1,n-7. In contrast to Elovl-5, Elovl-6 over- expression preferentially drove synthesis of 16:0 elongation products 18:0 and 18:1,n-9 but not 18:1,n-7. These findings demonstrate that coordinated induction of FA elongase and desaturase gene expression is required for balanced synthesis of specific n-7 versus 130 n-9 monounsaturated FA species. Furthermore, Elovl-6 is identified as a critical regulator in determining de novo synthesized FA end products. Introduction Diets high in carbohydrates and saturated fat are well established to cause altered fatty acid (FA) metabolism and elevated triglyceride accumulation, contributing to the development of obesity and type 2 diabetes. Elevated levels of carbohydrates specifically enriched in mono- and disaccharides induce the transcription of genes that increase glucose metabolism and lipogenesis in the liver (57, 326, 327), diverting excess catabolic metabolites into FAS for storage as triglycerides and cholesterol esters. FA elongase and desaturase enzymes catalyze the conversion of saturated F As synthesized de novo from glucose into monounsaturated FAs (MUFAs) such as palmitoleate or oleate. The accumulation of MUFAs have been associated with hypertriglyceridemia and adiposity (254, 328, 329), and inhibition of MUFA synthesis decreases triglyceride levels and protects from diet-induced obesity and insulin resistance (36, 48, 255). Interestingly, palmitoleate was recently identified as an adipose tissue-derived lipid hormone capable of enhancing muscle insulin sensitivity (330). These findings emphasize the importance of understanding the mechanisms regulating the production of MUFAs. Synthesis of de novo FAs involves the enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) which carboxylate cytosolic acetyl-CoA to malonyl-CoA and covalently bond malonyl-CoA C2 units to produce the C16 FA palmitate (16:0), respectively (8). After activation to palmitoyl-CoA, a FA elongase adds an additional malonyl-COA to make stearoyl-COA (18:0). MUFAs not derived from exogenous sources are synthesized by a delta-9 desaturase addition of a cis-double bond to 131 palmitoyl-CoA and stearoyl-CoA to form, palmitoleoyl-COA (16:1,n-7) and oleoyl-CoA (18:1,n-9), respectively. Palmitoleoyl-COA may also be elongated to vaccenyl-CoA (18:1,n-7). Stearoyl-CoA desaturases (SCDs) are delta-9 desaturases and rate limiting for MUFA synthesis (20). SCD subtypes identified in mammalian cells thus far include SCDs 1-5 (1 1-15, 18). Saturated FAS desaturated by SCDs contained chain lengths from Cl3-19 for SCDs 1, 2, and 4 and C12-16 for SCD3 (20). Synthesis of C18 MUFAs de novo from glucose requires chain elongation of C16 FAS by a FA elongase (Elovl), yet it remains uncertain which elongases are involved in synthesis of specific MUFA species (e.g. 18:1,n-7 versus 18:],n-9). Substrate specificity analyses using yeast and in vitro microsomal preparations identified the elongases Elovl-5 (FAEI, Relol, Helol) and Elovl-6 (LCE, FACE, rElo2) to be involved in MUFA synthesis (31-33, 331). Elovl-6 and SCD gene expression are induced by insulin, liver X receptors, sterol regulatory element binding protein-l (SREBP-1), and glucose induction of the carbohydrate-regulatory element binding protein/MAX-like factor X heterodimer (318, 325). In mice, hepatic Elovl-5, Elovl-6 and SCDs are induced by activation of peroxisome proliferator-activated receptor a and in leptin deficient, obese (ob/ob) mice, whereas they are suppressed by long-term feeding of diets high in saturated fat (318). Coordinated expression of elongases and desaturases by transcription factors that regulate lipogenic pathways, thus control the levels of MUFAs. In vitro assays have shown that Elovl-5 elongates unsaturated FAs, including 16:1,n-7, 18:3,n6 and 18:4,n3 (32, 33, 33]). Relative to Elovl-5, Elovl-6 more effectively elongates C12-16 saturated FAs and 16:1,n-7 (31, 32). Because oleate (18:1,n-9) is the 132 predominate MUFA in cells, Elovl-6 may play a larger role in MUFA synthesis than Elovl-5 by converting palmitate to stearate, a saturated precursor of oleate. Modulating the expression of FA elongase and desaturase genes has physiological significance, as mice with knockdowns of either Elovl-6 or SCD1 are protected from diet-induced obesity (36, 48, 255). The mechanism regulating the determination of specific de novo derived MUFA end products, however, remains to be defined. This study presents a comprehensive analysis of the effects of both decreased and increased expression of Elovl-5, Elovl-6, and SCDs on FAs synthesized de novo from glucose in a mammalian cell line. The results demonstrate that altering the expression of each enzyme causes significant changes in select MUFA species synthesized de novo. Results 5.]. Regulation of FA elongase and desaturase genes by glucose Studies characterizing the substrate specificity of FA elongases in mammalian cells have focused primarily on in vitro assays using cellular extracts or fractions. These in vitro assays indicated that Elovl-5 preferentially elongates 16:1,n-7 and polyunsaturated FAS, while Elovl-6 elongates 14:0, 16:0, and 16:1,n-7 (31-33, 331). Little is known, however, about their substrate preference in intact cells, particularly in regards to de novo derived FAS. To study the roles of Elovl-5 and Elovl-6, INS—1 cells were used to model intracellular de novo FA synthesis, as these cells are known to induce lipogenic gene expression in response to glucose (112, 234). INS-1 cells were treated with either 4 mM or 16.7 mM glucose to determine the glucose responsiveness of 133 lipogenic genes and FA elongase and desaturase genes indicated to be involved in MUF A synthesis (Figure 5.1A). Genes required for de novo synthesized palmitate (16:0), ACC and FAS, were increased 7.9- and 1.8-fold, respectively, by 16.7 mM glucose compared to 4 mM glucose (Figure 5.]B). The expression of Elovl-5 and Elovl-6 was increased 1.3-fold, whereas SCD expression was elevated 2.6-fold for SCD1 and 3.2-fold for SCD2. These results show that between the FA elongases and desaturases involved in MUFA synthesis, the expression of SCDs in INS-l cells is more responsive to glucose than Elovl-5 and Elovl- 6. Further, this suggests that modulating the expression of Elovl—5, Elovl-6, and SCDs could significantly alter the elongation and desaturation status of newly synthesized FAs. 5.2. Elevated glucose increases the abundance of de novo synthesized FAs and alters the ratio of saturated to monounsaturated FAs. Synthesis of de novo FAs in response to glucose was determined by measuring 14C-glucose incorporation into FAS in cells cultured for 48 hrs in either 4 mM or 16.7 mM glucose. INS-1 cells cultured in 16.7 mM glucose showed a 6- to 13-fold increase in 14C-labeled saturated FA and MUFA compared to cells cultured in 4 mM glucose (Figure 5.2A). The percent of labeled FAs was significantly increased for 16:1,n—7, 18:1,n-7 and 18:1,n-9, and decreased for 16:0 and 18:0 (Figure 5.28). Thus, there was a 38% decrease in the ratio of total saturated FA to MUFAs in 16.7 mM glucose treated cells (Figure 5.2C). These data demonstrate that elevated glucose increases both the abundance of de novo synthesized F As and the conversion of saturated FAs to MUFAs. INS-l cells cultured in 11.1 mM glucose were subsequently used to examine the role of Elovl—5 and Elovl-6 for synthesis of specific 18:] FA species. 134 A ACC FA Elon ase CoA (Elovl-6) Delta-9 Delta-9 Desaturase Desaturase (SCD) (SCD) 18:1 n9 Elongase 16:1 n7 ———->18:1n7 (EIovl-5/Elovl-6) B 101 * E 4 mM Glucose 2 - 16.7 mM Glucose Fold Response ACC FAS Elovl-5 Elovl-6 SCD1 SCDZ Figure 5.1. Glucose increased mRNA levels of genes involved in de novo lipogenesis and FA elongation and desaturation. A. Diagram of genes regulating end products of de novo FA synthesis. B. Levels of mRNA of de novo FA synthesis genes. Total mRNA was extracted from cells cultured for 48 hrs in 4 mM versus 16.7 mM glucose and analyzed by qRT—PCR. Data are relative to RPL32 expression and normalized to cells cultured in 4 mM glucose. compared to 4 mM glucose. 135 Data represent mean t SE. (n=3). Figure 5.2. Elevated glucose increased de novo FA abundance and monounsaturated FA synthesis. Cells were cultured for 48 hrs in 4 mM or 16.7 mM glucose. During the last 24 hrs the culture media was supplemented with [U- l4C]-glucose (specific activity of glucose was held constant). Total lipids were extracted, saponified, and 14C incorporation into F As was quantified by rp-HPLC. A. Newly synthesized FAs are represented as counts incorporated into specific FA species normalized to protein. B. Percentage of total labeled FA. The percentage of labeled 16:1,n-7, 18:0, 18:1,n-7, and 18:1,n-9 in high glucose cultured cells are significantly different when compared to cells cultured in low glucose (p < 0.05). C. Ratio of total saturated FAS to total MUFAs. Data are the mean : SE for three independent experiments. *, p < 0.03 when compared to low glucose. 136 A 50000- -14:o * -16:0 | | 40000- E218:0 E16:1n7 30000_ @18fln7 -18:1n9 20000- 10000- Counts Incorporated/pg protein 0- B 120- '0 0 . 110° - -18:1n9 g 30- .m1sz1n7 : ‘- E16z1n7 % 60- .Q 3 40- -18:0 ‘5‘, --16:0 g 20. -14:0 l _ mm:- 4 mM Glucose 16.7 mM Glucose 137 Figure 5.2. Continued C N w J:- I l I Ratio Saturated FA/MUFA 4 mM Glucose 16.7 mM Glucose 138 5.3. Selective knockdown of Elovl-5 or Elovl-6 impact synthesis of specific MUFA species. siRNAs were used to determine the relative contributions of Elovl-5, Elovl-6 or SCD1/2 on the de novo synthesis of specific FAs species in INS-1 cells cultured in elevated glucose. siRNAs selective against Elovl-5 or Elovl-6 decreased expression of the target mRNA by 75% and 81% with no significant effect on non-target mRNA levels (Figure 5.3A). SCD siRNA reduced SCD1 and SCD2 by 91% and resulted in a 1.68-fold increase in Elovl-6. Next, de novo FA synthesis was assessed in siRNA treated cells using 14C-acetic acid. Decreased expression of Elovl-5 mRNA led to increased 14C- labeling of 16:1,n-7 and decreased labeling of 16:0 (Figure 5.3B). There was no significant change in 14C-labeled 18:0 and 18:1,n-7, but there was a trend for increased 14C-labeled 18:1,n-9. These data are consistent with reduced elongation of 16:1,n-7 and increased flux to 18:1,n-9 production. Decreased expression of Elovl-6 led to decreased 14C-labeling of 18:0 and 18:1,n-9, and increased 14C-labeling of 16:0 and 16:1,n-7. There was no change in labeled 18:1,n-7. These data are consistent with Elovl—6 mediating elongation of 16:0 to 18:0. Reduction of SCD expression decreased 16:1,n-7, 18:1,n-7 and 18:1,n-9 synthesis and led to increased 14C-labeling of 16:0 and 18:0. Indexes of elongation and desaturation were calculated to determine the effects of decreased Elovl—5, Elovl-6, and SCD expression on the handling of specific FAs. In control cells, the elongation indexes for 16:0 and 16:1,n-7 are the same, whereas the desaturation index for 18:0 is approximately 3-fold greater than 16:0 (Figure 5.3C and D). Elovl-5 siRNA decreased 16:1,n-7 elongation and resulted in increased 16:0 elongation and desaturation, but caused no change in 18:0 desaturation. Elovl-6 siRNA 139 reduced 16:0 and 16:1,n-7 elongation and 18:0 desaturation. Elovl-6 siRNA had no effect on 16:0 desaturation. Reduced SCD expression decreased 16:0 elongation and caused a greater decrease in 16:1,n-7 elongation. As expected, desaturation of 16:0 and 18:0 with siSCD was also markedly decreased. These results demonstrate that decreased expression of Elovl-5 or Elovl-6 has dramatic effects on the synthesis of specific FA species derived de novo from glucose. 140 Figure 5.3. FA elongase and desaturase siRNA decreased gene expression and modulated MUFA synthesis. Cells electroporated with control (CTL), Elovl-5, Elovl-6 and SCD siRNA were cultured for 24 hrs in media containing 11.1 mM glucose followed by RNA extraction or overnight treatment with the same conditions plus [2-14C]-acetic acid. A. Levels of Elovl-5, Elovl-6, SCD1, and SCD2 mRNA, relative to RPL32 mRNA levels. Data are normalized to siCTL cells and represent the mean .4.- S.E. (n=3). *, p < 0.02 when compared to siCTL. B. Total lipids were extracted, saponified, and 14C incorporation into FAS was quantified by rp-HPLC. Data presented as percentage of total labeled FA species (n=3). C. Elongation index for each specific siRNA. Data are the mean :t SE. (n=3). *, p < 0.05 for 16:0, and #, p < 0.03 for 16:1n-7 when compared to siCTL. D. Desaturation index for each specific siRNA. Data are the mean t SE. (n=3). *, p < 0.04 for 16:0, and #, p < 0.02 for 18:0 when compared to siCTL. 141 A 2,0. EElovl-5 -E|ov|-6 -SCD1 -SCDZ Fold Response $ III/II/II/II/III/I 3(- III/I/IIIIE L siCTL siElovl-5 siElovl-6 siSCD B 120. N O I 1 U i100 ____________ -18:1n9 E 80‘ 1:.:.:.:.:.:.:.:.:.:.:- ______ IIIIII l i m 1 8 z 1 n7 '0 E16:1n7 % 60‘ Elmo E -14:0 3 3 D. O I siCTL siElovl-5 siElovl-6 siSCD 142 Figure 5.3. Continued C 2.0- I:I16:o -16:1n7 g 1.6- 'O 5 12- .5 ' * # 2’ # * 2 0.0- siCTL siElovI-5 siElovl-G siSCD U . E16:0 1 -18:0 Desaturation Index 9 .° .° .5 a) on I I 1 .° N l HI nl’i siCTL siElovl-5 siElovl-6 siSCD P O I 143 5.4. Over-expression of Elovl-5 or Elovl-6 leads to selective synthesis of specific MUFA species. To further examine the selectivity of Elovl-5, Elovl-6, and SCD2 in de novo FA synthesis, adenoviruses were constructed to over-express each individual gene and examined for their effect on de novo FA end products. Compared to a control adenovirus containing B—galactosidase (AdB—gal), over-expression of Elovl-6 resulted in a large increase in 14C-labeled 18:0 and 18:1,n-9 and decreased labeling of 16:0, 16:1,n-7, and 18:1,n-7 (Figure 5.4A). The elongation index from cells over-expressing Elovl-6 showed a large 3.6-fold increase in 16:0 elongation to 18:0 with no change in the rate of 18:0 desaturation to 18:1,n-9 (Figure 5.4B and C). The increased 16:0 elongation led to a 54% decrease in 16:0 desaturation and only a small, insignificant increase in 16:1,n-7 elongation. These results are consistent for production of 18:1,n-9 at the expense of 16:1,n-7 and 18:1,n-7. Elovl-5 over-expression had a limited effect on 14C-acetic acid incorporation into FAs compared to Elovl-6 over-expression. Elevated Elovl-5 expression led to decreased 14C-labeled 16:0 and 16:1,n-7, and increased 14C-labeled 18:0 and 18:1,n-7 (Figure 5.5A). The effect of Elovl-5 on 16:1,n-7 and 18:1,n-7 labeling corresponded with a significant increase in the elongation index of 16:1,n-7 (Figure 5.5B). The minimal effect of Elovl-5 might be due to low 16:1,n-7 substrate availability. To test this possibility, cells were treated with an adenovirus over-expressing SCD2 alone or in combination with Elovl-5 over-expression. SCD2 over-expression caused a decrease in 14C-labeled 16:0 and 18:0 and a marked increase in 14C-labeled 16:1,n-7 and 18:1,n-7. This resulted in a 5-fold increase in the 16:0 desaturation index and a 1.5-fold increase in the 18:0 144 desaturation index (Figure 5.5C). Compared to SCD2 alone, the combination of Elovl-5 and SCD2 further increased 18:1,n-7 and reduced 16:1,n-7, demonstrating that Elovl-5 elongates de novo synthesized 16:1,n-7. These results emphasize a larger role of Elovl-6 and SCD than Elovl-5 in de novo FA synthesis in this cell model and point to the elongase Elovl-6 as being critical for regulating newly synthesized FA end products. 145 Figure 5.4. Effect of increased Elovl-6 expression on de novo FA end product formation. IN S—l cells were infected with adenoviruses expressing B-galactosidase (B-gal) and Elovl—6 and were then cultured for 24 hrs in 11.1 mM glucose and [2- 14C]-acetic acid. Total lipids were extracted, saponified, and 14C incorporation into F As was quantified by rp-HPLC. A. Percentage of total labeled FA species (n=3). B. Effect of B-galactosidase and Elovl-6 on elongation index. Data are the mean :t SE (n=3). #, p < 0.01 for 16:0 when compared to B-gal. C. Effect of [3- galactosidase and Elovl-6 on desaturation index. Data are the mean :1: SE. (n=3). *, p < 0.006 for 16:0 when compared to B-gal. 146 'U 3 -18:1n9 g: m1sz1n7 3 -16:1n7 g Chem :1 -16:0 g -14:0 5 D. B-gal Elovl-6 6- |:|16:o >< 5. -16:1n7 # a: .1 2 4- C .2 3. '65 2’ 2- .0 LL] 1_ - B-gal EloiII-B 1.2- D16:0 ><1.0- -18:0 d) '0 50.3- C .9 50.6- 3 150.4- 10 0 00.2- 0.0- B-gal Elovl-6 147 Figure 5.5. Effect of increased Elovl-5 or SCD2 expression on de novo end product formation. INS-1 cells were infected with adenoviruses that express (3- galactosidase (B—gal), Elovl-5 or SCD2. Cells were cultured for 24 hrs in 11.1 mM glucose and [2-14C]-acetic acid. Total lipids were extracted, saponified, and 14C incorporation into FAs was quantified by rp-HPLC. A. Percentage of total labeled FA species (n=3). B. Effect of B-gal, Elovl-5 and SCD2 on elongation index. Data are the mean 1 SE. (n=3). #, p < 0.02 for 16:1,n-7 when compared to B-gal. C. Effect of B-gal, Elovl-5 and SCD2 on desaturation index. Data are the mean :1: SE. (n=3). *, p < 0.007 for 16:0, and #, p < 0.002 for 18:0 when compared to B-gal. 148 120- 'U '5 100- i -18:1n9 % 8 0" ':-;-;-;-;-;-;-:-;.;-;-; 1:53-32 552 E 1 8: 1 n7 I'-::16:1n7 d) jg 60‘ [:18:0 3 40- -16:0 E -14:0 2 2o- 0 D. o- Elongation Index ><1. Desaturation Inde Bgal Elovl-5/SCD2 Bgal Elovl-5 SCD2 Elovl-5ISCD2 Bgal Elovl-5 SCD2 Elovl-5/SCD2 149 Discussion The expression of FA elongases and desaturases is highly regulated by transcription factors involved in glycolytic and lipogenic gene expression (318). This coordinates changes in nutrient and hormone status with the activation of lipogenic genes and altered synthesis of specific FAs and complex lipids, which can impact the susceptibility to disease. Exposure to elevated carbohydrates activates transcription factors such as SREBP-1c and ChREBP that induce the expression of genes that enhance glucose metabolism as well as the synthesis and storage of FAS (69, 57). As shown here, elevated glucose induced the expression of ACC, FAS, and FA elongases and desaturases, which increased the synthesis of de novo derived FAs. The role FA of elongases in determining end products of de novo FA synthesis from glucose has been largely speculative. Although substrate specificities of Elovl-5 and Elovl-6 in vitro using yeast and microsomal preparations indicated elongation of 16 carbon PAS (31, 32), the effect of altered expression of these enzymes on the specific species of FA synthesized from glucose has not been addressed. This study is the first to characterize the effects of both reduced and enhanced expression of Elovl-5 and Elovl-6 on the intracellular end products of de novo derived FAs in mammalian cells. The findings reveal a significant role for FA elongase activity in regulating the synthesis of de novo derived MUFAs and establishing the balance between 16:1,n-7, 18:1,n-7, and 18:1,n-9. Elongation of FA by Elovl-5 is essential for control of hepatic lipid homeostasis as over-expression in liver decreased triglyceride content and knockdown led to activation of SREBP-1c, increased lipogenic gene expression, and hepatic steatosis (320, 332). Elovl-5 substrates include polyunsaturated FAs such as 18:4,n3, a precursor for 150 20:5,n3 FA synthesis, as well as 16:1,n-7 (32, 33, 331). Elevated levels of carbohydrate leads to increased synthesis of both 18:1,n-7 and 18:1,n-9 FA in INS-1 cells and liver (48). Targeted reduction of Elovl-5 expression decreased the elongation of 16:1,n-7 to 18:1,n-7 and increased elongation of 16:0 to 18:0 and the synthesis of 18:1,n-9, illustrating an ex vivo role for Elovl—5 in the elongation of 16:1,n-7. Findings in INS-l cells are in stark contrast to Elovl-5 null mice, which have increased rather than decreased hepatic levels of 18:1,n-7 (320). Although 18:1,n-7 levels were unexpectedly elevated in Elovl-5 null mice, PUFA levels were reduced as expected. The increased levels of hepatic 18:1,n-7 maybe associated with reduced synthesis of 22:6,n3, an inhibitor of SREBP-1 processing (8, 320). Indeed Elovl-5 null mice had increased SREBP-1c levels, de novo FA synthesis, and Elovl-6 expression, which can elongate 16:1,n—7 to 18:1,n-7 (320). Over-expression of Elovl-5 in INS-l cells decreased the amount of de novo derived 16:1,n-7 but only had a minimal effect on 18:1,n-7. The minimal effect of increased Elovl-5 expression was likely due to limited substrate availability as INS-1 cells have very low concentrations of 16:1,n-7 relative to 16:0. Consistent with this possibility, over-expression of SCD2 significantly increased 16: 1 ,n-7 synthesis, which was available for Elovl-5 to elongate to 18:1,n-7. Under many physiologic states, increased SCD expression occurs with increased expression of Elovl-5 (and Elovl-6) (318). Elovl-5 might function to keep cellular concentrations of 16:1,n-7 low, thereby preventing accumulation of 16:1,n-7 that can serve as a cell-signaling molecule (330). Although the mechanism is unknown, exposure of cells to 16:],n-7 enhances insulin signaling and its accumulation in the blood has been shown to correlate with increased muscle insulin sensitivity and protection from hepatic steatosis (3 6, 330). 151 The preferred substrate for triglyceride storage of excess FAs is the MUFA oleate (18:1,n-9) (21). Synthesis of 18:1,n-9 de novo requires the elongation of 16:0 to 18:0 prior to desaturation. Elovl-6 over—expression in INS-1 cells largely drove the elongation of 16:0 to 18:0 and promoted synthesis of 18:1,n-9 rather than elongation of 16:1,n-7 to 18:1,n-7. Conversely, reduced expression of Elovl-6 significantly decreased the products of 16:0 elongation (i.e. 18:0 and 18:1,n-9) while increasing 16:1,n-7. In addition, elongation of 16:1,n-7 to 18:1,n-7 was also decreased with siElovl-6. A role for Elovl-6 in 16:0 elongation is supported by Elovl-6 null mice, which displayed decreased hepatic accumulation of 18:0 and 18:1,n-9 and increased 16:0 and 16:1,n-7 (36). The effect of decreased elongation of both 16:0 and 16:1,n-7 by reduced Elovl-6 expression and activity is a shuttling of de novo synthesized FA towards the production of 16:1,n-7. Our data shows expression and activity of Elovl-6 is mostly involved in elongation of de novo synthesized 16:0 to produce n-9 MUFA species. Although Elovl-5 and Elovl-6 activities can influence synthesis of specific MUFA species, SCD activity clearly plays the predominate role in total MUFA synthesis. This was exemplified by the large reduction in 16:1,n-7, 18:1,n-7 and 18:1,n-9 in INS-1 cells with reduced SCD1 and SCD2. Interestingly, Elovl-6 mRNA, but not Elovl-5 mRNA, was induced in SCD deficient INS-1 cells. This finding supports the unique role of Elovl-6 in synthesis of 18:1,n-9, the predominate MUFA in cells, and suggests that MUFA provide negative-feedback control on Elovl-6 expression. Over-expression of SCD2 in IN S-l cells, in the absence of increased Elovl-5/Elovl-6, led to increased 16:1,n- 7 and 18:1,n-7, but had little impact on 18:1,n-9 synthesis. These data suggest that 152 without coordinate regulation of FA elongases, elevated SCD activity will disrupt the balance of 16:1,n-7, 18:1,n-7 and 18:1,n-9. In conclusion, this study presents a comprehensive analysis of the effects of altered expression of Elovl-5 and Elovl-6 on de novo synthesized MUFAs. Our results demonstrate that Elovl-5 preferentially converts 16:1,n-7 to 18:1,n-7, whereas Elovl-6 preferentially elongates 16:0 to 18:0, which can be further desaturated to 18:1,n-9. Loss of coordinate control of Elovl-5, Elovl-6 and SCD can disrupt production of specific MUFA species, which may negatively influence cell function. 153 Chapter 6. General Conclusions and Future Studies Chronic hyperglycemia and elevated FFA levels have been associated with the pathogenesis of (fl-cell dysfunction and T2D (4). Pancreatic [ii-cell FA metabolism is a growing area of research focused on identifying mechanisms to prevent the adverse effects of glucolipotoxicity. Recent studies demonstrated that [El-cells possess innately enhanced regulation of specific FA metabolic pathways that contribute to preserving proper function (6, 104, 234, 257). This dissertation provides novel information into how changes in FA metabolism through activation of LXRs and alterations in MUFA synthesis modulate B-cell firnction in response to glucolipotoxicity. Islet B-cells from hyperglycemic animal models of T2D exhibit diminished GSIS in conjunction with elevated lipogenic gene expression, de novo FA synthesis, and TAG accumulation (116, 233). This association between diminished GSIS and lipogenesis has been proposed to occur through activation of SREBP-1c, a major transcriptional regulator 0f lipogenic genes (116, 233). The findings presented here, however, demonstrate that enhanced activation of SREBP-1c, lipogenic gene expression, and TAG synthesis by aetivation of LXRs results in elevated basal insulin release and GSIS during chronic h)"perglycemia. These results conflict with studies showing over-expression of a c30nstitutively active SREBP-lo increases TAGS and causes loss of GSIS (116). The difference could be that B-cells are sensitive to the level of SREBP-1c activation, as its expression is required for LXR-mediated enhancement of insulin secretion (253). In addition, LXR activation may affect other FA metabolism pathways that impact B-cell 1:LlIIction. Consistent with this possibility, elevated basal insulin release from LXR- activated INS-1 cells was blocked by inhibition of acyl-CoA formation and FA oxidation, 154 Glucose Exogenous SFA (16:0) Malonyl- -CoA CPT-1 1. LC- CoA (16: o- -CoA) B-oxid. . 1FQSCDZ 11‘16'1/1821,n-7 LciCoADA DAG ATAG? F 119166 . ‘QA. v p" FA-induced ER stress Qtelycem'm'd 1 LXR -(UPR) 1TAG 1 SCDZ alone 1 MitoJER Oxidative stress -(ROS) 7?? LXR-mediated Increased TAG Synthesis, FA-Oxidation, and Elevated insulin Secretion SCDZ-mediated Reduction of Palmitate-Induced ER stress and Preservation of B-Cell Mass Figure 6.1. Mechanisms of protection from glucolipotoxicity by activation of LXRs and enhanced MUFA synthesis. LXR activation during chronic hyperglycemia drives de novo synthesis of 16:0, conversion of 16:0 to n-7 MUFAs, and TAG synthesis. In addition, LXR activation increased CPT-1 gene expression and FA oxidation. Subsequent lipoylsis of glycerolipid pools in LXR-activated B-cells increased basal insulin secretion, via the enhanced FA oxidation, and increased GSIS through generation of DAG. ER stress and loss of B—cell mass from exposure to excess exogenous palmitate was reduced by enhanced SCD2-mediated synthesis of n-7 MUFAs.‘ Whether LXR activation and increased MUFA synthesis affect oxidative stress remains to be determined. Furthermore, how SCD2 protects from palmitate toxicity is still unknown. Overall, utilization of mechanisms designed to enhance LXR and SCD2 activity could provide significant protection of B-cells from glucolipotoxicity. 155 which coincided with increased FA oxidation and expression of genes involved in mitochondrial B-oxidation. The link between LXR activation and increased CPT-1 gene expression and FA oxidation is unclear, but may involve FA-mediated regulation of AMPK and ACC or an effect of LXR on PPARor as shown in the intestine (292). Enhanced TAG synthesis and FA oxidation combined with the observation that TAGS are turned over rapidly indicated that the effect of LXR activation on GSIS involved turnover of neutral lipid pools. Inhibition of lipolysis by treatment of INS-1 cells with the general lipase inhibitor orlistat blocked the turnover of TAG and reduced GSIS. Turnover of TAG could, in turn, provide lipid signaling molecules such as DAG to enhance insulin secretion. In support of this hypothesis, elevated GSIS from LXR- activated cells was reduced by inhibition of DAG binding proteins. Thus, elevated GSIS by activation of LXRs involves TAG turnover and signaling through DAG. Our findings indicate that a balance between synthesis and turnover of neutral lipid pools is necessary for enhanced glycerolipid/FA cycling to protect B-cells from chronic hyperglycemia. This is supported by studies showing that altering only synthesis or turnover of neutral lipids causes reduced GSIS (104, 165, 239, 240). Taken together, LXR activation elevates insulin secretion through a mechanism involving increased de novo synthesis and turnover of TAG and enhanced mitochondrial B-oxidation. These findings support the emerging role of increased glycerolipid/FA cycling in B-cell compensation (99). Obese ZF rats maintain normoglycemia, in part, through enhanced islet glucose- and FA-stimulated insulin secretion (104). Interestingly, 156 ZF rat islets display increased FA esterification, lipolysis, and FA oxidation as well as increased LXRa gene expression (104, 257). Therefore, LXRs could be key regulators of B-cell glycerolipid/FA cycling. Future studies will be necessary to determine if LXR is required for glycerolipid/F A cycling. In conjunction with elevated TAG synthesis, LXR activation increased MUFA synthesis and expression of the FA desaturases SCD1 and SCD2. Characterization of FA elongase and desaturase genes in rat islets and INS-1 cells identified expression of the elongases Elovls 1, 2, 4, 5, 6 and 7, and the desaturases SCD1, SCD2, ASD and A6D. In contrast to the liver, SCD2 was the predominant SCD isoform expressed in B-cells, as shown recently (257). In addition, we demonstrated that SCD1 and SCD2 gene expression is elevated in pre-diabetic ZDF rat islets and reduced, along with Elovl-6, in diabetic ZDF rat islets. Prior to the onset of T2D, ZDF rats display gradually increasing concentrations of plasma FFAs (313), which are associated with loss of B-cell function (4). This suggested that regulation of genes involved in MUFA synthesis could be involved in B-cell compensation and failure during the development of T2D. In support of this hypothesis, we show that knockdown of SCD] and SCD2 increased the susceptibility of INS-l cells to palmitate-induced ER stress and apoptosis, confirming that SCD expression is required for protection against lipotoxicity (257). Increased palmitate toxicity in SCD knockdown cells was associated with reduced TAG, increased DAG, and Ca2+—dependent PKC activation. This correlates with studies showing that palmitate toxicity is associated with reduced incorportation into neutral lipids compared to MUFAs (241, 315). Although reduced palmitate toxicity in rat islets and MIN-6 [3- cells correlates with enhanced SCD1 and SCD2 expression (6, 257), enhanced regulation 157 of other FA metabolism pathways could account for this protection as well. For example, ZF rat islets and MIN-6 B-cells have increased expression of SCDS and CPT-1, which coincided with increased FA oxidation (6, 104, 257). Here, we show for the first time that elevated expression of only SCD2 protects from palmitate-induced ER stress and apoptosis. Protection by SCD2 over-expression, however, did not coincide with increased TAGS. Further studies are needed to determine how enhanced SCD2 expression modulates lipotoxicity. In addition, palmitate toxicity tended to be reduced by Elovl-6 knockdown, whereas it was enhanced by Elovl-6 over-expression. This correlates with Elovl-6 knockdown having beneficial effects in liver, as Elovl-6 null mice are protected from diet-induced insulin resistance (36). In the liver, increased MUFA synthesis coincides with increased expression of both SCD and Elovl-6 (318). Here, we show that elevated expression of either SCD2 or Elovl-6 alone alters the conversion of exogenous palmitate into specific MUFA species, n-7 versus n-9. The increased palmitate toxicity in cells over-expressing Elovl-6 is likely due to the significantly increased stearate production in the absence of a simultaneous increase in SCD expression. Interestingly, Elovl-6 drove the synthesis of both exogenous palmitate and de novo derived FAS towards stearate and oleate, rather and vaccenate. This demonstrates that although Elovl-6 elongates palmitate and palmitoleate in vitro (31, 32), its primary function is to elongate palmitate to provide the precursor for oleate synthesis. The FA elongase Elovl-5 is also involved in MUFA synthesis, as it elongates 16:1,n-7 to 18:1,n-7 (32, 33). Although Elovl-5 knockdown in INS-1 cells decreased elongation of de novo synthesized 16:1,n-7, its over-expression had a limited effect on 18:1,n-7 synthesis, which depended on SCD driven substrate availability of 16:1,n-7. 158 '~'_.__n3..m Taken together, our results Show that altered expression of either SCD, Elovl-6 or Elovl-5 significantly effects MUFA end products, emphasizing the importance of coordinated regulation of these genes for maintaining balanced synthesis of n-7 versus n-9 MUFAs, as found in liver (318). 159 ' 10. ll. BIBLIOGRAPHY Amos AF, McCarty DJ, Zimmet P 1997 The rising global burden of diabetes and its complications: estimates and projections to the year 2010. Diabet Med 14 Suppl 5:81-85 King H, Aubert RE, Herman WH 1998 Global burden of diabetes, 1995-2025: prevalence, numerical estimates, and projections. Diabetes Care 21:1414-1431 Prentki M, Joly E, El-Assaad W, Roduit R 2002 Malonyl-CoA Signaling, lipid partitioning, and glucolipotoxicity: role in beta-cell adaptation and failure in the etiology of diabetes. Diabetes 51 Suppl 3:8405-413 Poitout V, Robertson RP 2008 Glucolipotoxicity: fuel excess and beta—cell dysfunction. Endocrine reviews 29:351-366 Efanov AM, Sewing S, Bokvist K, Gromada J 2004 Liver X receptor activation stimulates insulin secretion via modulation of glucose and lipid metabolism in pancreatic beta-cells. Diabetes 53 Suppl 3:875-78 Busch AK, Gurisik E, Cordery DV, Sudlow M, Denyer GS, Laybutt DR, Hughes WE, Biden TJ 2005 Increased fatty acid desaturation and enhanced expression of stearoyl coenzyme A desaturase protects pancreatic beta-cells from lipoapoptosis. Diabetes 54:2917-2924 Sprecher H 2000 Metabolism of highly unsaturated n-3 and n-6 fatty acids. Biochimica et biophysica acta 1486:219-231 Jump DB 2004 Fatty acid regulation of gene transcription. Critical reviews in clinical laboratory sciences 41:41-78 Cho HP, Nakamura M, Clarke SD 1999 Cloning, expression, and fatty acid regulation of the human delta-5 desaturase. The Journal of biological chemistry 274:37335-37339 Ramanadham S, Zhang S, Ma Z, Wohltmann M, Bohrer A, Hsu FF, Turk J 2002 Delta6-, Stearoyl CoA-, and DeltaS-desaturase enzymes are expressed in beta- cells and are altered by increases in exogenous PUFA concentrations. Biochimica et biophysica acta 1580:40-56 Kaestner KH, Ntambi M, Kelly TJ, Jr., Lane MD 1989 Differentiation-induced gene expression in 3T3-L1 preadipocytes. A second differentially expressed gene encoding stearoyl-CoA desaturase. The Journal of biological chemistry 264:14755-14761 160 12. l3. 14. 15. 16. 17. 18. 19. 20. 21. 22. Ntambi JM, Buhrow SA, Kaestner KH, Christy RJ, Sibley E, Kelly TJ, Jr., Lane MD 1988 Differentiation-induced gene expression in 3T3-L1 preadipocytes. Characterization of a differentially expressed gene encoding stearoyl-CoA desaturase. The Journal of biological chemistry 263:17291-17300 Miyazaki M, Jacobson MJ, Man WC, Cohen P, Asilmaz E, Friedman JM, Ntambi JM 2003 Identification and characterization of murine SCD4, a novel heart- specific stearoyl-CoA desaturase isoform regulated by leptin and dietary factors. The Journal of biological chemistry 278:33904-33911 Zheng Y, Prouty SM, Harmon A, Sundberg JP, Stenn KS, Parimoo S 2001 Scd3-- a novel gene of the stearoyl-CoA desaturase family with restricted expression in skin. Genomics 71:182-191 Mihara K 1990 Structure and regulation of rat liver microsomal stearoyl-CoA desaturase gene. Journal of biochemistry 108:1022-1029 Wang J, Yu L, Wang H, Gao Y, Schrementi JP, Porter RK, Yurek DA, Kuo M, Suen CS, Cao G, Bean J S, Kauffinan RF, Qian Y 2008 Identification and characterization of hamster stearoyl-CoA desaturase isoforms. Lipids 43: 197-205 Zhang L, Ge L, Parimoo S, Stenn K, Prouty SM 1999 Human stearoyl-CoA desaturase: alternative transcripts generated from a single gene by usage of tandem polyadenylation sites. The Biochemical journal 340 ( Pt 1):255-264 Wang J, Yu L, Schmidt RE, Su C, Huang X, Gould K, Cao G 2005 Characterization of HSCDS, a novel human stearoyl-CoA desaturase unique to primates. Biochemical and biophysical research communications 332:735-742 Zhang S, Yang Y, Shi Y 2005 Characterization of human SCD2, an oligomeric desaturase with improved stability and enzyme activity by cross-linking in intact cells. The Biochemical journal 388:135-142 Miyazaki M, Bruggink SM, Ntambi JM 2006 Identification of mouse palmitoyl- coenzyme A Delta9-desaturase. Journal of lipid research 472700-704 Ntambi JM, Miyazaki M 2004 Regulation of stearoyl-CoA desaturases and role in metabolism. Progress in lipid research 43:91-104 Miyazaki M, Dobrzyn A, Elias PM, Ntambi JM 2005 Stearoyl-CoA desaturase-2 gene expression is required for lipid synthesis during early skin and liver development. Proceedings of the National Academy of Sciences of the United States of America 102:12501-12506 161 23. 24. 25. 26. 27. 28. 29. 30. 31. Green SC, Stewart ME, Downing DT 1984 Variation in sebum fatty acid composition among adult humans. The Journal of investigative dermatology 83:114-117 Turkish AR, Henneberry AL, Cromley D, Padamsee M, Oelkers P, Bazzi H, Christiano AM, Billheimer JT, Sturley SL 2005 Identification of two novel human acyl-CoA wax alcohol acyltransferases: members of the diacylglycerol acyltransferase 2 (DGAT2) gene superfamily. The Journal of biological chemistry 280: 14755-14764 Oh CS, Toke DA, Mandala S, Martin CE 1997 ELOZ and ELO3, homologues of the Saccharomyces cerevisiae ELO] gene, function in fatty acid elongation and are required for sphingolipid formation. The Journal of biological chemistry 272:]7376-17384 Tvrdik P, Westerberg R, Silve S, Asadi A, Jakobsson A, Cannon B, Loison G, Jacobsson A 2000 Role of a new mammalian gene family in the biosynthesis of very long chain fatty acids and sphingolipids. The Journal of cell biology 149:707-718 Agbaga MP, Brush RS, Manda] MN, Henry K, Elliott MH, Anderson RE 2008 Role of Stargardt-3 macular dystrophy protein (ELOVL4) in the biosynthesis of very long chain fatty acids. Proceedings of the National Academy of Sciences of the United States of America 105:12843-12848 Westerberg R, Mansson J E, Golozoubova V, Shabalina IG, Backlund EC, Tvrdik P, Retterstol K, Capecchi MR, Jacobsson A 2006 ELOVL3 is an important component for early onset of lipid recruitment in brown adipose tissue. The Journal of biological chemistry 281:4958-4968 Westerberg R, Tvrdik P, Unden AB, Mansson JE, Norlen L, Jakobsson A, Holleran WH, Elias PM, Asadi A, F lodby P, Toftgard R, Capecchi MR, Jacobsson A 2004 Role for ELOVL3 and fatty acid chain length in development of hair and skin firnction. The Journal of biological chemistry 279:5621-5629 Zhang K, Kniazeva M, Han M, Li W, Yu Z, Yang Z, Li Y, Metzker ML, Allikmets R, Zack DJ, Kakuk LE, Lagali PS, Wong PW, MacDonald IM, Sieving PA, Figueroa DJ, Austin CP, Gould RJ, Ayyagari R, Petrukhin K 2001 A 5-bp deletion in ELOVL4 is associated with two related forms of autosomal dominant macular dystrophy. Nature genetics 27:89-93 Moon YA, Shah NA, Mohapatra S, Warrington JA, Horton JD 2001 Identification of a mammalian long chain fatty acyl elongase regulated by sterol regulatory element-binding proteins. The Journal of biological chemistry 276:45358-45366 162 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. Inagaki K, Aki T, F ukuda Y, Kawamoto S, Shigeta S, Ono K, Suzuki 0 2002 Identification and expression of a rat fatty acid elongase involved in the biosynthesis of C18 fatty acids. Bioscience, biotechnology, and biochemistry 66:613-62] Leonard AE, Bobik EG, Dorado J, Kroeger PE, Chuang LT, Thurmond JM, Parker-Bames JM, Das T, Huang YS, Mukerji P 2000 Cloning of a human cDNA encoding a novel enzyme involved in the elongation of long-chain polyunsaturated fatty acids. The Biochemical journal 350 Pt 32765-770 Leonard AE, Kelder B, Bobik EG, Chuang LT, Lewis CJ, Kopchick JJ, Mukerji P, Huang YS 2002 Identification and expression of mammalian long-chain PUFA elongation enzymes. Lipids 37:733-740 Wang Y, Botolin D, Christian B, Busik J, Xu J, Jump DB 2005 Tissue-specific, nutritional, and developmental regulation of rat fatty acid elongases. Journal of lipid research 46:706-715 Matsuzaka T, Shimano H, Yahagi N, Kato T, Atsumi A, Yamamoto T, Inoue N, Ishikawa M, Okada S, Ishigaki N, Iwasaki H, Iwasaki Y, Karasawa T, Kumadaki S, Matsui T, Sekiya M, Ohashi K, Hasty AH, Nakagawa Y, Takahashi A, Suzuki H, Yatoh S, Sone H, Toyoshima H, Osuga J, Yamada N 2007 Crucial role of a long-chain fatty acid elongase, Elovl6, in obesity-induced insulin resistance. Nature medicine 13:] 193-1202 Carey MC, Small DM, Bliss CM 1983 Lipid digestion and absorption. Annual review of physiology 45:651-677 Mattson FH, Beck LW 1956 The specificity of pancreatic lipase for the primary hydroxyl groups of glycerides. The Journal of biological chemistry 219:735-740 Mattson FH, Volpenhein RA 1964 The Digestion and Absorption of Triglycerides. The Journal of biological chemistry 239:2772-2777 Mattson FH, Volpenhein RA 1968 Hydrolysis of primary and secondary esters of glycerol by pancreatic juice. Journal of lipid research 9:79-84 Hofmann AF, Borgstrom B 1963 Hydrolysis of long-chain monoglycerides in micellar solution by pancreatic lipase. Biochimica et biophysica acta 70:317-331 Borgstroem B 1964 Influence of Bile Salt, Ph, and Time on the Action of Pancreatic Lipase; Physiological Implications. Journal of lipid research 5:522-531 Mansbach CM, 2nd, Gorelick F 2007 Development and physiological regulation of intestinal lipid absorption. 11. Dietary lipid absorption, complex lipid synthesis, 163 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. and the intracellular packaging and secretion of chylomicrons. Am J Physiol Gastrointest Liver Physiol 2932G645-650 Wang H, Eckel RH 2009 Lipoprotein lipase: fiom gene to obesity. American journal of physiology 297:E27l-288 Wang S, Soni KG, Semache M, Casavant S, F ortier M, Pan L, Mitchell GA 2008 Lipolysis and the integrated physiology of lipid energy metabolism. Molecular genetics and metabolism 95:117-126 Aprahamian SA, Arslanian MJ, Wakil SJ 1982 Comparative studies on the kinetic parameters and product analyses of chicken and rat liver and yeast fatty acid h synthetase. Comparative biochemistry and physiology 7 1 2577-5 82 ' Wakil SJ 1989 Fatty acid synthase, a proficient multifunctional enzyme. Biochemistry 2824523-4530 Miyazaki M, Flowers MT, Sampath H, Chu K, Otzelberger C, Liu X, Ntambi JM 2007 Hepatic stearoyl-CoA desaturase-l deficiency protects mice from L carbohydrate-induced adiposity and hepatic steatosis. Cell metabolism 62484-496 ' Gonzalez-Baro MR, Lewin TM, Coleman RA 2007 Regulation of Triglyceride Metabolism. 11. Function of mitochondrial GPAT] in the regulation of triacylglycerol biosynthesis and insulin action. Am J Physiol Gastrointest Liver Physiol 292:G1195-1 199 Gimeno RE, Cao J 2008 Thematic review series: glycerolipids. Mammalian glycerol-3-phosphate acyltransferases: new genes for an old activity. Journal of lipid research 4922079-2088 Agarwal AK, Garg A 2003 Congenital generalized lipodystrophy: significance of triglyceride biosynthetic pathways. Trends in endocrinology and metabolism: TEM 142214-221 Shindou H, Hishikawa D, Harayama T, Yuki K, Shimizu T 2009 Recent progress on acyl CoA: lysophospholipid acyltransferase research. Journal of lipid research 50 Supp12S46-51 Reue K, Brindley DN 2008 Thematic Review Series: Glycerolipids. Multiple roles for lipins/phosphatidate phosphatase enzymes in lipid metabolism. Journal of lipid research 49:2493-2503 Kabashima T, Kawaguchi T, Wadzinski BE, Uyeda K 2003 Xylulose 5-phosphate mediates glucose-induced lipogenesis by xylulose 5-phosphate-activated protein phosphatase in rat liver. Proceedings of the National Academy of Sciences of the United States of America 10025107-5112 164 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. Ma L, Tsatsos NG, Towle HC 2005 Direct role of ChREBP.Mlx in regulating hepatic glucose-responsive genes. The Journal of biological chemistry 280212019- 12027 Stoeckman AK, Ma L, Towle HC 2004 Mlx is the functional heteromeric partner of the carbohydrate response element-binding protein in glucose regulation of lipogenic enzyme genes. The Journal of biological chemistry 279: 15662-15669 Yamashita H, Takenoshita M, Sakurai M, Bruick RK, Henzel WJ, Shillinglaw W, Amot D, Uyeda K 2001 A glucose-responsive transcription factor that regulates carbohydrate metabolism in the liver. Proceedings of the National Academy of Sciences of the United States of America 98:9116-9121 Ishii S, Iizuka K, Miller BC, Uyeda K 2004 Carbohydrate response element binding protein directly promotes lipogenic enzyme gene transcription. Proceedings of the National Academy of Sciences of the United States of America 101215597-15602 Guha P, Aneja KK, Shilpi RY, Haldar D 2009 Transcriptional regulation of mitochondrial glycerophosphate acyltransferase is mediated by distal promoter via ChREBP and SREBP-l. Archives of biochemistry and biophysics Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F 2004 SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 862839-848 Nohturfft A, Brown MS, Goldstein JL 1998 Topology of SREBP cleavage- activating protein, a polytopic membrane protein with a sterol-sensing domain. The Journal of biological chemistry 273:17243-17250 Yabe D, Komuro R, Liang G, Goldstein JL, Brown MS 2003 Liver-specific mRNA for Insig-2 down-regulated by insulin: implications for fatty acid synthesis. Proceedings of the National Academy of Sciences of the United States of America 10023155-3160 Yellaturu CR, Deng X, Park EA, Raghow R, Elam MB 2009 Insulin enhances biogenesis of the nuclear SREBP-1c by posttranscriptional downregulation of Insig-2a and its dissociation from SCAP-SREBP-lc complex. The Journal of biological chemistry Wang X, Sato R, Brown MS, Hua X, Goldstein J L 1994 SREBP-1, a membrane- bound transcription factor released by sterol-regulated proteolysis. Cell 77:53-62 Sakai J, Rawson RB, Espenshade P], Cheng D, Seegmiller AC, Goldstein JL, Brown MS 1998 Molecular identification of the sterol-regulated luminal protease 165 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. that cleaves SREBPS and controls lipid composition of animal cells. Molecular cell 22505-514 Brown MS, Goldstein J L 1999 A proteolytic pathway that controls the cholesterol content of membranes, cells, and blood. Proceedings of the National Academy of Sciences of the United States of America 96:11041-11048 Sheng Z, Otani H, Brown MS, Goldstein JL 1995 Independent regulation of sterol regulatory element-binding proteins 1 and 2 in hamster liver. Proceedings of the National Academy of Sciences of the United States of America 922935-938 Hegarty BD, Bobard A, Hainault I, Ferre P, Bossard P, F oufelle F 2005 Distinct roles of insulin and liver X receptor in the induction and cleavage of sterol regulatory element-binding protein- 1c. Proceedings of the National Academy of Sciences of the United States of America 102:791-796 Foretz M, Guichard C, F erre P, F oufelle F 1999 Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes. Proceedings of the National Academy of Sciences of the United States of America 96: 12737-12742 Amemiya—Kudo M, Shimano H, Hasty AH, Yahagi N, Yoshikawa T, Matsuzaka T, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Osuga J, Harada K, Gotoda T, Sato R, Kimura S, Ishibashi S, Yamada N 2002 Transcriptional activities of nuclear SREBP-la, -lc, and -2 to different target promoters of lipogenic and cholesterogenic genes. Journal of lipid research 43:]220-1235 Magana MM, Lin SS, Dooley KA, Osborne TF 1997 Sterol regulation of acetyl coenzyme A carboxylase promoter requires two interdependent binding sites for sterol regulatory element binding proteins. Journal of lipid research 38:1630—1638 Magana MM, Osborne TF 1996 Two tandem binding sites for sterol regulatory element binding proteins are required for sterol regulation of fatty-acid synthase promoter. The Journal of biological chemistry 271:32689-32694 Koo SH, Dutcher AK, Towle HC 2001 Glucose and insulin function through two distinct transcription factors to stimulate expression of lipogenic enzyme genes in liver. The Journal of biological chemistry 276:9437-9445 Kumadaki S, Matsuzaka T, Kato T, Yahagi N, Yamamoto T, Okada S, Kobayashi K, Takahashi A, Yatoh S, Suzuki H, Yamada N, Shimano H 2008 Mouse Elovl-6 promoter is an SREBP target. Biochemical and biophysical research communications 368:261-266 Bene H, Lasky D, Ntambi JM 2001 Cloning and characterization of the human stearoyl-CoA desaturase gene promoter: transcriptional activation by sterol 166 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. regulatory element binding protein and repression by polyunsaturated fatty acids and cholesterol. Biochemical and biophysical research communications 28421194- 1198 Tabor DE, Kim JB, Spiegelman BM, Edwards PA 1999 Identification of conserved cis-elements and transcription factors required for sterol-regulated transcription of stearoyl-CoA desaturase 1 and 2. The Journal of biological chemistry 274:20603-20610 Mitro N, Mak PA, Vargas L, Godio C, Hampton E, Molteni V, Kreusch A, Saez E 2007 The nuclear receptor LXR is a glucose sensor. Nature 4452219-223 Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA, Mangelsdorf DJ 1995 a LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes & development 9: 1033-1045 Costet P, Luo Y, Wang N, Tall AR 2000 Sterol-dependent transactivation of the ABC] promoter by the liver X receptor/retinoid X receptor. The Journal of a biological chemistry 275:28240-28245 Cha JY, Repa JJ 2007 The liver X receptor (LXR) and hepatic lipogenesis. The carbohydrate-response element-binding protein is a target gene of LXR. The Journal of biological chemistry 282:743-751 Yoshikawa T, Shimano H, Amemiya—Kudo M, Yahagi N, Hasty AH, Matsuzaka T, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Osuga J, Harada K, Gotoda T, Kimura S, Ishibashi S, Yamada N 2001 Identification of liver X receptor-retinoid X receptor as an activator of the sterol regulatory element-binding protein 1c gene promoter. Molecular and cellular biology 21:2991-3000 Terasaka N, Hiroshima A, Koieyama T, Ubukata N, Morikawa Y, Nakai D, Inaba T 2003 T-0901317, a synthetic liver X receptor ligand, inhibits development of atherosclerosis in LDL receptor-deficient mice. F EBS letters 53626-11 Joseph SB, Laffitte BA, Patel PH, Watson MA, Matsukuma KE, Walczak R, Collins JL, Osborne TF, Tontonoz P 2002 Direct and indirect mechanisms for regulation of fatty acid synthase gene expression by liver X receptors. The Journal of biological chemistry 27721 1019-] 1025 . Chu K, Miyazaki M, Man WC, Ntambi JM 2006 Stearoyl—coenzyme A desaturase 1 deficiency protects against hypertriglyceridemia and increases plasma high- density lipoprotein cholesterol induced by liver X receptor activation. Molecular and cellular biology 26:6786-6798 Grefhorst A, Elzinga BM, Voshol PJ, Plosch T, Kok T, Bloks VW, van der Sluijs F H, Havekes LM, Romijn JA, Verkade HJ, Kuipers F 2002 Stimulation of 167 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles. The Journal of biological chemistry 277:34182-34190 Chen G, Liang G, Ou J, Goldstein JL, Brown MS 2004 Central role for liver X receptor in insulin-mediated activation of Srebp-lc transcription and stimulation of fatty acid synthesis in liver. Proceedings of the National Academy of Sciences of the United States of America 101211245-11250 Denechaud PD, Bossard P, Lobaccaro JM, Millatt L, Staels B, Girard J, Postic C 2008 ChREBP, but not LXRS, is required for the induction of glucose-regulated genes in mouse liver. The Journal of clinical investigation 118:956-964 Kunau WH, Dommes V, Schulz H 1995 beta-oxidation of fatty acids in mitochondria, peroxisomes, and bacteria: a century of continued progress. Progress in lipid research 342267-342 Ruderman N, Prentki M 2004 AMP kinase and malonyl-CoA: targets for therapy of the metabolic syndrome. Nature reviews 3:340-351 Kahn BB, Alquier T, Carling D, Hardie DG 2005 AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell metabolism 1:15-25 McGarry JD, Brown NF 1997 The mitochondrial camitine palmitoyltransferase system. From concept to molecular analysis. European journal of biochemistry / FEBS 244:1-14 Hostetler HA, Petrescu AD, Kier AB, Schroeder F 2005 Peroxisome proliferator- activated receptor alpha interacts with high affinity and is conformationally responsive to endogenous ligands. The Journal of biological chemistry 280:18667-18682 Hostetler HA, Huang H, Kier AB, Schroeder F 2008 Glucose directly links to lipid metabolism through high affinity interaction with peroxisome proliferator- activated receptor alpha. The Journal of biological chemistry 283:2246-2254 Leone TC, Weinheimer CJ, Kelly DP 1999 A critical role for the peroxisome proliferator-activated receptor alpha (PPARalpha) in the cellular fasting response: the PPARalpha-null mouse as a model of fatty acid oxidation disorders. Proceedings of the National Academy of Sciences of the United States of America 96:7473-7478 Henquin JC, Ravier MA, Nenquin M, Jonas JC, Gilon P 2003 Hierarchy of the beta-cell signals controlling insulin secretion. European journal of clinical investigation 33 2742-750 168 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. Newgard CB, McGarry JD 1995 Metabolic coupling factors in pancreatic beta- cell signal transduction. Annual review of biochemistry 64:689-719 Straub SG, Sharp GW 2002 Glucose-stimulated signaling pathways in biphasic insulin secretion. Diabetes/metabolism research and reviews 182451-463 Jacobson DA, Philipson LH 2007 Action potentials and insulin secretion: new insights into the role of Kv channels. Diabetes, obesity & metabolism 9 Suppl 2:89-98 Nolan CJ, Madiraju MS, Delghingaro-Augusto V, Peyot ML, Prentki M 2006 Fatty acid signaling in the beta-cell and insulin secretion. Diabetes 55 Suppl 22S16-23 Roduit R, Nolan C, Alarcon C, Moore P, Barbeau A, Delghingaro-Augusto V, Przybykowski E, Morin J, Masse F, Massie B, Ruderrnan N, Rhodes C, Poitout V, Prentki M 2004 A role for the malonyl-CoA/long-chain acyl-CoA pathway of lipid signaling in the regulation of insulin secretion in response to both fuel and nonfuel stimuli. Diabetes 53:1007-1019 Newsholme P, Bender K, Kiely A, Brennan L 2007 Amino acid metabolism, insulin secretion and diabetes. Biochemical Society transactions 35:1180-1186 Drucker DJ 2006 The biology of incretin hormones. Cell metabolism 3: 153-165 Nolan CJ, Prentki M 2008 The islet beta-cell: fuel responsive and vulnerable. Trends in endocrinology and metabolism: TEM 192285-291 Nolan CJ, Leahy J L, Delghingaro-Augusto V, Moibi J, Soni K, Peyot ML, F ortier M, Guay C, Lamontagne J, Barbeau A, Przybytkowski E, Joly E, Masiello P, Wang S, Mitchell GA, Prentki M 2006 Beta cell compensation for insulin resistance in Zucker fatty rats: increased lipolysis and fatty acid signalling. Diabetologia 49:2120-2130 Schuit F, De Vos A, Farfari S, Moens K, Pipeleers D, Brun T, Prentki M 1997 Metabolic fate of glucose in purified islet cells. Glucose-regulated anaplerosis in beta cells. The Journal of biological chemistry 272:18572-18579 Farfari S, Schulz V, Corkey B, Prentki M 2000 Glucose-regulated anaplerosis and cataplerosis in pancreatic beta-cells: possible implication of a pyruvate/citrate shuttle in insulin secretion. Diabetes 492718-726 Liang Y, Matschinsky FM 1991 Content of CoA-esters in perifused rat islets stimulated by glucose and other fuels. Diabetes 40:327-333 169 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. Corkey BE, Glennon MC, Chen KS, Deeney JT, Matschinsky FM, Prentki M 1989 A role for malonyl-CoA in glucose-stimulated insulin secretion from clonal pancreatic beta-cells. The Journal of biological chemistry 264221608-21612 Macdonald MJ, Hasan NM, Longacre MJ 2008 Studies with leucine, beta- hydroxybutyrate and ATP citrate lyase-deficient beta cells support the acetoacetate pathway of insulin secretion. Biochimica et biophysica acta 1780:966-972 MacDonald MJ, Dobrzyn A, Ntambi J, Stoker SW 2008 The role of rapid lipogenesis in insulin secretion: Insulin secretagogues acutely alter lipid composition of INS-1 832/ 13 cells. Archives of biochemistry and biophysics 470:153-162 Wang H, Wollheim CB 2002 ChREBP rather than USF2 regulates glucose stimulation of endogenous L-pyruvate kinase expression in insulin-secreting cells. The Journal of biological chemistry 277:32746-32752 da Silva Xavier G, Rutter GA, Diraison F, Andreolas C, Leclerc I 2006 ChREBP binding to fatty acid synthase and L-type pyruvate kinase genes is stimulated by glucose in pancreatic beta-cells. Journal of lipid research 47:2482-2491 Brun T, Roche E, Kim KH, Prentki M 1993 Glucose regulates acetyl-CoA carboxylase gene expression in a pancreatic beta-cell line (INS-1). The Journal of biological chemistry 268: 18905-1 89] 1 Sandberg MB, Fridriksson J, Madsen L, Rishi V, Vinson C, Holmsen H, Berge RK, Mandrup S 2005 Glucose-induced lipogenesis in pancreatic beta-cells is dependent on SREBP-1. Molecular and cellular endocrinology 240294-106 Andreolas C, da Silva Xavier G, Diraison F, Zhao C, Varadi A, Lopez-Casillas F, Ferre P, F oufelle F, Rutter GA 2002 Stimulation of acetyl-CoA carboxylase gene expression by glucose requires insulin release and sterol regulatory element binding protein 1c in pancreatic MIN6 beta-cells. Diabetes 51:2536-2545 Diraison F, Parton L, Ferre P, F oufelle F, Briscoe CP, Leclerc I, Rutter GA 2004 Over-expression of sterol-regu1atory-element-binding protein-1c (SREBPlc) in rat pancreatic islets induces lipogenesis and decreases glucose-stimulated insulin release: modulation by 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR). The Biochemical journal 378:769-778 Gerin I, Dolinsky VW, Shackman JG, Kennedy RT, Chiang SH, Burant CF, Steffensen KR, Gustafsson J A, MacDougald 0A 2005 LXRbeta is required for adipocyte growth, glucose homeostasis, and beta cell function. The Journal of biological chemistry 280:23024-23031 170 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. Choe SS, Choi AH, Lee JW, Kim KH, Chung JJ, Park J, Lee KM, Park KG, Lee IK, Kim JB 2007 Chronic activation of liver X receptor induces beta-cell apoptosis through hyperactivation of lipogenesis: liver X receptor-mediated lipotoxicity in pancreatic beta-cells. Diabetes 56:1534-1543 Helleboid-Chapman A, Helleboid S, Jake] H, Timmerman C, Sergheraert C, Pattou F, F ruchart-Najib J, Fruchart JC 2006 Glucose regulates LXRalpha subcellular localization and function in rat pancreatic beta-cells. Cell research 162661-670 Rubi B, Antinozzi PA, Herrero L, Ishihara H, Asins G, Serra D, Wollheim CB, Maechler P, Hegardt F G 2002 Adenovirus-mediated overexpression of liver camitine palmitoyltransferase I in INSlE cells: effects on cell metabolism and insulin secretion. The Biochemical journal 364:2]9-226 Salt IP, Johnson G, Ashcroft SJ, Hardie DG 1998 AMP-activated protein kinase is activated by low glucose in cell lines derived from pancreatic beta cells, and may regulate insulin release. The Biochemical journal 335 ( Pt 3):533-539 Zhang S, Kim KH 1995 Glucose activation of acetyl-CoA carboxylase in association with insulin secretion in a pancreatic beta-cell line. J Endocrinol 147:33-41 Chen J, Jeppesen PB, Nordentoft I, Hermansen K 2007 Stevioside improves pancreatic beta-cell function during glucotoxicity via regulation of acetyl-CoA carboxylase. American journal of physiology 2922E1906-19l6 Roduit R, Morin J, Masse F, Segall L, Roche E, Newgard CB, Assimacopoulos- Jeannet F, Prentki M 2000 Glucose down-regulates the expression of the peroxisome proliferator-activated receptor-alpha gene in the pancreatic beta -cell. The Journal of biological chemistry 275:35799-35806 Ravnskjaer K, Boergesen M, Dalgaard LT, Mandrup S 2006 Glucose-induced repression of PPARalpha gene expression in pancreatic beta-cells involves PP2A activation and AMPK inactivation. J Mol Endocrinol 36:289-299 Khan A, Ling ZC, Landau BR 1996 Quantifying the carboxylation of pyruvate in pancreatic islets. The Journal of biological chemistry 271:2539-2542 Lu D, Mulder H, Zhao P, Burgess SC, Jensen MV, Kamzolova S, Newgard CB, Sherry AD 2002 13C NMR isotopomer analysis reveals a connection between pyruvate cycling and glucose-stimulated insulin secretion (GSIS). Proceedings of the National Academy of Sciences of the United States of America 9922708-2713 171 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. MacDonald MJ 1993 Estimates of glycolysis, pyruvate (de)carboxylation, pentose phosphate pathway, and methyl succinate metabolism in incapacitated pancreatic islets. Archives of biochemistry and biophysics 305:205-214 Guay C, Madiraju SR, Aumais A, Joly E, Prentki M 2007 A role for ATP-citrate lyase, malic enzyme, and pyruvate/citrate cycling in glucose-induced insulin secretion. The Journal of biological chemistry 282:35657-35665 Ronnebaum SM, Ilkayeva O, Burgess SC, Joseph JW, Lu D, Stevens RD, Becker TC, Sherry AD, Newgard CB, Jensen MV 2006 A pyruvate cycling pathway involving cytosolic NADP-dependent isocitrate dehydrogenase regulates glucose- stimulated insulin secretion. The Journal of biological chemistry 281230593- 30602 Joseph JW, Jensen MV, Ilkayeva O, Palmieri F, Alarcon C, Rhodes CJ, Newgard CB 2006 The mitochondrial citrate/isocitrate carrier plays a regulatory role in glucose-stimulated insulin secretion. The Journal of biological chemistry 281:35624-35632 Pongratz RL, Kibbey RG, Shulman GI, Cline GW 2007 Cytosolic and mitochondrial malic enzyme isoforms differentially control insulin secretion. The Journal of biological chemistry 282:200-207 MacDonald MJ 1995 Feasibility of a mitochondrial pyruvate malate shuttle in pancreatic islets. Further implication of cytosolic NADPH in insulin secretion. The Journal of biological chemistry 270:20051-20058 Cline GW, Lepine RL, Papas KK, Kibbey RG, Shulman GI 2004 13C NMR isotopomer analysis of anaplerotic pathways in INS-1 cells. The Journal of biological chemistry 27924437 0-44375 Stark R, Pasquel F, Turcu A, Pongratz RL, Roden M, Cline GW, Shulman GI, Kibbey RG 2009 Phosphoenolpyruvate cycling via mitochondrial pepck links anaplerosis and mitochondrial GTP with insulin secretion. The Journal of biological chemistry Ivarsson R, Quintens R, Dejonghe S, Tsukamoto K, in 't Veld P, Renstrom E, Schuit FC 2005 Redox control of exocytosis: regulatory role of NADPH, thioredoxin, and glutaredoxin. Diabetes 54: 2132- 2142 Reinbothe TM, Ivarsson R, Li DQ, Niazi O, Jing X, Zhang B, Stenson L, Brybom U, Renstrom E 2009 Glutaredoxin-l mediates NADPH-dependent stimulation of calcium-dependent insulin secretion. Molecular endocrinology (Baltimore, Md 23:893-900 172 138. 139. 140. 141. 142. 143. 144. 145. 146. Flamez D, Berger V, Kruhoffer M, Omtofi T, Pipeleers D, Schuit FC 2002 Critical role for cataplerosis via citrate in glucose-regulated insulin release. Diabetes 51:2018-2024 Brun T, Roche E, Assimacopoulos-Jeannet F, Corkey BE, Kim KH, Prentki M 1996 Evidence for an anaplerotic/malonyl-COA pathway in pancreatic beta-cell nutrient signaling. Diabetes 45: 190-198 Herrero L, Rubi B, Sebastian D, Serra D, Asins G, Maechler P, Prentki M, Hegardt F G 2005 Alteration of the malonyl-CoA/camitine palmitoyltransferase 1 interaction in the beta-cell impairs glucose-induced insulin secretion. Diabetes 54:462-471 Zhang S, Kim KH 1998 Essential role of acetyl-CoA carboxylase in the glucose- induced insulin secretion in a pancreatic beta-cell line. Cellular signalling 10235- 42 Gremlich S, Nolan C, Roduit R, Burcelin R, Peyot ML, Delghingaro-Augusto V, Desvergne B, Michalik L, Prentki M, Wahli W 2005 Pancreatic islet adaptation to fasting is dependent on peroxisome proliferator-activated receptor alpha transcriptional up-regulation of fatty acid oxidation. Endocrinology 146:3 75-3 82 Tordjman K, Standley KN, Bemal-Mizrachi C, Leone TC, Coleman T, Kelly DP, Semenkovich CF 2002 PPARalpha suppresses insulin secretion and induces UCP2 in insulinoma cells. Journal of lipid research 432936-943 Bratanova-Tochkova TK, Cheng H, Daniel S, Gunawardana S, Liu YJ, Mulvaney-Musa J, Schermerhorn T, Straub SG, Yajima H, Sharp GW 2002 Triggering and augmentation mechanisms, granule pools, and biphasic insulin secretion. Diabetes 51 Suppl 12S83-90 Itoh Y, Kawamata Y, Harada M, Kobayashi M, Fujii R, Fukusumi S, Ogi K, Hosoya M, Tanaka Y, Uejima H, Tanaka H, Maruyama M, Satoh R, Okubo S, Kizawa H, Komatsu H, Matsumura F, Noguchi Y, Shinohara T, Hinuma S, Fujisawa Y, Fujino M 2003 Free fatty acids regulate insulin secretion from pancreatic beta cells through GPR40. Nature 4222173-176 Tomita T, Masuzaki H, Iwakura H, Fujikura J, Noguchi M, Tanaka T, Ebihara K, Kawamura J, Komoto I, Kawaguchi Y, F ujimoto K, Doi R, Shimada Y, Hosoda K, Imamura M, Nakao K 2006 Expression of the gene for a membrane-bound fatty acid receptor in the pancreas and islet cell tumours in humans: evidence for GPR40 expression in pancreatic beta cells and implications for insulin secretion. Diabetologia 49:962-968 173 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. 157. Shapiro H, Shachar S, Sekler I, Hershfinkel M, Walker MD 2005 Role of GPR40 in fatty acid action on the beta cell line INS-1E. Biochemical and biophysical research communications 33 5297- 104 Latour MG, Alquier T, Oseid E, Tremblay C, Jetton TL, Luo J, Lin DC, Poitout V 2007 GPR40 is necessary but not sufficient for fatty acid stimulation of insulin secretion in vivo. Diabetes 56: 1087- 1094 Martins EF, Miyasaka CK, Newsholme P, Curi R, Carpinelli AR 2004 Changes of fatty acid composition in incubated rat pancreatic islets. Diabetes & metabolism 30221-27 Gonzalo S, Linder ME 1998 SNAP-25 palmitoylation and plasma membrane targeting require a functional secretory pathway. Molecular biology of the cell 92585-5 97 Chapman ER, Blasi J, An S, Brose N, Johnston PA, Sudhof TC, Jahn R 1996 Fatty acylation of synaptotagmin in PC12 cells and synaptosomes. Biochemical and biophysical research communications 225:326-332 Hu L, Deeney JT, Nolan CJ, Peyot ML, A0 A, Richard AM, Luc E, Faergeman NJ, Knudsen J, Guo W, Sorhede-Winzell M, Prentki M, Corkey BE 2005 Regulation of lipolytic activity by long-chain acyl-coenzyme A in islets and adipocytes. American journal of physiology 2892E1085-1092 Peter-Riesch B, Fathi M, Schlegel W, Wollheim CB 1988 Glucose and carbachol generate 1,2-diacylglycerols by different mechanisms in pancreatic islets. The Journal of clinical investigation 81:] 154-1 161 Yang C, Kazanietz MG 2003 Divergence and complexities in DAG signaling: looking beyond PKC. Trends in pharmacological sciences 242602-608 Prentki M, Matschinsky FM 1987 Ca2+, CAMP, and phospholipid-derived messengers in coupling mechanisms of insulin secretion. Physiological reviews 67:1185-1248 Yedovitzky M, Mochly-Rosen D, Johnson JA, Gray MO, Ron D, Abramovitch E, Cerasi E, Nesher R 1997 Translocation inhibitors define specificity of protein kinase C isoenzymes in pancreatic beta-cells. The Journal of biological chemistry 272:1417-1420 Pinton P, Tsuboi T, Ainscow EK, Pozzan T, Rizzuto R, Rutter GA 2002 Dynamics of glucose-induced membrane recruitment of protein kinase C beta I] in living pancreatic islet beta-cells. The Journal of biological chemistry 277:37702-37710 174 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. Biden TJ, Schmitz-Peiffer C, Burchfield JG, Gurisik E, Cantley J, Mitchell CJ, Carpenter L 2008 The diverse roles of protein kinase C in pancreatic beta-cell function. Biochemical Society transactions 362916-919 Sheu L, Pasyk EA, Ji J, Huang X, Gao X, Varoqueaux F, Brose N, Gaisano HY 2003 Regulation of insulin exocytosis by Munc13-l. The Journal of biological chemistry 278:27556-27563 Kwan EP, Xie L, Sheu L, Nolan CJ, Prentki M, Betz A, Brose N, Gaisano HY 2006 Muncl3-1 deficiency reduces insulin secretion and causes abnormal glucose tolerance. Diabetes 55:1421-1429 Rhee J S, Betz A, Pyott S, Reim K, Varoqueaux F, Augustin I, Hesse D, Sudhof P TC, Takahashi M, Rosenmund C, Brose N 2002 Beta phorbol ester- and diacylglycerol-induced augmentation of transmitter release is mediated by Muncl3s and not by PKCS. Cell 108:121-133 Noel RJ, Antinozzi PA, McGarry JD, Newgard CB 1997 Engineering of glycerol- : stimulated insulin secretion in islet beta cells. Differential metabolic fates of E glucose and glycerol provide insight into mechanisms of stimulus-secretion coupling. The Journal of biological chemistry 272: 18621-18627 Peyot ML, Nolan CJ, Soni K, Joly E, Lussier R, Corkey BE, Wang SP, Mitchell GA, Prentki M 2004 Horrnone-sensitive lipase has a role in lipid Signaling for insulin secretion but is nonessential for the incretin action of glucagon-like peptide 1 . Diabetes 5321733-1742 Mulder H, Yang S, Winzell MS, Holm C, Ahren B 2004 Inhibition of lipase activity and lipolysis in rat islets reduces insulin secretion. Diabetes 532122-128 Peyot ML, Guay C, Latour MG, Lamontagne J, Lussier R, Pineda M, Ruderman NB, Haemmerle G, Zechner R, Joly E, Madiraju SR, Poitout V, Prentki M 2009 Adipose triglyceride lipase is implicated in fuel- and non-firel-stimulated insulin secretion. The Journal of biological chemistry 284:16848-16859 Zawalich WS, Zawalich KC 1996 Regulation of insulin secretion by phospholipase C. The American journal of physiology 271:E409-416 Hughes WE, Elgundi Z, Huang P, Frohman MA, Biden TJ 2004 Phospholipase Dl regulates secretagogue-stimulated insulin release in pancreatic beta-cells. The Journal of biological chemistry 279:27534-27541 Zechner R, Strauss JG, Haemmerle G, Lass A, Zimmerrnann R 2005 Lipolysis: pathway under construction. Current opinion in lipidology 162333-340 175 169. 170. 171. 172. 173. 174. 175. 176. 177. 178. Raben DM, Baldassare JJ 2005 A new lipase in regulating lipid mobilization: hormone-sensitive lipase is not alone. Trends in endocrinology and metabolism: TEM 16:35-36 Wolf G 2005 The mechanism and regulation of fat mobilization from adipose tissue: desnutrin, a newly discovered lipolytic enzyme. Nutrition reviews 63: 166- 170 Baulande S, Lasnier F, Lucas M, Pairault J 2001 Adiponutrin, a transmembrane protein corresponding to a novel dietary- and obesity-linked mRNA specifically expressed in the adipose lineage. The Journal of biological chemistry 276233336- 33344 Lake AC, Sun Y, Li JL, Kim JE, Johnson JW, Li D, Revett T, Shih HH, Liu W, _ Paulsen J E, Gimeno RE 2005 Expression, regulation, and triglyceride hydrolase 1,. activity of Adiponutrin family members. Journal of lipid research 4622477-2487 i Soni KG, Lehner R, Metalnikov P, O'Donnell P, Semache M, Gao W, Ashman K, Pshezhetsky AV, Mitchell GA 2004 Carboxylesterase 3 (EC 3.1.1.1) is a major L adipocyte lipase. The Journal of biological chemistry 279:40683-40689 ' Mulder H, Holst LS, Svensson H, Degerrnan E, Sundler F, Ahren B, Rorsman P, Holm C 1999 Hormone-sensitive lipase, the rate-limiting enzyme in triglyceride hydrolysis, is expressed and active in beta-cells. Diabetes 482228-232 Winzell MS, Svensson H, Arner P, Ahren B, Holm C 2001 The expression of hormone-sensitive lipase in clonal beta-cells and rat islets is induced by long-term exposure to high glucose. Diabetes 50:2225-2230 Fex M, Olofsson CS, Fransson U, Bacos K, Lindvall H, Sorhede-Winzell M, Rorsman P, Holm C, Mulder H 2004 Hormone-sensitive lipase deficiency in mouse islets abolishes neutral cholesterol ester hydrolase activity but leaves lipolysis, acylglycerides, fat oxidation, and insulin secretion intact. Endocrinology 145:3746-3753 Holm C, Osterlund T, Laurel] H, Contreras JA 2000 Molecular mechanisms regulating hormone—sensitive lipase and lipolysis. Annual review of nutrition 202365-393 Fex M, Haemmerle G, Wierup N, Dekker-Nitert M, Rehn M, Ristow M, Zechner R, Sundler F, Holm C, Eliasson L, Mulder H 2009 A beta cell-specific knockout of hormone-sensitive lipase in mice results in hyperglycaemia and disruption of exocytosis. Diabetologia 522271-280 176 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. Larsson S, Wierup N, Sundler F, Eliasson L, Holm C 2008 Lack of cholesterol mobilization in islets of hormone-sensitive lipase deficient mice impairs insulin secretion. Biochemical and biophysical research communications 3762558-562 Lindvall H, Nevsten P, Strom K, Wallenberg R, Sundler F, Langin D, Winzell MS, Holm C 2004 A novel hormone-sensitive lipase isoform expressed in pancreatic beta-cells. The Journal of biological chemistry 27923828-3 836 Mathias PC, Best L, Malaisse WJ 1985 Stimulation by glucose and carbamylcholine of phospholipase C in pancreatic islets. Cell biochemistry and function 32173-177 Prentki M, Janjic D, Biden TJ, Blondel B, Wollheim CB 1984 Regulation of Ca2+ transport by isolated organelles of a rat insulinoma. Studies with endoplasmic reticulum and secretory granules. The Journal of biological chemistry 259:10118-10123 Prentki M, Biden TJ, Janjic D, Irvine RF, Berridge MJ, Wollheim CB 1984 Rapid mobilization of Ca2+ fiom rat insulinoma microsomes by inositol-1,4,5- trisphosphate. Nature 309:562-564 Biden TJ, Prentki M, Irvine RF, Berridge MJ, Wollheim CB 1984 Inositol 1,4,5- trisphosphate mobilizes intracellular Ca2+ from permeabilized insulin-secreting cells. The Biochemical journal 223:467-473 Zawalich WS, Zawalich KC, Kelley GG 1995 Regulation of insulin release by phospholipase C activation in mouse islets: differential effects of glucose and neurohumoral stimulation. Endocrinology 13624903-4909 Exton JH 2002 Phospholipase D-structure, regulation and fimction. Reviews of physiology, biochemistry and pharmacology 14421-94 Liscovitch M, Czarny M, Fiucci G, Lavie Y, Tang X 1999 Localization and possible functions of phospholipase D isozymes. Biochimica et biophysica acta 1439:245-263 Jones D, Morgan C, Cockcroft S 1999 Phospholipase D and membrane traffic. Potential roles in regulated exocytosis, membrane delivery and vesicle budding. Biochimica et biophysica acta 1439:229-244 Kooijman EE, Chupin V, dc Kruijff B, Burger KN 2003 Modulation of membrane curvature by phosphatidic acid and lysophosphatidic acid. Traffic (Copenhagen, Denmark) 4:162-174 Poitout V 2008 Phospholipid hydrolysis and insulin secretion: a step toward solving the Rubik's cube. American journal of physiology 2942E214-216 177 191. 192. 193. 194. 195. 196. 197. 198. 199. Gross RW, Ramanadham S, Kruszka KK, Han X, Turk J 1993 Rat and human pancreatic islet cells contain a calcium ion independent phospholipase A2 activity selective for hydrolysis of arachidonate which is stimulated by adenosine triphosphate and iS specifically localized to islet beta-cells. Biochemistry 32:327- 336 Ramanadham S, Bohrer A, Mueller M, Jett P, Gross RW, Turk J 1993 Mass spectrometric identification and quantitation of arachidonate-containing phospholipids in pancreatic islets: prominence of plasmenylethanolamine molecular species. Biochemistry 32:5339-5351 Wolf BA, Pasquale SM, Turk J 1991 Free fatty acid accumulation in secretagogue-stimulated pancreatic islets and effects of arachidonate on depolarization-induced insulin secretion. Biochemistry 30:63 72-6379 Jacobson DA, Weber CR, Bao S, Turk J, Philipson LH 2007 Modulation of the pancreatic islet beta-cell-delayed rectifier potassium channel Kv2.1 by the polyunsaturated fatty acid arachidonate. The Journal of biological chemistry 282:7442-7449 Ma Z, Ramanadham S, Wohltmann M, Bohrer A, Hsu FF, Turk J 2001 Studies of insulin secretory responses and of arachidonic acid incorporation into phospholipids of stably transfected insulinoma cells that overexpress group VIA phospholipase A2 (iPLA2beta ) indicate a signaling rather than a housekeeping role for iPLA2beta. The Journal of biological chemistry 276213198-13208 Bao S, Bohrer A, Ramanadham S, J in W, Zhang S, Turk J 2006 Effects of stable suppression of Group VIA phospholipase A2 expression on phospholipid content and composition, insulin secretion, and proliferation of INS-l insulinoma cells. The Journal of biological chemistry 281:187-198 Bao S, Song H, Wohltmann M, Ramanadham S, Jin W, Bohrer A, Turk J 2006 Insulin secretory responses and phospholipid composition of pancreatic islets from mice that do not express Group VIA phospholipase A2 and effects of metabolic stress on glucose homeostasis. The Journal of biological chemistry 281:20958-20973 Bao S, Jacobson DA, Wohltmann M, Bohrer A, Jin W, Philipson LH, Turk J 2008 Glucose homeostasis, insulin secretion, and islet phospholipids in mice that overexpress iPLA2beta in pancreatic beta-cells and in iPLA2beta-null mice. American journal of physiology 2942E217-229 Unger RH, Grundy S 1985 Hyperglycaemia as an inducer as well as a consequence of impaired islet cell firnction and insulin resistance: implications for the management of diabetes. Diabetologia 282119-121 178 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. 211. Shafiir E, Gutman A 1965 Patterns of Decrease of Free Fatty Acids During Glucose Tolerance Tests. Diabetes 14:77-83 Reaven GM, Hollenbeck C, Jeng CY, Wu MS, Chen YD 1988 Measurement of plasma glucose, free fatty acid, lactate, and insulin for 24 h in patients with NIDDM. Diabetes 37: 1020- 1 024 Paolisso G, Tataranni PA, Foley JE, Bogardus C, Howard BV, Ravussin E 1995 A high concentration of fasting plasma non-esterified fatty acids is a risk factor for the development of NIDDM. Diabetologia 38:1213-1217 Unger RH 1995 Lipotoxicity in the pathogenesis of obesity-dependent NIDDM. Genetic and clinical implications. Diabetes 442863-870 Eizirik DL, Cardozo AK, Cnop M 2008 The role for endoplasmic reticulum stress in diabetes mellitus. Endocrine reviews 29:42-61 Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K 2001 XBP] mRNA is induced by ATF6 and Spliced by IREl in response to ER stress to produce a highly active transcription factor. Cell 107:881-891 Calfon M, Zeng H, Urano F, Till JI-I, Hubbard SR, Harding HP, Clark SG, Ron D 2002 IRE] couples endoplasmic reticulum load to secretory capacity by processing the XBP-l mRNA. Nature 415292-96 Lee AH, Chu GC, Iwakoshi NN, Glimcher LH 2005 XBP-l is required for biogenesis of cellular secretory machinery of exocrine glands. The EMBO journal 24:4368-4380 Lee AH, Iwakoshi NN, Glimcher LB 2003 XBP-l regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Molecular and cellular biology 23:7448-7459 Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, Yoshida H, Mori K, Kaufman RJ 2002 IREl-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBPl in signaling the unfolded protein response. Genes & development 162452-466 Lee AH, Scapa EF, Cohen DE, Glimcher LH 2008 Regulation of hepatic lipogenesis by the transcription factor XBP]. Science (New York, NY 32021492- 1496 Domer AJ, Wasley LC, Kaufman RJ 1992 Overexpression of GRP78 mitigates stress induction of glucose regulated proteins and blocks secretion of selective proteins in Chinese hamster ovary cells. The EMBO journal 1121563-1571 179 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D 2000 Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nature cell biology 22326-332 Liu CY, Schroder M, Kaufman RJ 2000 Ligand-independent dimerization activates the stress response kinases IRE] and PERK in the lumen of the endoplasmic reticulum. The Journal of biological chemistry 275224881-24885 Shen J, Chen X, Hendershot L, Prywes R 2002 ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Developmental cell 3299-111 Hinnebusch AG 1993 Gene-specific translational control of the yeast GCN4 gene by phosphorylation of eukaryotic initiation factor 2. Molecular microbiology 102215-223 Harding HP, Novoa 1, Zhang Y, Zeng H, Wek R, Schapira M, Ron D 2000 Regulated translation initiation controls stress-induced gene expression in mammalian cells. Molecular cell 621099-1 108 Harding HP, Zhang Y, Zeng H, Novoa 1, Lu PD, Calfon M, Sadri N, Yun C, Popko B, Paules R, Stojdl DF, Bell JC, Hettmann T, Leiden JM, Ron D 2003 An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Molecular cell 112619-633 Yoshida H, Haze K, Yanagi H, Yura T, Mori K 1998 Identification of the cis- acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. The Journal of biological chemistry 273233741-33749 Okada T, Yoshida H, Akazawa R, Negishi M, Mori K 2002 Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. The Biochemical journal 366:585-594 Elouil H, Bensellam M, Guiot Y, Vander Mierde D, Pascal SM, Schuit FC, Jonas JC 2007 Acute nutrient regulation of the unfolded protein response and integrated stress response in cultured rat pancreatic islets. Diabetologia 50: 1442-1452 Marchetti P, Bugliani M, Lupi R, Marselli L, Masini M, Boggi U, Filipponi F, Weir GC, Eizirik DL, Cnop M 2007 The endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients. Diabetologia 5022486-2494 Cunha DA, Hekerman P, Ladriere L, Bazarra-Castro A, Ortis F, Wakeham MC, Moore F, Rasschaert J, Cardozo AK, Bellomo E, Overbergh L, Mathieu C, Lupi 180 223. 224. 225. 226. 227. 228. 229. 230. 231. 232. R, Hai T, Herchuelz A, Marchetti P, Rutter GA, Eizirik DL, Cnop M 2008 Initiation and execution of lipotoxic ER stress in pancreatic beta-cells. Journal of cell science 12122308-2318 Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A 2006 Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-l pancreatic beta-cell apoptosis. Endocrinology 14723398-3407 Laybutt DR, Preston AM, Akerfeldt MC, Kench JG, Busch AK, Biankin AV, Biden TJ 2007 Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 502752-763 Grankvist K, Marklund SL, Taljedal [B 1981 CuZn-superoxide dismutase, Mn- superoxide dismutase, catalase and glutathione peroxidase in pancreatic islets and other tissues in the mouse. The Biochemical journal 1992393-398 Tiedge M, Lortz S, Drinkgern J, Lenzen S 1997 Relation between antioxidant enzyme gene expression and antioxidative defense status of insulin-producing cells. Diabetes 46: 1733-1742 Robertson RP 2004 Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. The Journal of biological chemistry 279242351-42354 Tanaka Y, Gleason CE, Tran PO, Harmon J S, Robertson RP 1999 Prevention of glucose toxicity in HIT-T15 cells and Zucker diabetic fatty rats by antioxidants. Proceedings of the National Academy of Sciences of the United States of America 96210857-10862 Hohmeier HE, Thigpen A, Tran W, Davis R, Newgard CB 1998 Stable expression of manganese superoxide dismutase (MnSOD) in insulinoma cells prevents IL-lbeta- induced cytotoxicity and reduces nitric oxide production. The Journal of clinical investigation 10] 2181 1-1820 Oprescu AI, Bikopoulos G, Naassan A, Allister EM, Tang C, Park E, Uchino H, Lewis GF, F antus IG, RozakiS-Adcock M, Wheeler MB, Giacca A 2007 Free fatty acid-induced reduction in glucose-stimulated insulin secretion: evidence for a role of oxidative stress in vitro and in vivo. Diabetes 56:2927-2937 Moore PC, Ugas MA, Hagman DK, Parazzoli SD, Poitout V 2004 Evidence against the involvement of oxidative stress in fatty acid inhibition of insulin secretion. Diabetes 5322610-2616 Zhou YT, Shimabukuro M, Lee Y, Koyama K, Higa M, Ferguson T, Unger RH 1998 Enhanced de novo lipogenesis in the leptin-unresponsive pancreatic islets of 181 233. 234. 235. 236. 237. 238. 239. 240. 241. prediabetic Zucker diabetic fatty rats: role in the pathogenesis of lipotoxic diabetes. Diabetes 47: 1904-1908 Kakuma T, Lee Y, Higa M, Wang Z, Pan W, Shimomura I, Unger RH 2000 Leptin, troglitazone, and the expression of sterol regulatory element binding proteins in liver and pancreatic islets. Proceedings of the National Academy of Sciences of the United States of America 9728536-8541 Diraison F, Ravier MA, Richards SK, Smith RM, Shimano H, Rutter GA 2008 SREBP] is required for the induction by glucose of pancreatic beta-cell genes involved in glucose sensing. Journal of lipid research 492814-822 Takahashi A, Motomura K, Kato T, Yoshikawa T, Nakagawa Y, Yahagi N, Sone H, Suzuki H, Toyoshima H, Yamada N, Shimano H 2005 Transgenic mice overexpressing nuclear SREBP-1c in pancreatic beta-cells. Diabetes 542492-499 Kato T, Shimano H, Yamamoto T, Ishikawa M, Kumadaki S, Matsuzaka T, Nakagawa Y, Yahagi N, Nakakuki M, Hasty AH, Takeuchi Y, Kobayashi K, Takahashi A, Yatoh S, Suzuki H, Sone H, Yamada N 2008 Palmitate impairs and eicosapentaenoate restores insulin secretion through regulation of SREBP-10 in pancreatic islets. Diabetes 5722382-2392 El-Assaad W, Buteau J, Peyot ML, Nolan C, Roduit R, Hardy S, Joly E, Dbaibo G, Rosenberg L, Prentki M 2003 Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death. Endocrinology 14424154-4163 Hellemans K, Kerckhofs K, Hannaert JC, Martens G, Van Veldhoven P, Pipeleers D 2007 Peroxisome proliferator-activated receptor alpha-retinoid X receptor agonists induce beta-cell protection against palmitate toxicity. The FEBS journal 274:6094-6105 Kelpe CL, Johnson LM, Poitout V 2002 Increasing triglyceride synthesis inhibits glucose-induced insulin secretion in isolated rat islets of langerhans: a study using adenoviral expression of diacylglycerol acyltransferase. Endocrinology 14323326- 3332 Winzell MS, Svensson H, Enerback S, Ravnskjaer K, Mandrup S, Esser V, Amer P, Alves-Guerra MC, Miroux B, Sundler F, Ahren B, Holm C 2003 Pancreatic beta-cell lipotoxicity induced by overexpression of hormone-sensitive lipase. Diabetes 52:2057-2065 Cnop M, Hannaert JC, Hoorens A, Eizirik DL, Pipeleers DG 2001 Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation. Diabetes 5021771-1777 182 242. 243. 244. 245. 246. 247. 248. 249. 250. 251. 252. Haataja L, Gurlo T, Huang CJ, Butler PC 2008 Islet amyloid in type 2 diabetes, and the toxic oligomer hypothesis. Endocrine reviews 292303-316 Ma Y, Brewer JW, Diehl JA, Hendershot LM 2002 Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. Journal of molecular biology 31821351-1365 Jiang HY, Wek SA, McGrath BC, Lu D, Hai T, Harding HP, Wang X, Ron D, Cavener DR, Wek RC 2004 Activating transcription factor 3 is integral to the eukaryotic initiation factor 2 kinase stress response. Molecular and cellular biology 2421365-1377 Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, Ron D 2000 Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE]. Science (New York, NY 287:664-666 Nishitoh H, Matsuzawa A, Tobiume K, Saegusa K, Takeda K, Inoue K, Hori S, Kakizuka A, Ichijo H 2002 ASK] is essential for endoplasmic reticulum stress- induced neuronal cell death triggered by expanded polyglutamine repeats. Genes & development 16:1345-1355 Song B, Scheuner D, Ron D, Pennathur S, Kaufman RJ 2008 Chop deletion reduces oxidative stress, improves beta cell function, and promotes cell survival in multiple mouse models of diabetes. The Journal of clinical investigation 11823378-3389 - Chinnaiyan AM 1999 The apoptosome: heart and soul of the cell death machine. Neoplasia (New York, NY 125-15 Hill MM, Adrain C, Duriez PJ, Creagh EM, Martin SJ 2004 Analysis of the composition, assembly kinetics and activity of native Apaf-l apoptosomes. The EMBO journal 23:2134-2145 Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, Nagata S 1998 A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391243-50 Odinokova IV, Sung KF, Mareninova OA, Hermann K, Evtodienko Y, Andreyev A, Gukovsky I, Gukovskaya AS 2009 Mechanisms regulating cytochrome c release in pancreatic mitochondria. Gut 582431-442 Parton LE, McMillen PJ, Shen Y, Docherty E, Sharpe E, Diraison F, Briscoe CP, Rutter GA 2006 Limited role for SREBP-1c in defective glucose-induced insulin secretion from Zucker diabetic fatty rat islets: a functional and gene profiling analysis. American journal of physiology 2912E982-994 183 253. 254. 255. 256. 257. 258. 259. 260. 261. Zitzer H, Wente W, Brenner MB, Sewing S, Buschard K, Gromada J, Efanov AM 2006 Sterol regulatory element-binding protein 1 mediates liver X receptor-beta- induced increases in insulin secretion and insulin messenger ribonucleic acid levels. Endocrinology 147:3898-3905 Attie AD, Krauss RM, Gray-Keller MP, Brownlie A, Miyazaki M, Kastelein JJ, Lusis AJ, Stalenhoef AF, Stoehr JP, Hayden MR, Ntambi JM 2002 Relationship between stearoyl-CoA desaturase activity and plasma triglycerides in human and mouse hypertriglyceridemia. Journal of lipid research 4321899-1907 Ntambi JM, Miyazaki M, Stoehr JP, Lan H, Kendziorski CM, Yandell BS, Song Y, Cohen P, Friedman JM, Attie AD 2002 Loss of stearoyl-CoA desaturase-l function protects mice against adiposity. Proceedings of the National Academy of Sciences of the United States of America 99211482-11486 Flowers JB, Rabaglia ME, Schueler KL, Flowers MT, Lan H, Keller MP, Ntambi JM, Attie AD 2007 Loss of stearoyl-CoA desaturase-1 improves insulin sensitivity in lean mice but worsens diabetes in leptin-deficient obese mice. Diabetes 56:]228-1239 Hellemans KH, Hannaert JC, Denys B, Steffensen KR, Raemdonck C, Martens GA, Van Veldhoven PP, Gustafsson JA, Pipeleers D 2009 Susceptibility of pancreatic beta cells to fatty acids is regulated by LXR/PPARalpha-dependent stearoyl-coenzyme A desaturase. PloS one 42e7266 Cousin SP, Hugl SR, Wrede CE, Kajio H, Myers MG, Jr., Rhodes CJ 2001 Free fatty acid-induced inhibition of glucose and insulin-like growth factor I-induced deoxyribonucleic acid synthesis in the pancreatic beta-cell line INS-1. Endocrinology 1422229-240 Wells WW, Xu DP, Washburn MP, Cirrito HK, Olson LK 2001 Polyhydroxybenzoates inhibit ascorbic acid activation of mitochondrial glycerol- 3-phosphate dehydrogenase: implications for glucose metabolism and insulin secretion. The Journal of biological chemistry 276:2404-2410 Linning KD, Tai MH, Madhukar BV, Chang CC, Reed DN, Jr., Ferber S, Trosko JE, Olson LK 2004 Redox-mediated enrichment of self-renewing adult human pancreatic cells that possess endocrine differentiation potential. Pancreas 292e64- 76 Botolin D, Jump DB 2003 Selective proteolytic processing of rat hepatic sterol regulatory element binding protein-l (SREBP-1) and SREBP-2 during postnatal development. The Journal of biological chemistry 278:6959-6962 184 262. 263. 264. 265. 266. 267. 268. 269. 270. 271. 272. Ye DZ, Tai MH, Linning KD, Szabo C, Olson LK 2006 MafA expression and insulin promoter activity are induced by nicotinamide and related compounds in INS-1 pancreatic beta-cells. Diabetes 552742-750 Zawalich WS, Matschinsky FM 1977 Sequential analysis of the releasing and fuel function of glucose in isolated perifused pancreatic islets. Endocrinology 10021-8 Pawar A, Xu J, Jerks E, Mangelsdorf DJ, Jump DB 2002 Fatty acid regulation of liver X receptors (LXR) and peroxisome proliferator-activated receptor alpha (PPARalpha ) in HEK293 cells. The Journal of biological chemistry 277239243- 39250 Parkera SM, Moorea PC, Johnsona LM, Poitout V 2003 Palmitate potentiation of glucose-induced insulin release: a study using 2-bromopalmitate. Metabolism 5221367-1371 Foretz M, Pacot C, Dugail I, Lemarchand P, Guichard C, Le Liepvre X, Berthelier-Lubrano C, Spiegelman B, Kim JB, Ferre P, Foufelle F 1999 ADDl/SREBP-lc is required in the activation of hepatic lipogenic gene expression by glucose. Molecular and cellular biology 19:3760-3 768 Kim JB, Sarraf P, Wright M, Yao KM, Mueller E, Solanes G, Lowell BB, Spiegelman BM 1998 Nutritional and insulin regulation of fatty acid synthetase and leptin gene expression through ADDl/SREBPI. The Journal of clinical investigation 10121-9 Wang H, Maechler P, Antinozzi PA, Herrero L, Hagenfeldt-Johansson KA, Bjorklund A, Wollheim CB 2003 The transcription factor SREBP-1c is instrumental in the development of beta-cell dysfunction. The Journal of biological chemistry 278:16622-16629 Yamashita T, Eto K, Okazaki Y, Yamashita S, Yamauchi T, Sekine N, Nagai R, Noda M, Kadowaki T 2004 Role of uncoupling protein-2 up-regulation and triglyceride accumulation in impaired glucose—stimulated insulin secretion in a beta-cell lipotoxicity model overexpressing sterol regulatory element-binding protein-1c. Endocrinology 145 23566-3577 Lee Y, Hirose H, Zhou YT, Esser V, McGarry JD, Unger RH 1997 Increased lipogenic capacity of the islets of obese rats: a role in the pathogenesis of NIDDM. Diabetes 462408-413 Mohan R, Heyman RA 2003 Orphan nuclear receptor modulators. Curr Top Med Chem 321637-1647 Teboul M, Enmark E, Li Q, Wikstrom AC, Pelto-Huikko M, Gustafsson JA 1995 OR-l, a member of the nuclear receptor superfamily that interacts with the 9-cis- 185 273. 274. 275. 276. 277. 278. 279. 280. 281. retinoic acid receptor. Proceedings of the National Academy of Sciences of the United States of America 92:2096-2100 Lehmann JM, Kliewer SA, Moore LB, Smith-Oliver TA, Oliver BB, Su JL, Sundseth SS, Winegar DA, Blanchard DE, Spencer TA, Willson TM 1997 Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway. The Journal of biological chemistry 27223137-3140 Repa JJ, Turley SD, Lobaccaro JA, Medina J, Li L, Lustig K, Shan B, Heyman RA, Dietschy JM, Mangelsdorf DJ 2000 Regulation of absorption and ABC]- mediated efflux of cholesterol by RXR heterodimers. Science (New York, NY 28921524-1529 Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, Tontonoz P 2001 LXRS control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proceedings of the National Academy of Sciences of the United States of America 982507-512 Peet DJ, Turley SD, Ma W, J anowski BA, Lobaccaro JM, Hammer RE, Mangelsdorf DJ 1998 Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 93 2693-704 Repa JJ, Berge KE, Pomajzl C, Richardson JA, Hobbs H, Mangelsdorf DJ 2002 Regulation of ATP-binding cassette sterol transporters ABCGS and ABCG8 by the liver X receptors alpha and beta. The Journal of biological chemistry 277: 18793-18800 Schultz JR, Tu H, Luk A, Repa JJ, Medina J C, Li L, Schwendner S, Wang S, Thoolen M, Mangelsdorf DJ, Lustig KD, Shan B 2000 Role of LXRS in control of lipogenesis. Genes & development 14:2831-2838 Repa JJ, Liang G, On J, Bashmakov Y, Lobaccaro JM, Shimomura I, Shan B, Brown MS, Goldstein J L, Mangelsdorf DJ 2000 Regulation of mouse sterol regulatory element-binding protein-lo gene (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes & development 14:2819-2830 Stulnig TM, Steffensen KR, Gao H, Reimers M, Dahlman-Wright K, Schuster GU, Gustafsson JA 2002 Novel roles of liver X receptors exposed by gene expression profiling in liver and adipose tissue. Mol Pharrnacol 62: 1299-1305 Shalev A, Pise-Masison CA, Radonovich M, Hoffmann SC, Hirshberg B, Brady JN, Harlan DM 2002 Oligonucleotide microarray analysis of intact human pancreatic islets: identification of glucose-responsive genes and a highly regulated TGF beta signaling pathway. Endocrinology 14323695-3698 186 282. 283. 284. 285. 286. 287. 288. 289. 290. 291. 292. Olson LK, Qian J, Poitout V 1998 Glucose rapidly and reversibly decreases INS-1 cell insulin gene transcription via decrements in STF-l and C1 activator transcription factor activity. Molecular endocrinology (Baltimore, Md 122207-219 Maedler K, Oberholzer J, Bucher P, Spinas GA, Donath MY 2003 Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose on human pancreatic beta-cell turnover and function. Diabetes 52:726- 733 Kridel SJ, Axelrod F, Rozenkrantz N, Smith J W 2004 Orlistat is a novel inhibitor of fatty acid synthase with antitumor activity. Cancer Res 64:2070-2075 Roche E, F arfari S, Witters LA, Assimacopoulos-Jeannet F, Thumelin S, Brun T, Corkey BE, Saha AK, Prentki M 1998 Long-term exposure of beta-INS cells to high glucose concentrations increases anaplerosis, lipogenesis, and lipogenic gene expression. Diabetes 47: 1086-1094 Steffensen KR, Gustafsson JA 2004 Putative metabolic effects of the liver X receptor (LXR). Diabetes 53 Suppl 12836-42 Joseph JW, Odegaard ML, Ronnebaum SM, Burgess SC, Muehlbauer J, Sherry AD, Newgard CB 2007 Normal flux through ATP-citrate lyase or fatty acid synthase is not required for glucose-stimulated insulin secretion. The Journal of biological chemistry 282231592-31600 Ronnebaum SM, Joseph JW, Ilkayeva O, Burgess SC, Lu D, Becker TC, Sherry AD, Newgard CB 2008 Chronic suppression of acetyl-CoA carboxylase 1 in beta- cells impairs insulin secretion via inhibition of glucose rather than lipid metabolism. The Journal of biological chemistry 283: 14248-14256 Zhou YP, Grill VB 1994 Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. The Journal of clinical investigation 932870-876 Alstrup KK, Brock B, Hermansen K 2004 Long-Term exposure of INS-1 cells to cis and trans fatty acids influences insulin release and fatty acid oxidation differentially. Metabolism 5321 158-1165 Zhang Y, Xiao M, Niu G, Tan H 2005 Mechanisms of oleic acid deterioration in insulin secretion: Role in the pathogenesis of type 2 diabetes. Life Sci Colin S, Bourguignon E, Boullay AB, Tousaint JJ, Huet S, Caira F, Staels B, Lestavel S, Lobaccaro JM, Delerive P 2008 Intestine-specific regulation of PPARalpha gene transcription by liver X receptors. Endocrinology 14925128- 5135 187 293. 294. 295. 296. 297. 298. 299. 300. 301. 302. 303. Assimacopoulos-Jeannet F, Thumelin S, Roche E, Esser V, McGarry JD, Prentki M 1997 Fatty acids rapidly induce the camitine palmitoyltransferase I gene in the pancreatic beta-cell line IN S-l. The Journal of biological chemistry 27221659- 1664 Boni LT, Rando RR 1985 The nature of protein kinase C activation by physically defined phospholipid vesicles and diacylglycerols. The Journal of biological chemistry 260: 10819-10825 Bimer-Gruenberger R, Susani-Etzerodt H, Waldhuber M, Riesenhuber G, Schmidinger H, Rechberger G, Kollroser M, Strauss JG, Lass A, Zimmermann R, Haemmerle G, Zechner R, Hermetter A 2005 The lipolytic proteome of mouse adipose tissue. Mol Cell Proteomics 421710-1717 Duque M, Graupner M, Stutz H, Wicher I, Zechner R, Paltauf F, Hermetter A 1996 New fluorogenic triacylglycerol analogs as substrates for the determination and chiral discrimination of lipase activities. Journal of lipid research 372868-876 Prentki M, Madiraju SR 2008 Glycerolipid metabolism and signaling in health and disease. Endocrine reviews 292647-676 Soupene E, Kuypers FA 2008 Mammalian long-chain acyl-CoA synthetases. Exp Biol Med (Maywood) 233:507-521 Antinozzi PA, Segall L, Prentki M, McGarry JD, Newgard CB 1998 Molecular or pharrnacologic perturbation of the link between glucose and lipid metabolism is without effect on glucose-stimulated insulin secretion. A re-evaluation of the long-chain acyl-CoA hypothesis. The Journal of biological chemistry 273216146- 16154 Straub SG, Shanmugam G, Sharp GW 2004 Stimulation of insulin release by glucose is associated with an increase in the number of docked granules in the beta-cells of rat pancreatic islets. Diabetes 5323179-3183 Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC 2003 Beta- cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 522102-110 Clark A, Wells CA, Buley 1D, Cruickshank JK, Vanhegan RI, Matthews DR, Cooper GJ, Holman RR, Turner RC 1988 Islet amyloid, increased A-cells, reduced B-cells and exocrine fibrosis: quantitative changes in the pancreas in type 2 diabetes. Diabetes Res 92151-159 Kloppel G, Lohr M, Habich K, Oberholzer M, Heitz PU 1985 Islet pathology and the pathogenesis of type 1 and type 2 diabetes mellitus revisited. Surv Synth Pathol Res 42110-125 188 304. 305. 306. 307. 308. 309. 310. 311. 312. 313. Yoon KH, Ko SH, Cho JH, Lee JM, Ahn YB, Song KH, Yoo SJ, Kang MI, Cha BY, Lee KW, Son HY, Kang SK, Kim HS, Lee IK, Bonner-Weir S 2003 Selective beta-cell loss and alpha-cell expansion in patients with type 2 diabetes mellitus in Korea. J Clin Endocrinol Metab 88:2300-2308 Sakuraba H, Mizukami H, Yagihashi N, Wada R, Hanyu C, Yagihashi S 2002 Reduced beta-cell mass and expression of oxidative stress-related DNA damage in the islet of Japanese Type 11 diabetic patients. Diabetologia 45285-96 Kelpe CL, Moore PC, Parazzoli SD, Wicksteed B, Rhodes CJ, Poitout V 2003 Palmitate inhibition of insulin gene expression is mediated at the transcriptional level via ceramide synthesis. The Journal of biological chemistry 278230015- 30021 Carlsson C, Borg LA, Welsh N 1999 Sodium palmitate induces partial mitochondrial uncoupling and reactive oxygen species in rat pancreatic islets in vitro. Endocrinology 140:3422-3428 Maedler K, Spinas GA, Dyntar D, Moritz W, Kaiser N, Donath MY 2001 Distinct effects of saturated and monounsaturated fatty acids on beta-cell turnover and function. Diabetes 50:69-76 Lalloyer F, Vandewalle B, Percevault F, Torpier G, Kerr-Conte J, Oosterveer M, Paumelle R, F ruchart JC, Kuipers F, Pattou F, Fievet C, Staels B 2006 Peroxisome proliferator-activated receptor alpha improves pancreatic adaptation to insulin resistance in obese mice and reduces lipotoxicity in human islets. Diabetes 55:1605-1613 Sol EM, Sargsyan E, Akusjarvi G, Bergsten P 2008 Glucolipotoxicity in INS-1E cells is counteracted by camitine palmitoyltransferase ] over-expression. Biochemical and biophysical research communications 3752517-521 Borg J, Klint C, Wierup N, Strom K, Larsson S, Sundler F, Lupi R, Marchetti P, Xu G, Kimmel A, Londos C, Holm C 2009 Perilipin is present in islets of Langerhans and protects against lipotoxicity when overexpressed in the beta-cell line INS-1. Endocrinology 15023049-3057 Lai E, Bikopoulos G, Wheeler MB, RozakiS-Adcock M, Volchuk A 2008 Differential activation of ER stress and apoptosis in response to chronically elevated free fatty acids in pancreatic beta-cells. American journal of physiology 2942E540-550 Lee Y, Hirose H, Ohneda M, Johnson JH, McGarry JD, Unger RH 1994 Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent diabetes mellitus of obese rats: impairment in adipocyte-beta-cell relationships. Proceedings of the National Academy of Sciences of the United States of America 91210878-10882 189 314. 315. 316. 317. 318. 319. 320. 321. 322. 323. 324. Martinez SC, Tanabe K, Cras-Meneur C, Abumrad NA, Bemal-Mizrachi E, Permutt MA 2008 Inhibition of Foxo] protects pancreatic islet beta-cells against fatty acid and endoplasmic reticulum stress-induced apoptosis. Diabetes 572846- 859 Listenberger LL, Han X, Lewis SE, Cases S, F arese RV, Jr., Ory DS, Schaffer JE 2003 Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proceedings of the National Academy of Sciences of the United States of America 10023077-3082 Kakuma T, Lee Y, Unger RH 2002 Effects of leptin, troglitazone, and dietary fat on stearoyl CoA desaturase. Biochemical and biophysical research communications 297: 1259- l 263 Toyama T, Kudo N, Mitsumoto A, Kawashima Y 2005 Regulation of palmitoyl- CoA chain elongation by clofibric acid in the liver of Zucker fa/fa rats. Lipids 402463-470 Wang Y, Botolin D, Xu J, Christian B, Mitchell E, Jayaprakasam B, Nair 'MG, Peters JM, Busik JV, Olson LK, Jump DB 2006 Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and obesity. Journal of lipid research 4722028-2041 Botolin D, Wang Y, Christian B, Jump DB 2006 Docosahexaneoic acid (22:6,n-3) regulates rat hepatocyte SREBP-1 nuclear abundance by Erk- and 26S proteasome-dependent pathways. Journal of lipid research 472181-192 Moon YA, Hammer RE, Horton JD 2009 Deletion of ELOVLS leads to fatty liver through activation of SREBP-1c in mice. Journal of lipid research 502412-423 Waters KM, Ntambi JM 1994 Insulin and dietary fructose induce stearoyl-CoA desaturase 1 gene expression of diabetic mice. The Journal of biological chemistry 269227773-27777 Hrelia S, Bordoni A, Celadon M, Turchetto E, Biagi PL, Rossi CA 1989 Age- related changes in linoleate and alpha-linolenate desaturation by rat liver microsomes. Biochemical and biophysical research communications 1632348-355 Bordoni A, Biagi PL, Turchetto E, Hrelia S 1988 Aging influence on delta-6- desaturase activity and fatty acid composition of rat liver microsomes. Biochem Int 17:1001-1009 Pirot P, Ortis F, Cnop M, Ma Y, Hendershot LM, Eizirik DL, Cardozo AK 2007 Transcriptional regulation of the endoplasmic reticulum stress gene chop in pancreatic insulin-producing cells. Diabetes 5621069-1077 190 325. 326. 327. 328. 329. 330. 331. 332. Green CD, Jump DB, Olson LK 2009 Elevated insulin secretion from liver X receptor-activated pancreatic beta-cells involves increased de novo lipid synthesis and triacylglyceride turnover. Endocrinology 15022637-2645 F oufelle F, Ferre P 2002 New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-lo. The Biochemical journal 3662377- 391 Nagai Y, Nishio Y, Nakamura T, Maegawa H, Kikkawa R, Kashiwagi A 2002 Amelioration of high fructose-induced metabolic derangements by activation of PPARalpha. American journal of physiology 2822B] 180-1190 Okada T, Furuhashi N, Kuromori Y, Miyashita M, Iwata F, Harada K 2005 Plasma palmitoleic acid content and obesity in children. Am J Clin Nutr 822747- 750 Paillard F, Catheline D, Duff FL, Bouriel M, Deugnier Y, Pouchard M, Daubert JC, Legrand P 2008 Plasma palmitoleic acid, a product of stearoyl-coA desaturase activity, is an independent marker of triglyceridemia and abdominal adiposity. Nutr Metab Cardiovasc Dis 182436-440 Cao H, Gerhold K, Mayers JR, Wiest MM, Watkins SM, Hotamisligil GS 2008 Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell 134:933-944 Parker-Bames JM, Das T, Bobik E, Leonard AE, Thurmond JM, Chaung LT, Huang YS, Mukerji P 2000 Identification and characterization of an enzyme involved in the elongation of n-6 and n-3 polyunsaturated fatty acids. Proceedings of the National Academy of Sciences of the United States of America 9728284- 8289 Wang Y, Torres-Gonzalez M, Tripathy S, Botolin D, Christian B, Jump DB 2008 Elevated hepatic fatty acid elongase-5 activity affects multiple pathways controlling hepatic lipid and carbohydrate composition. Journal of lipid research 4921538-1552 191