. zn-ov- u v.6ulw-1-h-M -.--«;-. .4 I. -‘~~v~v ' ' ‘ ' ‘ I ’ " 4.. U .‘ u. ... ,, “ ‘MH. 7’”. L!L.t2"1Hi'iY mung..- Italy! “ii-fin State L “:‘s£‘,ersfty 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 THE ROLE OF FATTY ACID ELONGASES IN GENE EXPRESSION AND LIPID METABOLISM By YUN WANG A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Physiology 2007 ABSTRACT ROLE OF FATTY ACID ELONGASES ON HEPATIC GENE EXPRESSION AND LIPID METABOLISM By Yun Wang Fatty acid elongation and desaturation represent key metabolic steps that determine the fatty acyl composition of complex lipids in cells. Of these two metabolic pathways, factors controlling fatty acid elongase expression in mammalian tissues remain poorly defined. My studies established that of the 7 elongase subtypes expressed in mammals, adult rat liver expresses mRN A encoding 4 elongase (Elovl) subtypes; ElovlS > Elovll = Elov12 = Elovl6. Once the hepatic profile for elongase subtype expression was established, I examined the nutritional and developmental regulation of hepatic fatty acid elongase as well as the role of key transcription factors in the control of hepatic elongase and desaturase expression in rat primary hepatocytes and in liver in vivo. These studies established that hepatic elongase expression is controlled by several transcription factors; including PPARa, SREBP-l, LXR, ChREBP and MLX. The in vivo studies show that changes in hepatic lipid composition induced by diabetes or obesity correlated with changes in elongase and desaturase expression. Even though individual elongase subtypes have different substrate preferences, tissue specific expression, and are regulated differently, the significance of this diversity is unknown. In an effort to better understand the impact of altered elongase expression on hepatic physiology, I over expressed Elov12 and ElovlS in primary hepatocytes using recombinant adenovirus technology. Overexpression of both Elov12 (Ad-Elov12) and ElovlS (Ad-ElovlS) enhanced 20:5,n-3 conversion to 22:5,n-3, but only Elov12 stimulated the formation of 24:5,n-3. The enhanced rate of 20:5 ,n—3 to elongated fatty acids attenuated 20:5-mediated induction on PPARa regulated transcripts. Over expressed Elov12, but not over expressed Elov15, also enhanced 20:5-mediated suppression of SREBP-l nuclear content. To gain a better understanding of the function of these enzymes in vivo, I used the recombinant adenovirus to over express ElovlS in livers of C57BL/6 male mice. When compared to a control virus, overexpression of Ad-ElovlS suppressed hepatic glycogen content and lowered blood glucose. I attributed this response to suppressed expression of several genes involved in glucose metabolism, i.e., Glut2, L-PK, PepCK. As in primary hepatocytes, overexpressed ElovlS also suppressed several PPARa target genes. Over expressed ElovlS also promoted changes in both the hepatic and plasma lipid profile consistent with ElovlS overexpression. Over expressed Elovl5 also significantly suppressed blood triglyceride during refeeding. Taken together, these studies indicate that changes in hepatic Elov15 activity, alone, are sufficient to impact both hepatic and plasma glucose and lipid composition. In conclusion, these studies establish a role for specific transcriptional regulatory networks in the control of hepatic elongase gene expression and hepatic lipid composition. Based on my overexpression studies, fatty acid elongases play an important role in hepatic and whole body glucose and lipid metabolism. iv To my parents and' my fius5anJ ACKNOWLEDGEMENTS This thesis is the result of four and half years of work in Michigan State University where I have been accompanied and supported by many people. It is pleasant that, now, I have the opportunity to express my gratitude to all of them. First a very special thank to my major advisor Dr. Donald B. Jump. His enthusiasm and integral view on science has made a deep impression on me. I owe him lots of gratitude for having shown me this way of research. He could not even realize how much I have learned from him. Besides being an excellent supervisor, Don was as close as a family member and a good fiiend to me. It is my pleasure to get to know him in my life. I would also like to gratefully acknowledge my committee members, Dr. Dale Romsos, Dr. Kathy Gallo, Dr. Laura McCabe and Dr. L. Karl Olson, for their monitoring of my work and took effort in providing me with valuable suggestions and ideas on some experiments of this thesis. I extend my thanks to Dr. Olson, Dr. Jeannine Scott and Dr. Andrea Amalfitano who provide helpful guidance about the construction and application of adenovirus and share their equipment with me. I warmly thank Dr. Busik for her valuable advice and friendly help. Her extensive discussions around my work have been very helpful for my study. I owe my thanks to my lab mates, Dr. J inghua Xu, Dr. Daniela Botolin, Barbara Christian and Dr. Olivier Demeure, for their kind help in daily research work and warm friendship in life. During this thesis work I have collaborated with many colleagues for whom I have great regard, I thank Dr. Kate Claycomb and Dale Romsos for their generous supply of liver from lean and obese mice; thank Dr. Howard C. Towle at the University of Minnesota for generously providing recombinant adenovirus expressing SREBP-lc nuclear form and the dominant negative MLX. For the liver histology reported in this thesis, I would like to acknowledge technical support by: Amy S. Porter (HT) ASCP QIHC, Kathleen A. Campbell (HT) ASCP QIHC, Ricky A. Rosebury (HT) ASCP QIHC of the Investigative HistoPathology Laboratory, Department of Physiology. I wish to extend my warmest thanks to all those who have helped me with my work in the Department of Physiology. I thank Dr. Heidemann and Dr. Weber for sharing their microscopes; thank the help from Dr. Kreulen’s lab for the use of real time PCR equipment; thank Chris Green, Katrina Linning, Carol Hall, Dr. Xiaopeng Li for their kind help in use of the equipment in their labs. I also appreciate the precious friendship from Dr. Diana Ye, Dr. Weiqin Chen, Dr. Xiaoling Dai, Dr. Wei Ni, Hui-yuan Tang, Wei Li, and Xiaohong Wang and his husband Hui Wang. I thank them for their kind help in my life in Michigan. I am very grateful for my dear husband Fengyuan, for his love and patience during my PhD study. One of the best experiences during this period was the birth of our lovely daughter Anji, who provided a joyful time to our life. Finally, I have a deep sense of gratitude to my dear parents for their 30 years’ support without any reservation and persistent inspiration in my journey to PhD. vi TABLE OF CONTENTS LIST OF FIGURES x LIST OF TABLES xi LIST OF ABBREVIATIONS xii INTRODUCTION .................................................................................... 1 Chapter 1. Literature Review ........................................................................ 3 1.1 Significance of fatty acid metabolism .......................................................... 3 1.1.1 Fatty acid metabolism ..................................................................... 3 1.1.1.1 General features of fatty acid ................................................... 3 1.1.1.2 Dietary fatty acid metabolism ................................................... 4 1.1.1.3 De novo synthesis of fatty acids ................................................ 6 1.1.1.4 Modification of dietary fatty and endogenous fatty acids: elongation and desaturation .................................................................................. 8 1.1.2 Fatty acid regulation of gene transcription ............................................ 15 1.1.2.1 PPARa .................................................................................................... 16 1.1.2.2 SREBP-lc ........................................................................ 18 1.1.2.3. ChREBP/MLX .................................................................. 19 1.1.2.4 Other transcriptional factor regulated by fatty acids ........................ 20 1.1.3. Fatty acid and chronic disease ......................................................... 22 1.1.3.1 .Cardiovascular disease ......................................................... 22 1.1.3 .2.Diabetes ........................................................................... 23 1.1.3.3.Inflammation ..................................................................... 24 1.1.3.4.Cancer ............................................................................ 25 1.2 Fatty acid elongases ............................................................................ 27 1.2.1 Elovll ...................................................................................... 31 1.2.2 Elov12 ...................................................................................... 32 1.2.3 Elovl3 ...................................................................................... 32 1.2.4 Elovl4 ...................................................................................... 34 1.2.5 ElovlS ...................................................................................... 35 1.2.6 Elovl6 ...................................................................................... 35 1.2.7 Elovl7 .................................................................................... 36 1.3 Statement of the problem and Hypothesis ................................................ 36 Chapter2. Tissue-Specific, nutritional and Development Regulation of Rat Fatty Acid Elongases ............................................................................................. 39 2.1 Introduction ...................................................................................... 39 2.2 Methods and Materials ......................................................................... 41 2.3 Results ............................................................................................ 47 2.3.1. Tissue-specific expression of rat fatty acid elongases .............................. 47 vii 2.3.2. Dietary Effects on Hepatic Elongase and Desaturase Gene Expression ......... 47 2.3.3. Development regulation of Hepatic fatty acid elongase and desaturase gene expression ........................................................................................... 56 2.4. Disccussion ..................................................................................... 60 Chapter 3. Regulation of Hepatic Fatty Acid Elongase and Desaturase Expression in Diabetes and Obesity ............................................................................... 68 3.1 Introduction ...................................................................................... 68 3.2 Methods and Materials ........................................... ' .............................. 69 3.3 Results ............................................................................................ 76 3.3.1. Elongase and desaturase expression in rat and mouse liver ....................... 76 3.3.2. Role of PPARa in the control of hepatic elongase and desaturase expression..76 3.3.3. Regulation of elongase and desaturase expression in primary rat hepatocytes.79 3.3.4. Metabolism of fatty acids by fatty acid elongases .................................. 86 3.3.5. Regulation of elongase and desaturase expression in animal models of metabolic disease ................................................................................ 90 3.4. Disccussion ..................................................................................... 103 Chapter 4. Effect of Fatty Acid Elongases on Gene Expression and Lipid Metabolism in Rat Primary Hepatocytes ......................................................................... 112 4.1 Introduction ..................................................................................... 1 12 4.2 Methods and Materials ........................................................................ 113 4.3 Results .......................................................................................... 1 18 4.3.1. Efficacy of Ad-Elov12 and Ad-ElovlS infection in rat primary hepatocytes .118 4.3.2. Effect of Elov12 and Elov15 on fatty acid metabolism in rat primary hepatocytes ...................................................................................... 123 4.3.3. Effect of Elov12 and Elov15 on gene expression in rat primary hepatocytes 123 4.3.4. Effect of Elov12 and Elov15 on SREBPs nuclear content in rat primary hepatocytes .................................................................................... 125 4.4. Disccussion ..................................................................................... 129 Chapter 5. Effect of ElovlS on Hepatic Function and Lipid metabolism in Vivo .................................................................................................. 133 5.1 Introduction ..................................................................................... 133 5.2 Methods and Materials ........................................................................ 134 5.3 Results .......................................................................................... 139 53.1 .Generation of the mice overexpressing ElovlS in liver ............................ 139 5.3.2. Metabolic characterization of the mice overexpressing ElovlS in liver. . . 141 5.3.3. Alteration of glucose metabolism in Ad-ElovlS injected mice .................. 143 5.3.4. Decreased expression if genes involved in lipid metabolism in Ad-Elov15 injected mice ................................................................................... 148 5.3.5. Effect of ElovlS overexpression in hepatic and plasma fatty acid profile. . 1 51 5 .4. Disccussion ..................................................................................... 15 1 Chapter 6. Conclusions and Future Directions ................................................ 160 APPENDIX ......................................................................................... 165 viii PUBLICTIONS .................................................................................... 165 BIBLIOGRAPHY ................................................................................. 167 ix LIST OF FIGURES Figure 1.1 .............................................................................. 5 Figure 1.2 .............................................................................. 7 Figure 1.3 .............................................................................. 10 Figure 1.4 ............................................................................ 11 Figure 2.1 ............................................................................ 48 Figure 2.2 ............................................................................ 49 Figure 2.3 ............................................................................ 53 Figure 2.4 ............................................................................ 55 Figure 2.5 ............................................................................ 57 Figure 2.6 ............................................................................ 59 Figure 2.7 ............................................................................ 61 Figure 3.1 ............................................................................ 77 Figure 3.2 ............................................................................ 80 Figure 3.3 ............................................................................ 82 Figure 3.4 ............................................................................ 84 Figure 3.5 ............................................................................ 87 Figure 3.6 ............................................................................ 89 Figure 3.7 ............................................................................ 91 Figure 3.8 ............................................................................ 94 Figure 3.9 ............................................................................ 96 Figure 3.10 .............................................................................. 100 Figure 3.11 .......................................................................... 104 Figure 4.1 ........................................................................... 119 Figure 4.2 ........................................................................... 124 Figure 4.3 ........................................................................... 127 Figure 4.4 ........................................................................... 128 Figure 4.5 ........................................................................... 130 Figure 5.1 ........................................................................... 140 Figure 5.2 ........................................................................... 142 Figure 5.3 ........................................................................... 144 Figure 5.4 ........................................................................... 147 Figure 5.5 .......................................................................... 149 Figure 5.6 .......................................................................... 152 Figure 5.7 .......................................................................... 154 Figure 5.8 .......................................................................... 155 Figure 5.9 .......................................................................... 156 LIST OF TABLES Table 1.1 ............................................................................................. 29 Table 1.2 ............................................................................................. 30 Table 2.1 ............................................................................................. 46 Table 3.1 ............................................................................................. 73 Table 3.2 ............................................................................................ 107 Table 4.1 ............................................................................................ 117 Table 5.1 ............................................................................................ 138 Images in this dissertation are presented in color xi LIST OF ABBREVIATIONS AA: arachidonic acid ACC: acetyl CoA Carboxylase ACS: acyl-CoA synthetase ACAT: acyl-CoA cholesterol acyltransferase Ad: Adenovirus ALA: Alpha-linolenic acid AOX: Acyl CoA oxidase bHLH/ZIP: basic helix-loop—helix/leucine zipper ChoRE: carbohydrate regulatory elements ChREBP: Carbohydrate regulatory element-binding protein Cig 30: Cold induced glycoprotein with molecular weight about 30kDa CNS: central nerve system CPT: camitine palmitoyltransferase CVD: cardiovascular disease CYP4A: cytochrome P450 DGAT: diacylglycerol acyltransferase DHA: docosahexaenoic acid ASD: fatty acid desaturase 5 A6D: fatty acid desaturase 6 A9D: fatty acid desaturase 9 EFAD: essential fatty acid deficiency xii Elovl-l: fatty acid elongase family member 1 Elovl-2: fatty acid elongase family member 2 Elovl-3: fatty acid elongase family member 3 Elov1-4: fatty acid elongase family member 4 Elovl-S: fatty acid elongase family member 5 Elov1-6: fatty acid elongase family member 6 Elovl-7: fatty acid elongase family member 7 EPA: eicosapentaenoic acid ER: endoplasmic reticulum FAS: fatty acid synthase GK: glucokinase GLUTZ: glucose transporter 2 HNF-4 a: hepatic nuclear factor-4a H&E: hepatoxylin and eosin HMGCSZ: 3-hydroxy-3-methylglutaryl-Coenzyme A synthase 2 LA: linoleic acid LBD: ligand binding domain LPK: L-pyruvate kinase LTB4; leukotriene B4 LUC: luciferase LXR: liver X receptor MLX: MAX-like factor MTE-I: mitochondial thioesterase I xiii MUFA: monounsaturated fatty acid NEFA: non-esterified fatty acid NFKB: nuclear factor KB ORO: oil red O PAS: Periodic acid shift PEPCK: phosphoenolpyruvate carboxykinase PPAR: peroxisome proliferator-activated receptor PGEz; prostaglandin E2 PUFA: polyunsaturated fatty acids RXRa: retinoid X receptor a SCAP: SREBP-cleavage activating protein SCD: Stearoyl-CoA desaturase SREBP: sterol regulatory element-binding protein SREs: sterol —regulatory elements SIP: Site 1 protease SZP: Site 2 protease VLCFA: very long chain fatty acid xiv INTRODUCTION Fatty acids affect many structural, metabolic, and regulatory components of cells. It has become increasingly apparent that fatty acids are capable of regulating, either directly or indirectly, the expression of numerous genes, both positively and negatively [1-4]. The impact of fatty acids on regulatory systems is determined by the type and quantity of fatty acid. The enzymes involved in the modification of fatty acid structure include fatty acyl CoA synthetases and thioesterases, fatty acid elongases and desaturases, peroxisomal 0 oxidation (chain shortening) and phospholipid and triglyceride lipases. Our lab has an interest in how the fatty acid elongases lead to structural changes in fatty acids that control transcription via the control of fatty acid- regulated transcription factors, peroxisomal proliferate activator a (PPARa) and sterol regulatory element binding protein-1 (SREBP-l) and carbohydrate regulatory element binding protein (ChREBP). Four fatty acid elongases (Elovll, 2, 5 and 6) and 3 desaturases [A5 desaturase (A5D), A6 desaturase (A6D) and A9 desaturase (A9D)] are expressed in rat, mouse and human liver. Among these enzymes, the 3 desaturases are well studied, especially A9D. These enzymes are regulated by hormones, development and dietary lipids [5-10]. A9D knockout mice are resistant to dietary fat-induced hepatic steatosis and have improved insulin sensitivity in muscle [1 1-13]. As such, this enzyme has been recognized as a drug target for obesity treatment recently. Compared to the desaturases, however, we are still in the early stages of understanding how fatty acid elongases are regulated and what their roles are in physiology. Moreover, fatty acid structure plays a key role in the regulation of transcriptional factors [14]. As such, enzymes that modify fatty acid structure are also likely to impact gene expression. The goal of my research has been to gain information about the regulation of the fatty acid elongase family. In addition, I wanted to determine if there was any evidence in support or against the notion that fatty acid elongases play a role in the control of hepatic gene transcription and lipid metabolism in vitro and in vivo. Chapter 1 Literature Review 1.1. Significance of fatty acid metabolism 1.1.1. Fatty acid metabolism 1.1.1.1. General features of fatty acid 1.1.1.1.1. Classification of fatty acids Fatty acids are biochemical molecules that are composed of a hydrocarbon chain and a carboxyl group. According to the number of double bonds in the polycarbon chain, fatty acids can be classified to saturated fatty acid and unsaturated fatty acid. Saturated fatty acid, such as palmitic acid (16:0), contains no double bonds in the carbon chain. Unsaturated fatty acids, like oleic acid (18:1,n-9) contain one double bond while linoleic (18:2,n-6) and arachidonic acid (20:4,n-6) contain more than 1 double bonds. Therefore, unsaturated fatty acids are further classified into monounsaturated fatty acid (MU FA), such as oleic acid, and polyunsaturated. fatty acid (PUF A) (Fig. 1.1). Depending on the position of the first double bond relative to the methyl end of fatty acid, PUFA can be classified into two major groups, n-3 and n-6. Sometimes the symbol to is subtituted for the lowercase letter 11, making it (0-3 and (0-6. These two groups of PUFA are not convertible and have different biological fimctions. Alpha(a)-linolenic (ALA, 18:3,n-3), docosahexaenoic (DHA, 22:6, n-3) and eicosapentaenoic (EPA, 20:5, n-3) acids are representative n-3 fatty acids. Linoleic acid (LA, 18:2, n-6) and arachidonic acid (AA, 20:4, n-6) are n-6 fatty acids (Fig. 1.1). 1.1.1.1.2. Essential fatty acids The human body can produce all but two of the fatty acids it needs. These two essential fatty acids are 18:2,n-6 and 18:3,n-3; both are widely distributed in plant and fish oils. Since these fatty acids can not be made in the body from other fatty acid substrates and must be supplied by the diet, they are called essential fatty acids. Essential fatty acids are parent compounds of the n-3 and n-6 fatty acid series, respectively. In the body, essential fatty acids are assimilated into membranes as complex phospholipids; they are also used to produce hormone-like substances that regulate a wide range of functions, including blood pressure, blood clotting, blood lipid levels, the immune response, and the inflammation response [15]. 1.1.1.2. Dietary fatty acid metabolism Dietary lipids are absorbed from the small intestines and transported in the blood to various tissues. The transport vehicles for these lipids are large lipoprotein complexes called chylomicrons. The liver removes chylorrricron remnants fi'om the circulation; cholesterol and triglycerides are repackaged into very low-density lipoproteins (V LDLs) and secreted from the liver for delivery to various tissues. 16:0 CH3 HOOC /\/\/\/\/\/\/\/ HOOC CH3 HOOC $3311 COOH 20:511'3 Figure 1.1. Structure of fatty acids. According to the number of double bond in the carbon chain, fatty acid can be classified as a saturated fatty acid (SFA) [Palmitic acid,l6:0], monounsaturated fatty acid (MUFA) [Oleic acid, 18:1,n-9], or polyunsaturated fatty acid (PUFA), such as linoleic acid (18:2,n-6), arachidonic acid (20:4,n-6) and eicosapentaenoic acid (20:5,n-3). Plasma lipids can be derived from the diet (chylorrricrons), liver (V LDL) or mobilized from storage depots (adipose tissue) as non-esterified fatty acids (NEFA). Fatty acids in complex lipids, like triglycerides, are excised by the action of lipoprotein lipase. Plasma NEFAs are transported in the blood, loosely bound to albumin; NEFA are taken up by multiple tissues, including the liver, adipose tissue and muscle. Fatty acid can also be synthesized de novo fiom glucose [16]. When plasma fatty acids reach cell membrane, they enter cells via diffussion or by fatty acid transport proteins, e.g., FATP or CD36 [17]. Once inside the cell, fatty acids are rapidly converted to fatty acyl CoA by the action of fatty acyl CoA synthetases (ACS); some members of the FATP family also can convert NEFA to Fa-CoA during the transport process [18]. The formation of fatty acyl CoA is the rate-limiting step for nearly all subsequent metabolic transformations of fatty acid, such as elongation, desaturation, [i-oxidation (mitochondrial or peroxisomal), or assimilation into complex lipids. Microsomal mono-oxidation and eicosanoid synthesis, however, utilizes the NEFA form of the fatty acid as substrate [14](Fig. 1.2). 1.1.1.3. De novo synthesis of fatty acids Fatty acid synthesis occurs primarily in the cytoplasm of liver, adipose tissue, central nervous system and lactating mammary gland [19]. Among these tissues, liver has the highest rate for fatty acid synthesis. During caloric excess, the liver converts glucose to pyruvate. Pyruvate is converted to citrate in the mitochondrial (tricarboxylic acid cycle), Excessive citrate (not used in aerobic respiration) is transported out of the mitochondria to the cytosol where ATP citrate lyase converts citrate to acetyl-CoA. Intracellular [High] {FABP} ACS {ACBP} Eioosanoids‘ [N EF A] [NEFA]—->[FA-CoA] * TE . I LPL Elongation (W cm Desaturation I Ifl-Oxidation l«——’ P i .l Neutral Lipid '0‘” "'— Stora evs Export Acyiation ~ g TAG HSL Extracellular VLDl. Chylomlcron NEFA Figure l. 2. Overview of cellular fatty acid metabolism. When the plasma fatty acid reach the cell membrane, they are either via diffussion or by fatty acid transprot proteins (F ATP or CD3 6) to enter into the cell. Inside, fatty acid must be activated before proceeding through metabolism. Activation consists of conversion of the NEFA to its CoA derivative under the control of acyl CoA synthetases (ACS). The formation of fatty acyl CoA is the rate-limiting step for fatty acid further modification, including elongation/desaturation, B-oxidation (mitochondrial or peroxisomal), or assimilation into complex lipids. Acetyl-CoA is a substrate for acetyl CoA carboxylase 1 (ACCl) which converts acetyl- CoA to malonyl CoA. Malonyl CoA is the rate-limiting substrate for fatty acid synthesis. A multi-enzyme complex, called fatty acid synthase (FAS), then uses malonyl-CoA, acetyl-CoA, and NADPH to elongate fatty acids in two-carbon increments in the cytosol [20]. The usual end product for this reaction in rodents is palmitate (16:0) [21]. Insulin, triiodothyronine (T3), glucocorticoids and glucose induce, while 2C18 PUFA, glucagon, and epinephrine suppress de novo fatty acid synthesis [14]. 1.1.1.4. Modification of dietary and endogenous fatty acids: elongation and desaturation The liver plays a central role in whole body glucose, fatty acid and cholesterol metabolism. Once fatty acids enter hepatocytes, they are converted to their active form, acyl CoA. They enter pathways that lead to their elongation, desaturation, B-oxidation or assimilation into complex lipids (phospholipids, triglycerides and cholesterol esters). Fatty acid elongation, desaturation and B-oxidation are three chemical modifications that alter fatty acid structure. Each of these reactions occurs in most tissues. Our focus in the following discussion is on hepatic fatty acid elongation and desaturation. Fatty acid elongation occurs in both the endoplasmic reticulum (ER) and the mitochondria. The elongation in ER represents the predominant pathway and malonyl CoA is the source of the added carbons. The process resembles the elongation steps of de novo fatty acid synthesis, but involves separate enzymes. These reactions consists of four sequential and independent steps: 1) a condensation between fatty acyl CoA and malonyl CoA to form 3-ketoacyl-CoA mediated by a fatty acid elongase (Elovl) [also known as [3- acylketo CoA synthase] ; 2) a reduction of the 3-ketoacyl-CoA to form 3-hydroxyacyl- CoA using NADPH, mediated by B-acylketo CoA reductase; 3) a dehydration of 3- hydroxyacyl-CoA to trans-2,3-enoyl-CoA mediated by (B-hydroxyacyl CoA dehydrase); 4) a reduction of trans-2,3-enoyl-COA to acyl CoA, mediated by trans-2-enol CoA reductase [22]. The overall reactions are shown schematically in Fig. 1.3 [23]. The condensation step catalyzed by the fatty acid elongase (Elovl) is the regulated and rate- limiting step for fatty acid elongation [24]. Unlike de novo fatty acid synthesis, these reactions are separable and are not catalyzed by a multifunctional enzyme. Mitochondrial elongation is a minor pathway and acetyl CoA is the source of the added carbons. In the mitochondria, fatty acid elongation occurs by a reversal of [3- oxidation, except that one NADPH and one NADH are required (IS-oxidation yields two NADH). Mitochondrial fatty acid elongation acts primarily on fatty acyl CoA substrates shorter than 16 carbons [25]. Desaturation of fatty acids introduces a double bond into the fatty acid; this reaction also occurs in ER of many tissues. This reaction requires an electron transport system involving: cytochrome b5, desaturase and NADPH- cytochrome b5 reductase [9]. In mammals, there are 3 distinct fatty acid desaturases (A5, A6 and A9 desaturase). Each has a different substrate requirement and generates a double bond at specific carbon bonds relative to the carboxyl end. For example, A5-desaturase inserts a double bond between the 5th and 6th carbon bond from the carboxyl end. A minimum chain length of 16-18 carbons is required for the desaturation, mammals can not synthesize n-6 and n-3 fatty acids de novo. Desaturation and 2-carbon elongation fiinction together in metabolic Fatty acyl-CoA Condensation Elongase 3-Ketoacyl-CoA Reduction KAR 3-HydroxyacyI-CoA Dehydration Dehydratase Trans-2,3-enoyl-CoA Reduction I TER Elongated Fatty acyl-CoA Figure 1. 3. Microsomal fatty acyl elongation. The elongation in ER represents the predominant pathway and malonyl CoA is the source of the added carbons. The process consists of four sequential but independent reactions: 1) a condensation between fatty acyl CoA and malonyl CoA to form 3-ketoacyl-CoA ; 2) a reduction of the 3-ketoacyl- CoA to form 3-hydroxyacyl-CoA using NADPH; 3) a dehydration of 3-hydroxyacyl-COA to trans-2,3-enoyl-COA; 4) a reduction of trans-2,3-enoyl-CoA to acyl CoA KAR, 3- ketoacyl-CoA reductase. TER, trans-2,3-enoyl-CoA reductase. 10 Elovl6 1.4A q 1620 —> 1830 491;] I 16:1 —" 18:1 1-43 Elov15&6 Diet N6; 18:2 —>18:3 ->20:3 ->20:4 ->22:4 ->24:4 ->24:5 ->22:5 A60 Elovl5 A50 Elov12 Elov12 A‘D pfi-Ox N3: 18:3 —> 18:4 ->20:4 ->20:5 ->22:5 ->24:5 ->24:6 ->22:6 Essential Fatty Acids Figure 1. 4. Scheme of endogenous fatty acid synthesis pathway. 4A described the de novo fatty acid synthesis from glucose. ElovlS, Elovl6 and A9-desaturase (A9D) are involved in this pathway. The end products of this pathway (16:0, 16:1 and 18: l) accumulate in tissues. 4B illustrates the polyunsaturated fatty acid synthesis pathway from essential fatty acids. 18:2n6, and 18:3n3 are converted to long chain unsaturated fatty acids in vivo by a series of elongation (Elov12 & -5) and desaturation (As-desaturase [A5 D] and A6 -desaturase [A6D] reactions that take place in the endoplasmic reticulum and the peroxisome via fl-oxidation (pfl-Ox) 11 pathways to generate polyunsaturated fatty acids. As illustrated in Fig. 1.4, these two chemical modifications often alternate in the synthesis of long chain PUFA. The predominant saturated and monounsaturated fatty acids synthesized in mammals are long chain fatty acids (Fig. 4A). The major products of this pathway are palmitate (16:0), palmitoleate (16:1,n-7), oleate (18:1,n-9) and vaccinate (18:1,n-7). Mead acid (20:3,n-9) can arise from this pathway when essential fatty acids (18:2,n-6 and 18:3,n-3) are deficient or when animals are fed olive oil and treated with peroxisome proliferators, like WY14,643 [26]. Stearoyl-CoA desaturase (SCD), also known as A9 desaturase, is the rate-limiting enzyme catalyzing the synthesis of 18:1 and 16:1. 18:1 is the preferred substrate for acyl- CoAzcholesterol acyltransferase (ACAT) and diacylglycerol acyltransferase (DGA'I), the enzymes responsible for cholesteryl ester and triglyceride synthesis, respectively [27]. In addition, 18:1 is the major MUFA in human adipose tissue and in the membrane phospholipid of most non-neuronal cells. The ratio of 18:0 to 18:1 has been implicated in the regulation of cell growth and differentiation through effects on membrane fluidity and signal transduction [10]. MUFAs also influence apoptosis and may have some role in mutagenesis of some tumors [28-30]. Overall, these fatty acids are important components of phospholipids; they also represent the largest energy storage reservoir in the form of triglycerides, and are the preferred fatty acids used for the esterification of cholesterol. A second pathway is for synthesis of unsaturated fatty acids utilizes the essential fatty acid precursors (LA, 18:2,n-6 and ALA, 18:3,n—3) to form the long-chain derivatives 12 arachidonic acid (AA, 20:4,n-6) and docosahexaenoic acid (DHA, 22:6,n-3), respectively (Fig. 1.4B). The first part of this metabolic pathway takes place in the endoplasmic reticulrun and consists of sequential alternating elongation and desaturation steps catalyzed by fatty acid elongase-2 (Elov12) and -5 (Elov15) and the desaturases, A5- desaturase (A5D) and A‘s-desaturase (A6D). The A6-desaturase is the rate-limiting step of the pathway. A part of the pathway that applies only to the synthesis of DHA (22:6,n-3) involves the elongation of eicosapentaenoic acid (EPA, 20:5,n-3) to 22:5,n-3 and 24:5,n- 3 by Elov12, followed by desaturation of 24:5,n—3 by A6D to form 24:6,n-3. 24:6,n-3 is then chain shortened in the peroxisome, i.e., peroxisomal B-oxidation, to form 22:6,n-3. These pathways, defined by Sprecher and his colleagues [31], are the only metabolic routes to generate AA (20:4, n6) and DHA (22:6, n3) from dietary precursors, 18:2,n-6 and 18:3,n-3 (Fig. 1.4 B). 18:2,n-6, 20:4,n-6 and 22:6,n-3 are the major PUFA accumulating in membranes of all cells. Of the two essential fatty acids (18:2,n-6 and 18:3,n-3), linoleic acid (18:2,n-6) is the predominant essential fatty acid in the diet. Dietary abundance, coupled with the fact that much of dietary 18:3,n-3 is B-oxidized limits the availability of 18:3,n-3 for PUFA synthesis [32]. As a result, little 18:3,n—3 accumulates in tissues, appears in the blood or is converted to long-chain PUFA. The products of these reactions provide substrates for complex lipid synthesis; such as: 1) incorporation into phosphoglycero- and phospho-ether lipids in membranes (AA and DHA), neural tissue is enriched in DHA. Little AA and DHA appear in sphingolipids; 2) substrates for eicosanoid (prostaglandins, thromboxanes, leukotrienes & 13 prostacyclins are derived from 20:4,n-6) and docosanoid synthesis (resolvins & protectins are derived from 22:6,n-3); 3) B-oxidation for energy metabolism; 4) Stored as neutral lipids, triglycerides and cholesterol esters. AA, but little DHA, appears in cholesterol esters (unpublished observation). Cunnane’s laboratory used isotope tracer and other methods to show that membrane lipids in liver and brain preferentially use exogenous long chain PUFAs rather than those produced endogenously [33]. AA that is not incorporated into membranes may be used by eicosanoid metabolism which requires a readily available supply at all times. AA is the major precursor for the synthesis of eicosanoids, which are powerful cellular regulatory substances and mediators of inflammation, platelet aggregation, T-cell proliferation, lymphocyte migration, vasoconstriction and dilation, and the production of several immune and inflammatory substances [34]. EPA (20:5,n—3) is a poor substrate for cyclooxygenase-l and -2. Eicosanoids derived from 20:5,n-3 typically have weak activity when compared to the AA-derived eicosanoids [17,35,36]. Thus, EPA can moderate the production and activity of AA-derived eicosanoids. It is believed that eicosanoids derived from n-6 fatty acids contribute to the development and deterioration of several chronic diseases [3 7-40]. DHA, which is the end product of the n-3 PUFA pathway, is the most abundant n-3 PUFA in all tissues. It is critical for the development of central neural system and retina [41-43]. Deficiency of DHA causes a similar phenotype to essential fatty acid deficiency, including severe macular dystrophy and impaired cognitive function. 14 1.1.2. Fatty acid regulation of gene transcription Fatty acids regulate the expression of numerous genes, both positively and negatively [14,44-46]. Fatty acids directly bind to and regulate several nuclear receptors, including members of the peroxisome proliferator-activated receptor (PPAR) family (PPARa, 0/8, 71 & 72), liver X receptor (LXRu, but not LXRB), retinoid X receptor a (RXRu) , hepatic nuclear factor-4a (HNF-4 a) [47-55]. In this case, fatty acids act like hydrophobic hormones to control the activity of these key transcriptional factors. Of these, however, the PPAR family are generally considered bona fide in vivo targets of fatty acids [14]. In contrast, fatty acids also regulate other transcriptional factors through indirect mechanism by changing their nuclear abundance or activity. These transcription factors include sterol regulatory element binding protein-1 (SREBP-l, a & c) [56], MAX-like factor-X (MLX), carbohydrate regulatory element-binding protein (ChREBP) complex [55,57] and nuclear factor KB (NFicB) [14]. In vivo studies with rats and mice have established that hepatic PPARa, SREBP-l, ChREBP and MLX are target for control by dietary fatty acids [5 8-61 ]. PPARa induces fatty acid oxidation, whereas SREBP-l induces fatty acid synthesis. ChREBP/MLX induce glucose utilization for fatty acid synthesis [55,57,62]. These mechanisms control, not only hepatic lipid metabolism, but also whole body lipid metabolism by affecting VLDL composition and secretion. As such, these regulatory pathways may contribute to the onset and progression of chronic disease, such as 15 atherosclerosis, diabetes and obesity [44,63]. This review will mainly focus on the three transcriptional factors, PPARoc, SREBP-l and the ChREBP/MLX heterodimer. 1.1.2.1 PPARa PPARa is the predominant PPAR subtype expressed in liver. It can be activated by PUFA and their oxidized derivatives as well as by hypoliporrric drugs of the fibrate family, including fenofibrate or gemfibrozil [51]. PPARa plays a role in the regulation of an extensive network of genes involved in glucose and lipid metabolism [58,64-67]. PPARa, as well as other PPARs, bind DNA as a heterodimer with RXR at direct repeat in the 5’ flanking regions of many genes, such as camitine palmitoyltransferase (CPT), acyl- CoA oxidase (AOX), and fatty acyl-CoA synthetase-1 (ACSl) [68-70]. Like other nuclear receptors, ligand binding recruits co-regulators to the promoter and changes gene transcription [71]. PPARa was the first transcription factor identified as a prospective fatty acid receptor [51]. PUFAs are recognized as the PPARa activators [53]. This finding parallels the metabolic findings that PUFA induce fatty acid oxidation [72]. In cells, non-esterified fatty acids (NEF A) serve as PPARa ligands. Since NEFA bind PPARa with different affinities, the composition of intracellular NEFA is an important determinant in the control of PPAR activity. Intracellular NEFA composition is affected by the concentration of exogenous fatty acids entering cells, their rate of removal via acyl CoA-synthetase— dependent and -independent mechanisms, e.g. microsomal mono-oxygenation, and the return of NEF A or oxidized lipids to the NEFA pool as a result of lipid metabolism [14]. The relative abundance of several putative fatty acid ligands for PPARa in rat primary l6 hepatocytes is 20:4,n-6 = 18:2,n-6 = 18:1,n-9 > 22:6,n-3 > 18:3,n-3 or n-6 = 20:5,n-3 [53]. Challenging cells with 18:1,n-9, 18:2,n-6 or 20:4,n-6 has little impact on the mass of these fatty acids in the NEFA fraction of cells. In contrast, the addition of 20:5,n-3, 22:5,n-3 and 22:6,n-3 to hepatocytes significantly enriches the NEFA fraction of cells with these fatty acids. Of the 3 n-3 PUFA, 20:5,n-3 significantly induced PPARa activity and PPARa target genes, like AOX and CYP4A [53]. Structural analysis of PPARB indicated that the carboxyl end of fatty acid inserts into the hydrophobic pocket of ligand binding domain (LBD) [73]. 20:5,n-3 and 22:6,n-3 have a similar structure near the carboxyl end of carbon chain. However, elongation of 20:5,n-3 to 22:5,n-3 increases the rigidity of the fatty acid, which may influence its binding to PPARa [14]. This indicates that major changes in intracellular NEFA composition and mass alone may not be sufficient to trigger a PPARa response. Fatty acid structure may be a key factor in the control of PPARa activity. Although PUFA are weak agonists of PPAR compared with pharmacological agonists (e.g., fibrates and thiazolidinediones), PUFA have significant effects on cell physiology. PUFA-mediated induction of PPARa-regulated genes shifts hepatic metabolism away from lipid synthesis and storage toward lipid oxidation [74,75]. It also affects insulin sensitivity in various tissues, particularly skeletal muscle [76]. As such, PUFA composition of cells will likely impact gene expression and sensitivity to key hormonal regulatory systems. 17 1.1.2.2. SREBP-l SREBPs are basic helix-loop-helix-leucine zipper (bHLH-LZ) transcription factors involved in cholesterol, fatty acid and complex lipid synthesis [77]. SREBPs bind DNA cis-regulatory elements called sterol-regulatory elements (SREs) as dimers. There are two separate genes encoding the SREBP-1 and SREBP-2. SREBP-1 regulates the expression of genes involved in fatty acid and triglyceride synthesis, while SREBP-2 regulates the expression of genes involved in cholesterol synthesis [78]. Two isoforrns of SREBP-1 have been described. These isoforms, SREBP-1a and SREBP-1c, arise from differential promoter and exon usage. SREBP-1c is the predominant SREBP-l isoforrn expressed in rodent and human liver. Despite its greater abundance, SREBP-1c is reported to be the weaker of the two transcription factors in activating gene transcription. This is because SREBP-lo lacks a 29 acidic rich arrrino acid region present in SREBP-1a. When SREBPs are first translated as large precursor proteins (~125 kd). These proteins are tethered to the endoplasmic reticulum (ER). SREBP binds a second membrane protein SREBP-cleavage activating protein (SCAP) at its C-tenninal end. A third family of proteins, INSIGs, interacts with SCAP in the endoplasmic reticulum. Depletion of sterol from cells leads to the movement of the SCAP-SREBP complex fiom the endoplasmic reticulum to the Golgi. SCAP plays an escort role for this movement, while INSIG interacts with SCAP and prevent this movement when intracellular sterols are high [79,80]. Two proteases, called Site 1 protease (SIP) and Site 2 protease (SZP) are present in the Golgi. These proteases cleave SREBP precursors to release the mature nuclear form of SREBP (nSREBP, ~65 kd) from the Golgi. SREBPs dirnerized and bind l8 to importin-B for transport to the nucleus. Once in nuclei, SREBP bind as dimers at SRE in promoters of target genes. SREBP binding to SREs promotes recruitment of co- activators, e.g., CBP, RNA polymerase II and initiates gene transcription. There are no reports of SREBP functioning as receptors for either cholesterol or fatty acids. As such, genes regulated by SREBPs are controlled by factors affecting the nuclear abundance of SREBP. In the liver, SREBP-2 is more sensitive to the cholesterol mediated inhibition than SREBP-1. In contrast to SREBP-2, the nuclear content of SREBP-1 is suppressed by unsaturated fatty acids. This phenomenon is observed in livers of animals fed PUFA- containing diets as well as in primary hepatocytes and some, but not all, SREBP-1c- expressing cells lines treated with PUFA [74,81]. The effects of PUFA on SREBP-1 levels are very complex. It has been attributed to inhibition of transcription of the SREBP-1c gene as well as enhanced turnover of the mRNA encoding SREBP-1, interference with the SREBP-1 processing, and enhanced proteasomal degradation [61,82]. The effect of 20-22 carbon PUFA on SREBP-1 mRNA and gene transcription is similar. 22:6,n-3, however, has an added impact on controlling SREBP-1 nuclear content. 22:6,n-3enhances 26S proteasomal degradation of nuclear SREBP-1, but not the SREBP- 1 precursor or nuclear SREBP-2 [83]. As such, 22:6,n-3 is the most potent fatty acid suppressor of SREBP-1 nuclear abundance and SREBPl-mediated gene expression. 1.1.2.3. ChREBP/MLX ChREBP and MLX are bHLH-LZ transcription factors. They bind as heterodimers to carbohydrate regulatory elements (ChoRE) located in promoters of 19 glucose-responsive genes, like L-type pyruvate kinase (LPK), ACC, FAS, 814, SCD-l and GLUT2 [62,84-86]. Both our group [55] and the Postic group [57] have reported that components in the ChREBP/MLX heterodimer are regulated by PUFA in liver and primary hepatocytes. However, the results from these two groups are not identical. Postic’s group indicated that PUFA suppressed ChREBP activity by enhancing ChREBP mRNA decay as well as inhibiting its nuclear translocation. Our group reported that n-3 PUFA have no effect on ChREBP nuclear abundance, but suppressed MLX nuclear abundance. This controversy may come from the different animal species as well as the experiment design. Whatever the case, finding that PUFA control ChREBP/MLX nuclear abundance provides an explanation for those fatty acid-regulated hepatic genes that were independent of PPARa and SREBP-l [5 5]. 1.1.2.4. Other transcription factors regulated by fatty acids 1.1.2.4.1 Liver X receptor (LXRa) LXRs were first identified as fatty acid regulated transcriptional factors by Brown and Goldstein group [87]. LXRs, like PPARs, belong to the steroid-thyroid receptor superfarnily. There are two subtypes, LXRa and LXR B in this family. LXRa is the predominant one expressed in liver. LXRs bind oxysterols and regulate the expression of genes involved in hepatic bile acid synthesis, reverse cholesterol synthesis, lipogenesis and glucose uptake [81,88,89]. PUFA antagonize oxysterol activation of LXRu in HEK 293 and hepatoma cell lines by interfering with oxysterol binding [54,87]. However, a recent study from our group showed that LXR-regulated transcripts, such as CYP7A, ABCAl, ABCGS and ABCG8, were not sensitive to PUFA suppression in rat primary 20 hepatocytes and in liver [52]. The condition used to access PUFA control of transcriptional factors in this study was sufficient to suppress SREBP-1c and induce PPARa targeted genes. These observations raised doubt as to whether LXRa were bona fide targets for PUFA control in vivo. 1.1.2.4.2 Hepatic nuclear factor-4 (HNF4) HNF 4 is an orphan nuclear receptor regulating an extensive network of genes controlling glycolysis and lipogenesis in liver and other tissues [14]. Few of these genes are regulated by dietary PUFA in vivo except L-PK, glucose 6 phosphatases, apolipoproteins A1 and C111. Our laboratory first reported that HN F -4 recognition sequence is a component of the PUFA-response region of L-PK gene [90]. Later, the Bar- tana group showed evidence that HNF4a bound fatty acyl CoAs. They suggested that fatty acyl CoA might account for the effect of fatty acids on transferring and Apo CIII promoter activity [47]. However, structural studies on HNF-4 protein indicated that fatty acids and not fatty acyl CoA co-crystallized with HNF-4 ligand binding domain. These findings triggered controversy over HNF-4 ligands. Recent studies from our laboratory showed that 20:5,n-3 treatment in rat primary hepatocytes promotes transient change HNF-4u—LPK promoter interaction, but no effect on HNF4 transactivation on LPK promoter [55]. 20:5, n-3 is a minor fatty acid in the pool of NEFA and esterified lipid fraction of liver and hepatocytes [83]. Greater than 99% of intracellular 20:5,n-3 was esterified after hepatocytes were treated. Intracellular non-esterified 20:5,n-3 is highest at 1.5 and lowest at 24 hours after challenging cells with 20:5,n-3 [55]. This suggest that non-esterified 20:5,n-3 can regulate HNF-4 binding activity to LPK promoter. The 21 mechanism for this control, however, is unknown. In the view of the Jump lab, this control does not involve direct binding of 20:5,n-3 by I-INF-4. This discussion of fatty acid regulated transcription factors highlights those factors that are well-established targets for PUFA control, i.e., PPARa and SREBP-1, and those that are either recently identified (ChREBP/MLX) or there is some controversy (LXRor and HNF-4a). An underlying fact is that the type and quantity of intracellular fatty acid controls these transcription factors and their regulatory networks. This thesis focuses on a metabolic pathway (fatty acid elongation) that alters the type of fatty acid in the cells. I am predicting that changes in fatty acid elongation will impact certain transcriptional regulatory networks. 1.1.3. Fatty acid and chronic disease 1.1.3.1. Cardiovascular disease Numerous studies have revealed the benefits of n-3 PUFA on cardiovascular disease (CVD) [91-93]. N-3 PUFA can be obtained fiom two dietary sources: seafood and certain nut and plant oils. Fish or fish oil contains EPA and DHA, whereas canola and walnut oil contain ALA. ALA is less potent than EPA and DHA. Mechanisms by which n-3 fatty acids reduce CVD risk are under active investigation. Research to date, however, suggests there are several mechanisms involved in the protective effect of PUFA on cardiovascular health. 22 Evidence fi'om epidemiological studies and clinical trials suggest that n-3 PUF A have an important anti-arrhythmic effect in patients with CVD [94,95]. However, the details of the anti-arrhythmic action of n-3 PUFA remain to be elucidated. In addition, n- 3 PUFA is reported to decrease risk for thrombosis due to decreases in platelet aggregation. A decrease in thromboxane (TXAz) coupled with increases in prostaglandin (PG12 and PGI3) production, decrease in whole blood viscosity and an increase in bleeding time [96]. As described above, eicosanoid metabolic products from AA (n-6 PUFA) are more biologically active than those derived from EPA (n-3 PUFA) [14]. Supplementation of n-3 PUFA will replace n-6 PUFA as the precursor of eicosanoids, while increasing the amount of n-6 PUFA consumption contributes to the development of thrombi and athomas. PUFA is also reported to decrease triglyceride and remnant lipoprotein levels by replacing saturated fat in diet. Additional benefit is shown by n-3 PUFA which consistently lowers blood triacylglycerol levels in hypertriglyceridemic patients. In contrast, n-6 fatty acids either have no effect or increase blood triglycerides [97]. In addition, PUFA can decrease rate of growth of the atherosclerotic plaque by improving endothelial function and reducing inflammatory responses [98]. 1.1.3.2 Diabetes Type 2 diabetes (non-insulin dependent diabetes melittus, NIDDM) is the most prevalent form of diabetes in western societies. NIDDM is a metabolic disorder characterized by hyperglycemia in the presence of insulin resistance and hypertriglyceridemia. Skeletal muscle is the principal site of insulin-mediated glucose disposal. Campbell and colleagues showed that insulin resistance and hyperinsulinemia 23 are inversely associated with the amount of C20 and C22 fatty acids in the phospholipids of muscle cell membranes in patients with coronary heart disease and normal volunteers [99]. Increasing the amount of C20 and C22 PUFA in membrane leads to an increase in membrane fluidity, insulin receptor and insulin action [100]. High dietary intake of saturated and trans fat interferes with the elongation and desaturation of essential fatty acids to AA, EPA and DHA. These fatty acids also contribute to the development of insulin resistance [100,101]. Skeletal muscle triglyceride levels are also inversely related to insulin action [102]. Clinical trials of dietary supplemention of n-3 PUFA to type 2 diabetic patients lowers triacylglycerol concentrations with no adverse effects on glucose control [103]. Leptin is a hormone that suppresses appetite and enhances basal metabolic rate. As such, it has a remarkable effect on the regulation of body fat [104]. It is reported that serum leptin levels are influenced in patients with type 1 diabetes by the type of fat in the diet [105]. In particular, n-3 PUFA has been found to decrease leptin gene expression both in vivo and in vitro [106]. 1.1.3.3 Inflammation Inflammation is characterized symptomatically by pain, redness, and swelling. The disordered or excessive inflammation also contributes to the loss of function. This results from the release of inflammatory mediators, predominantly from activated leukocytes that migrate into the target area. Among the key inflammatory mediators are prostaglandin E2 (PGEz) and leukotriene B4 (LTB4), which are derived from the n-6 PUFA, AA [107]. Eicosanoids derived from AA and EPA have very similar molecular 24 structures, but very different biologic effects. Those derived fiom EPA are generally much less potent in inflammation than those derived from AA. As such, a majority of n-6 fatty acids will result in a proinflarrrmatory status by producing prostaglandins of the :2 series and leukotrienes of the 4 series. As the ratio of n-3 to n-6 increases, m0re prostaglandins of the 3 series and leukotrienes of the 5 series are produced. As such, n-3 fatty acids exert anti-inflammatory effects via competitive inhibition of the n-6-derived proinflammatory eicosanoids [108]. The immune response also plays a role in the development of inflammation by producing immunologic mediators such as cytokines. PUFA have been shown to have effects on immune response [109]. Dietary supplementation of n-3 fatty acids influences the cytokine production and lymphocyte proliferation. This effect is more dramatic on older versus younger women. 1.1.3.4 Cancer Cancer is a disease characterized by abnormal cell division without any control. Many cancers have a high mortality rate. Epidemiological studies indicate that women in countries with high-fat diets have a higher risk (5-fold) of breast cancer than women in countries with low-fat consumption. Such observations strongly suggest that a high intake of dietary fat could increase breast cancer risk [110,111]. This also provides evidence that dietary or exogenously derived fatty acids may play an important role in the carcinogenesis, evolution and/or progression of breast cancer. Several studies also showed that diets containing corn oil, with high levels of n-6 PUFA, enhance breast and colon tumorgenesis in rodents. In contrast, diets enriched in n-3 PUFA, e.g., fish oil, 25 reduces carcinogenesis [112]. Saturated fatty acids have no discernible effect on mammary carcinogenesis or progression. Several mechanisms were proposed to explain the mechanism by which the intake of n-3 PUFA might lower the risk of cancer. Inhibition of eicosanoid biosynthesis from AA is the most important one among these mechanisms. High intake of n-6 PUFA experimentally induces various physiological and metabolic effects [112,113]. It can increase ornithine decarboxylase activity in colon mucosa which leads to the enhanced epithelial polyamine levels and increase colon crypt cell proliferation. It can also up-regulate activities of protein kinases, such as protein kinase C in rodent mammary gland and increases number of estrogen receptor binding sites. Prostaglandins, thromboxanes, leukotrienes and hydroxy and hydroxyperoxy fatty acids are involved in tumor initiation and promotion, cell proliferation, tissue invasion and metastatis. Tumor cells produce more eicosanoids than their normal cell counterparts; oxidized lipids ultimately derived from linoleic acid have been linked to increased cell growth and tissue metastasis. The finding that oleic acid and n-3 PUFA, specifically EPA, inhibits the desaturation reaction, the first step from linoleic acid to arachidonic acid and eventually to eicosanoid production, may partially explain their inhibitory effects on turnorigenesis. Clearly, the impact of dietary fat on chronic disease is significant. What is less clear is how chronic disease might impact enzymes involved in fat metabolism. Equally unclear is how changes in these enzymes impact cellular fat composition and impact the onset and progression of these diseases. This thesis will begin to address these important medical issues. 26 1.2. Fatty acid elongases The focus of this thesis is on fatty acid elongases. This section will review our current understanding to the role these enzymes play in lipid metabolism. Fatty acid elongases (Elovls) are the condensing enzymes that interact with 3-keto acyl-CoA reductase, a dehydratase and trans-2,3-enoyl-COA reductase to elongate fatty acids (Fig. 1.3). The rate of fatty acid elongation is determined by the activity of elongase (condensing enzyme). There are 7 distinct elongase subtypes (Elovl-l, -2, -3, -4, -5, -6 & -7) present in the mouse, rat and human genomes. Each has a specific substrate preference in terms of chain length and the degree of fatty acid unsaturation. The members of this family are predicted to have 5 or 6 transmembrane spanning domain that traverse the endoplasmic reticulum. Since the third transmembrane stretch is very long, it is not clear whether 5 or 6 transmembrane domains in this family protein [114]. Also, this protein family share a common structure of a single histidine-box motif (HXXHH), and an ER retention signal (KKXX-like) [115]. Elovl3 was the first elongase to be identified by Jacobsson group in 1997 [116]. It was originally named cig30 (Cold induced glycoprotein with molecular weight about 30kDa) because the gene can be induced significantly in brown adipose tissue when the mice are exposed to cold environment. In silica search identified two homologues of cig30, which were first termed Sscl and Ssc2 (sequence similarity to cig30) [114]. The official names for these two enzymes are Elovll and Elov12, respectively. Shortly after that, a murine long chain fatty acid elongase was identified by gene array analysis from 27 SREBP] transgenic mice liver [117]. This enzyme was called Elovl6. ElovlS was the first human elongase identified [118]. Later, the rat ElovlS was reported to have similarity to human Elov15 [119]. Mutation in the human Elovl4 gene were described as being responsible for an autosomal dominant macular degeneration [120] (Table 1.1). Because of their separate isolations and cloning over the years, fatty acid elongases have been given many names, e.g, Cig30, SSCl, etc. With the discovery of multiple subtypes in human and rodent genomes, the nomenclature was revised to the following. Elovl, e_lgngase of yery long fatty acid. ( Table 1.2). 28 nm_019422 bab31310 af170908 nm_134255 nm_148941 bc016468 ay053453 nm_019422 bab31310 af170908 nm_134255 nm_14894l bc016468 ay053453 nm_019422 bab31310 af170908 nm_134255 nm_148941 bc016468 ay053453 nm_019422 bab31310 af170908 nm_134255 nm_148941 bc016468 ay053453 nm_019422 bab31310 af170908 nm_134255 nm_14894l bc016468 ayos3453 nm_019422 bab31310 af170908 nm_134255 nm_148941 bc016468 ay053453 1 7 2 5 4 3 6 \Jk‘ 01W p m N d H m w 9 m M q H Oioa.> Uina~u p. 9“” *‘UIh° air» much nr-q F‘ Table 1. 1. Alignment of mouse fatty acid elongases Accession# Elov1# .................. MEAVVNLYHELMKHADPRIQSYPLMGSPLLITSILLTYVYFI ............ MAFSDLTSRTVRFYDNWIKDADPRVEDYLLMSSPLPQTIILGLYVYFV ----------- MEQLKAFDNEVNAFLDNMFGPRDSRVRGWFLLDSYLPTFILTITYLLSI .............. MEHFDASLSTYFKAFLGPRDTRVKGWFLLDNYIPTFVCSVIYLLIV MGLLDSEPGSVLNAMSTAFNDTVEFYRWTWTIADKRVADWPLMQSPWPTISISTLYLLFV -------- MDTSMNFSRGLKMDLMQPYDFETFQDLR---PFLEEYWVSSFLIVVVYLLLI ............ MNMS----VLTLQEYEFEKQFNENEAIQWMQENWKKSFLFSALYAAFI . . * LSLGPRIMANRKPFQLRGFMIVYNFSLVILSLY-IVYEFLMSGWLSTYTWRCDPIDFSNS TSLGPKLMENRKPFELKKAMITYNFFIVLFSVY-MCYEFVMSGWGTGYSFRCDIVDYSQS W-LGNKYMKNRPALSLRGILTLYNLAITLLSAY-MLVELILSSWEGGYNLQCQNLDSAGE W-LGPKYMKNRQPFSCRGILQLYNLGLTLLSLY-MFYELVTGVWEGKYNFFCQGTRSAGE W-LGPKWMKDREPFQMRLVLIIYNFGMVLLNLF-IFRELFMGSYNAGYSYICQSVDYSND V-VGQTYMRTRKSFSLQRPLILWSFFLAIFSILGTLRMWKFMATVMFTVGLKQTVCFAIY F-GGRHLMNKRAKFELRKPLVLWSLTLAVFSIFGALRTGAYMLYILMTKGLKQSVCDQSF * * i :. ooooo nil-Box PEALRMVRVAWLFMLSKVIELMDTVIFILRKKDGQVTFLHVFHHSVLPWSWWWGIKIAPG PRAMRMVHTCWLYYFSKFIELLDTIFFVLRKKNSQVTFLHVFHHTIMPWTWWFGVKFAAG G-DVRVAKVLWWYYFSKLVEFLDTIFFVLRKKTNQITFLHVYHHASMFNIWWCVLNWIPC S-DMKIIRVLWWYYFSKLIEFMDTFFFILRKNNHQITVLHVYHHATMLNIWWFVMNWVPC VNEVRIAAALWWYFVSKGVEYLDTVFFILRKKNNQVSFLHVYHHCTMFTLWWIGIKWVAG TDDAVVRFWSFLFLLSKVVELGDTAFIILRKR--PLIFVHWYHHSTVLLFTSFGYKNKVP YNGPVSKFWAYAFVLSKAPELGDTIFIILRKQ--KLIFLHWYHHITVLLYSWYSYKDMVA .I'* i it :::*i*. : l:* :i* : GMGSFHAMINSSVHVVMYLYYGLSALGPVAQPYLWWKKHMTAIQLIQFVLVSLHISQYYF GLGTFHAFLNTAVHVVMYSYYGLCAMGPAYQKYLWWKKHLTSLQLVQFVLVTIHIGQIFF GQSFFGPTLNSFIHILMYSYYGLSVF-PSMHKYLWWKKYLTQAQLVQFVLTITHTLSAVV GHSYFGATLNSFIHVLMYSYYGLSSI-PSMRPYLWWKKYITQGQLVQFVLTIIQTTCGVF GQAFFGAQMNSFIHVIMYSYYGLTAFGPWIQKYLWWKRYLTMLQLVQFHVTIGHTALSLY SGGWF-MTMNFGVHSVMYTYYTMKAA--KLKHPNLLPMVITSLOILQMVLGTIFGILNYI GGGWF-MTMNYGVHAVMYSYYALRAA--GFRVSRKFAMFITLSQITQMLMGCVINYLVFN * :i :* :a+ it :i t: a: MP8 ----- CNYQYPIIIHLIWMYGTIFFILFSNFWYHSYTKG ----- KRLPRAVQQ---- MED ----- CNYQYPVFLYIIMSYGCIFLLLFLHFWYRAYTKG ----- QRLPKTLE ----- KP ------ CGFPFGCLIFQSS-YMMTLVILFLNFYIQTYRKK---PVKKELQEKE ----- WP ------ CSFPLGWLFFQIG-YMISLIALFTNFYIQTYNKKGASRRKDHLKGHQNGSVA TD ------ CPFPKWMHWALIA-YAISFIFLFLNFYTRTYNEP ----- KQSKTGKTATNGI WRQEKG--CHTTTEHFFWSFMLYGT-YFILFAHFFHRAYLRP ----- KGKVASKSQ"" WMQHDNDQCYSHFQNIFWSSLMYLS-YLVLFCHFFFEAYIG ------ KVKKATKAE---- * a . it :t: _:* : --NGAPATTKVKAN ------------ --NGN---CKSKRH ------------ VKNGFPKAHLIVANGMTDKKA -~-- AVNGHTNSFPSLENSVKPRKQRKD-- SSNGVNKSEKALENGKPQKNGKPKGE 29 Elongase Sub e lovl 1 Genbank # Alias Chromosome # Location, mb Genbank # Alias Chromosome# Location, mb Genbank# Alias Chromosome # Location mb Genbank # Alias Chromosome # Location, mb Genbank # Alias Chromosome # Location, mb Genbank # Alias Chromosome # Location, mb Genbank # Alias Chromosome # Location, mb Mouse, Rat and Human. Table 1. 2. Chromosomal Location of Elongase Subtypes in Mme Rat HIM NM_019422 AAL71993 SSCl CGI-88, SSCl 4 1 1 117.3 138.9 43.2 AF 1 70908 AAH60809 SSC2 SSC2 13 17 6 41.2 29.5 11.1 BC016468 AAH34344 c1030 BLOB 19 1 10 46.3 251.6 103.7 NM_148941 Q9GZR5 AAH3 8506 ELO4 9 8 6 86.5 88.9 80.6 NM_134255 NM_134382 AAF70631 FAEl RELOl HELOl 9 s 5 80.5 83 53.2 AY053453 NM_134383 NPO76995 LCE, FACE E102 3 2 4 133.1 226.9 111.4 BAB31310, AKA050441 9130013K24RIK Q9NT66 13 2 5 106.8 39.5 60.1 30 1.2.1 Elovll Elovll was first discovered by its homology to Elov1—3. Complementation studies with yeast mutants with defective fatty acid elongation with the murine Elovll indicated that Elovl-l was involved in elongation of very long chain fatty acids (up to 26 carbons) and sphingolipid formation [121]. Sphingolipids are essential for cell proliferation. Abnormal sphingolipid synthesis hinders cell growth [122]. Numerous studies show that sphingolipids, as well as very long chain fatty acid synthase (V LCFAS) are important for the formation of functional lipid microdomains (lipid rafts) in plasma membrane [123]. Disruption of the yeast Elovll homologue, E103, leads to a broad range of defects including altered expression of the plasma membrane ATPase and lower glucose uptake [124]. The overlapping functions of yeast E103 and murine Elovll indicates that mammalian enzymes are also important in membrane related functions associated with the lipid rafts. Elovll is ubiquitously expressed in mouse tissues. This means most mammalian tissues are endowed with the capacity to synthesize VLCF A up to C26. Interestingly, the mRNA level of Elovll is particularly high in stomach, lung, kidney, skin and intestine. The common feature of these tissues is that they have separate compartments with widely different water content. Therefore, it is conceivable that higher Elovll levels are needed for VLCFA synthesis in order to reduce water permeability of the epithelial barrier [125]. Quaking and J irnpy are two mouse mutant strains that have dramatically decreased Elovll mRNA level. Both strains are deficient in nerve myelination throughout the central nerve system (CNS), which results in severe motor problems and early death. 31 Moreover, both strains show decreased elongation activity towards 20:0-CoA and 22:0- CoA and lower levels of VLCFA in the brain [126]. The severity of elongation defect is proportional to Elovll mRNA expression. 1.2.2 Elov12 When Elov12 (SSC2) gene was first described in 2000, there was no functional characterization for this enzyme. Investigators hypothesized that it may play a role in PUFA synthesis based on the complementation studies from yeast system [114]. Later on, both mouse Elov12 and human Elov12 were shown to use arachidonic acid (20:4,n-6), eicosapentaeonic acid (20:5,n-3), docosatetraenoic acid (22:4,n-6) and docosapentaenoic acid (22:5,n-3) in yeast and mammalian HEK 293 cells when they are transfected with the vector expressing Elov12 gene [117,127]. In contrast, Elov12 did not elongate saturated and monounsaturated fatty acids. Northern blot analysis revealed that the highest level of expression of Elov12 was found in mouse liver and testis, followed by brain and kidney [127]. 1.2.3 Elovl3 Compared to other mammalian fatty acid elongases, Elovl3 is the one most studied. Elovl3 was first cloned from brown adipose tissue (BAT) of mice exposed to cold temperature [116]. When mice were exposed to cold temperature for 3 days, the rrricrosomal fraction from BAT of Elovl3 ablated mice has decreased ability to elongate saturated C16-C22 substrates. There was, however, little difference in elongation activity after mice were exposed to low temperatures for 3 weeks [128,129]. Further analysis of 32 fatty acid profile in these mice revealed a transient alteration of several different fatty acid species in the mice afier 3 days of cold exposure but not in the mice exposed to cold for 3 weeks cold [129]. Moreover, there are almost no lipid droplets in BAT of Elovl3 ablated mice when they are under therrnoneutral conditions (3 0°C). Such studies support a role for short term, but not long term, adaptive changes in BAT lipid metabolism following cold exposure. Jacobsson’s group showed that cold exposure induced Elovl3 gene expression required PPARa regulation [130]. In cultured brown adipocytes, a mixture of norepinephrine, dexarnethasone, and the PPARa ligand Wy14,643, which rendered the adipocytes a high oxidative state, was required for substantial induction of Elovl3 expression. In contrast, this treatment suppressed Elovll mRNA levels. Interestingly, the LXR agonist, T0901317, repressed Elovl-3 expression [130]. A most prominent phenotypic feature in Elovl3 -/- mice was found in the skin. Skin fimctions as a water barrier and very long chain fatty acids are important for this physiological function. Elov13-/- mice showed an impaired water barrier function which was attributed to a change of VLCFA synthesis [128]. Although most of identified fatty acid elongases are expressed in murine skin [114,127], nothing was known about the specific roles of the individual enzymes in skin. In situ hybridization indicated that Elovl3 has a distinct expression in the skin that was restricted to the sebaceous glands and the epithelial cells of the hair follicles [128]. Recent studies from the Baler group revealed circadian regulation of Elovl3 in liver that involved RevErba and SREBP-1. This regulation was sex- and tissue-specific 33 [131]. The reason for circadian regulation of Elovl3 can be explained by the needs for the liver to synthesize long-chain fatty acids. The main fraction of intracellular VLCFAs in liver is esterified in various lipids, predominantly in Sphingolipids. As such, circadian expression of Elovl3 as well as other lipogenic enzymes may contribute to maintenance membrane fluidity, which consequently regulates a rhythm of the membrane-linked signaling pathways [13 1]. 1.2.4 Elovl4 Tissue expression of Elovl4 is one of the most restricted amongst the elongases; it is expressed predominantly in the retina and to a less extent in brain, and testis [120]. The high expression of Elovl4 in the retina suggest an involvement in the synthesis of docosahexaenoic acid, 22:6,n-3. Nearly 50% of all acyl chains in phospholipids are DHA, the highest content of all mammalian tissues. Mutation of Elovl-4 was the first mutation of a fatty acid elongase linked to human disease. Mutation of the Elovl4 locus is found in patients with the genetic autosomal dominant disease, Stargardts-like macular dystrophy [120]. Stargardts-macular dystrophy is one of several forms of macular dystrophy. This disease is characterized by decreased visual acuity, macular atrophy and extensive fundus flecks. Genetic mapping by Zhang’s group identified a 5-bp deletion in the Elovl4 gene that may be responsible for this for of inherited macular degeneration [120]. About the same time, Allikmets, et al [132] found another mutation in Elovl4 gene associated with this disease. This mutation results in a frame shifi and the truncation of the ELOVL4 protein, similar to the effect of 5-bp deletion. 34 In vitro studies show that the mutant Elovl4 protein exerts a dominant-negative effect by recruiting wild-type protein into perinuclear cytoplasmic inclusions [133]. The interaction between the wild-type and mutant forms of Elovl4 and the resultant alteration in the trafficking of the wild-type ELOVL4 protein suggest a mechanism for the pathology associated with in patients harboring the autosomal dominant Stargardts-like macular dystrophy genotype. 1.2.5 ElovlS The Elov15 gene encodes a protein of 299 amino acids, which shares 56.4% amino acid sequence identity with Elov12. Elov15 is involved in the synthesis of mono and polyunsaturated fatty acids of C16-C22, but not polyunsaturated fatty acids beyond C22 [127]. Elov15 is widely expressed with high levels in testis, adrenal gland, lung and brain [115]. This is consistent with the fact that these tissues contain very high levels of docosapentanoic acid, 22:5,n-3 and adrenic acid, 22:4, n-6. 1.2.6. Elovl6 The identification of Elovl-6 was described by Moon et al. in 2001. It was discovered in livers of transgenic mice overexpressing SREBP-1 [117]. This enzyme is ubiquitously expressed. High fat-containing tissues such as white and brown adipose tissue, liver, and brain, displayed high levels of Elovl6 transcripts, whereas the low fat- containing tissues like spleen, skeletal muscle, and heart exhibit low levels of the Elovl6 transcript. In vitro microsomal fatty acid elongation assays demonstrated that Elovl6 possessed elongase activity specific for C1 2-C16 saturated and monounsaturated fatty 35 acids. The enzyme is induced by SREBP]. Its expression is also induced in the liver and adipose tissue in the refed state after fasting, but suppressed by the presence of dietary PUFAs [117,134]. It encodes 267 amino acid residues and has 96% identity with the human homologue of 265 amino acids [115]. 1.2.7 Elovl7 In silica analysis of the human and mouse genomes revealed the existence of the seventh member in elongase family. This gene is located on chromosome #13 in mouse, and #2 in rat. It encodes 281 amino acids residues and contains a histidine-rich motif (HXXHH), and ER retension signal (KKXX-like), both typical for members of fatty acid elongase family. The histidines in the HXXHH motif may act with aspartate and glutamate to coordinate an Fe-O-F e cofactor to receive electrons from cytochrome b5 or a cytochrome b5-like domain in an NAD(P)H-dependent manner [114,117]. The similarities in sequence organization amongst the 7 proteins suggest Elovl7, i.e., BAB31310, mouse; Q9NT66, human, is a fatty acid elongase. Unfortunately, there is no experimental data on Elovl-7 substrate specificity or its regulation. 1.3. Statement of the Problem and Hypothesis. Multiple mammalian fatty acid elongases and fatty acid desaturases play a major role in determining cellular lipid content. Of these enzymes, the most extensively studied is A9D. Multiple factors regulate A9D expression. Studies with the D9D -/- mouse implicate this enzyme in the management of hepatic and whole body lipid metabolism, 36 energy expenditure and tissue sensitivity to insulin [12,135]. These studies have targeted A9D as a candidate enzyme for the development of inhibitors of its activity to manage of whole body lipid metabolism. In contrast to desaturases like D9D, we are in the early stage of understanding the role fatty acid elongases play in cell function and composition. We have little information on their regulation; we are unclear of their substrates and products. We do not know how they are regulated in chronic disease or which transcriptional regulatory networks control their expression. Studies in our laboratory have established a strong link between fatty acid structure and the control of key transcriptional regulatory networks controlling glucose and lipid metabolism [83,136]. Elongase activity alters fatty acid structure. As such, these enzymes have the potential to impact cellular lipid composition and affect key transcriptional regulatory networks. If so, then, elongases might join with A9D as targets for drug development in the management of fatty acid-linked chronic diseases. The goal of this study is to gain information about the regulation of the elongase family of enzymes. A second goal is to gain evidence in support or against the notion that fatty acid elongases play a dual role in hepatic physiology: to control hepatic lipid composition and to impact hepatic gene transcription both in cells (in vitro) and in the mouse (in vivo). I hypothesized that fatty acid elongases play a dual role in hepatic physiology: a) to regulate cellular levels of long chain unsaturated fatty acids, and b) to regulate cellular levels of ligands controlling hepatic gene expression. The aims are: 37 Aim 1: To identify substrates of elongases in rat primary hepatocytes. To identify regulatory factors controlling fatty acid elongase family expression. Aim 2: To elucidate the role of Elovl-2 & Elov15 on hepatic gene expression in rat primary hepatocytes. Aim 3: To determine how over expression of specific fatty acid elongases affect hepatic function and blood lipid composition. The outcome of these studies will provided new information on the role fatty acid elongases play in hepatic physiology. 38 Chapter 2 Tissue-Specific, Nutritional and Developmental Regulation of Rat Fatty Acid Elongases. 2.1. Introduction Most cells have the capacity to synthesize fatty acids from glucose, de novo. This pathway utilizes products from glycolysis and along with the two enzymes, acetyl CoA carboxylase and fatty acid synthase, generates palmitate (16:0). Insulin, T3, glucocorticoids and glucose induce, while C20 PUFA, glucagon and epinephrine suppress de novo lipogenesis [14,46,137]. Many cells also have the capacity to modify fatty acid structure through metabolic pathways that include desaturation, elongation, mono- oxidation and peroxisomal B-oxidation (chain shortening). Such modifications occur to fatty acids generated de novo as well as fatty acids derived from the diet. These metabolic pathways play an important role in the maintenance of membrane lipid composition, the generation of precursors for certain signaling molecules, like eicosanoids. These pathways may also contribute to the control of cellular fatty acids impacting specific nuclear receptors, e.g., PPARa [14,53,54]. Of the various pathways known to affect fatty acid structure, physiological control of fatty acid elongation remains poorly defined. The predominant pathway for fatty acid elongation occurs in the endoplasmic reticulum and uses malonyl-CoA and fatty acyl- CoA as substrates for C2 additions to fatty acids. Elongases are condensing enzymes that interact with 3-keto acyl-CoA reductase, a dehydratase and trans 2,3-enoyl CoA 39 reductase to elongate fatty acids [24,115,138]. The rate of fatty acid elongation is determined by the activity of the elongase (condensing enzyme) and not the reductases or the dehydratase. Six distinct fatty acid elongase subtypes (Elovll through Elovl6) are present in the mouse, rat and human genomes. Elovll (Sscl) and Elovl6 (LCE, FACE, rE102) elongate saturated and monounsaturated fatty acids. Disorders of sphingolipid- metabolism, like the Quaking and J impy phenotypes [114] have been associated with impaired Elovll activity. Elovl6 is induced in transgenic mice over expressing SREBP-1 [117,134,139]. Elov12 (Ssc2) substrates include, C2o.22 PUFA, while Elov15 (FAEl, Relol, Helol) may utilize a broad substrate array, C1622 [117,119]. Elov12 and Elov15 likely play a role in endogenous PUF A synthesis; i.e., the conversion of essential fatty acid precursors, linoleic acid (18:2n6) and a-linolenic acid (18:3n3) to arachidonic acid (20:4n6) and docosahexaenoic acid (22:6n3) [139]. Elovl3 (Cig30, E103) and Elovl4 (E104) are expressed in the skin [140] and retina [141], respectively. Both Elovl3 and Elovl4 elongate a broad array of fatty acids 5C26. Elovl3 is induced in brown adipose tissue following exposure of animals to the cold [142]. Stargardt-like macular dystrophy and autosomal dominant macular dystrophy is associated with defective Elovl4 expression [120]. Many studies have examined the regulation of mammalian elongases at the level of enzyme activity [25,116,117,119,127,134,138,143-156]. Overlapping substrate/product profiles for these enzymes has made it difficult to ascertain which enzyme subtypes are regulated under specific physiological conditions. More recent studies have examined nutritional and tissue-specific regulation of elongase gene expression [116,117,134,140,153,155]. We are unaware of any study that has examined 40 the expression of all elongase subtypes in specific tissues. We have cloned 5 of the 7 fatty acid elongases known to be expressed in mammals and examined their expression in rat liver. Elovl4 was not examined because of its retina-specific expression. Elovl7 was not studies because it is poorly expressed in liver (unpublished observation). Our studies included an analysis of the tissue-specific expression of elongases as well as the effect of starvation, fish oil and PPARa agonist feeding on hepatic elongase expression and elongase enzymatic activity. Developmental regulation of hepatic elongase expression and activity is also included. Of the 4 fatty acid elongases expressed in rat liver, Elov15 emerges as the predominant elongase expressed, based on mRNA abundance. Elov15 may account for much of the developmental and nutritional regulation of rat hepatic fatty acid elongation. 2.2. Methods and Materials Cloning of mouse and rat fatty acid elongases and desaturases. cDNAs for fatty acid elongases and desaturases were cloned by reverse transcriptase-polymerase chain reaction (RT-PCR) methods. Primers corresponding to the open reading frame of each elongase and desaturase (Table 2.1) were designed based on sequences obtained from Genebank and the location of the open reading frame as determined using DNA-Star. Mouse liver was used as template to clone all desaturases and elongases, except Elov15; Elov15 was cloned fi'om rat liver. The RT-PCR products were inserted into TOPO- cloning-plasmids (Invitrogen, Carlsbad, CA) and screened by blue-white selection. Positive clones were selected and sequenced at the Genomic Technology Support Facility at Michigan State University. Sequences were verified by alignment to sequences in 41 Genebank (Blast, http://www.ncbi.nlm.nih.gov/blast/ blZseq/bl2.html). Rat and mouse elongases are 285% homologous at the protein sequence level. Animals. All procedures for the use and care of animals for laboratory research have been approved by the All University Committee for Animal Use and Care at Michigan State University. Rats are maintained on Harlan-Teklad laboratory chow (#8640) and water ad lib. Tissues for the analysis of elongase expression were derived fiom 60 day old Spraque-Dawley male rats maintained on a chow diet ad lib. Nutritional regulation of elongases used a meal-feeding protocol [53,157,158]. Briefly, rats were acclimated to meal-feeding Of a high carbohydrate (glucose) diet (HiCHO) [ICN Biochemicals, Aurora, OH] supplemented with olive (Pompiean, Baltimore, MD) at 10% w/w for 7 days. The meal began at 8 AM and ended at noon. After the acclimation period, rats were either maintained on the olive oil diet or switched to a HiCHO diet supplemented with fish oil (Dyets, Inc., Bethlehem, PA) at 10% w/w or a HiCHO diet supplemented with WY14,643 at 0.1% w/w [158]. Animals were maintained on the olive oil, fish oil or WY14, 643 diets for 7 days. Fasted rats were meal fed the HiCHO-olive oil diet 7 days, but were fasted for 24 hrs before euthanasia. Two hours after completion of the final meal animals were euthanized for tissue and blood collection. The developmental studies used timed pregnant female Spraque-Dawley rats from Charles River Laboratories (Kalamazoo, MI). Fetal livers were obtained at 18-19 days post coitum and pooled for analysis. Male suckling pups (1-20 days of age) and weaned male animals (30 days of age) were used as a source of liver. Rats were weaned onto Harlan-Teklad laboratory chow at 21 days postpartum. 42 Primary hepatocytes. Rat primary hepatocytes fi'om Teklad chow-fed (ad lib) male Spraque-Dawley rats were prepared and treated with fatty acids or WY14,643 as previously described [52,136]. In vitro fatty acid elongation assay. Rat liver microsomes were isolated by differential centrifugation [159]. Elongation reactions were carried out with modifications to the procedure described by Moon et al [117]. Briefly, reaction mixtures contained 50 pg microsomal proteins in a total reaction volume of 100 pl. The elongation assay used fatty acyl CoA as substrates. The reaction constituents are: 50 mM potassium phosphate buffer- pH 6.5, 5 pM rotenone (Sigma), 40 pM fatty acyl CoA (Avanti Polar Lipids, Alibaster, Al and Sigma, St. Louis, MO), 60 pM malonyl CoA (Sigma) [6.5 dpm/pmol] [2-‘4C]-malonyl CoA (Perkin Elmer), 1 mM NADPH (Sigma) 20 pM BSA (fatty acid free). Reactions (at 37°C) were initiated with the addition of NADPH. Reactions were terminated after 20 minutes with the addition of 100 pl 5 N KOH + 10% methanol; lipids were saponified for 1 hr at 65°C. The saponification reaction was acidified with 100 pl 5 N HCL; 100 pl ethanol was added to aid hexane extraction of fatty acids. Elongated fatty acids were collected by 2 independent extractions with hexane (800 pl). Hexane extracts were pooled and l4C-radioactivity was quantified by B -scintillation counting. Restrlts are expressed as Elongase Activity Units, mnoles l4C-malonyl CoA incorporated/mg protein/2O mins. Formation of reaction products was dependent on the presence of NADPH and the fatty acid CoA. Fatty acid elongation products were verified by reverse- phase-HPLC chromatography using a flow-through B—scintillation counter [136]. RNA extraction, northern analysis and RT -PCR. RNA was extracted and separated in denaturing (formaldehyde) agarose gels, transferred to nitrocellulose and hybridized with 43 32P-cDNAs. The cDNAs for the various transcripts has been described previously [52,136,159]. Hybridization was visualized by phosphoimager analysis (Molecular Dynamics, Sunnyvale, CA). To ensure an accurate assessment of all elongases, reverse transcriptase-polymerase chain reaction (RT-PCR) was used; transcript-specific primers are described in Table 2.1. Immunoblotting. Hepatic nuclear and rrricrosomal proteins were separated by SDS- PAGE and transferred to nitrocellulose. SREBP-1, SREBP-2 and CYP4A were detected as previously described [159]. PPARa was measured using antibodies obtained from Santa Cruz Biotechnology. Anti-goat antibodies were also obtained from Santa Cruz. The detection system employed the Super Signal West Pico chemiluminescence kit (Pierce). Quantitation of hepatic and plasma mead acid (20:3,n-9) levels. Total lipid was extracted from liver or plasma in chloroform:methanol (2:1) plus 1 mM butylated hydroxytoluene [136]. 7-Nonadecenoic acid (19:1) was added as a recovery standard at the time of extraction. Protein (Bio-Rad) was measured in extracts after the initial homogenization step. Total lipids were saponified, fractionated and quantified by reverse phase HPLC (RP-HPLC) using a YMC J-Sphere (ODS-H80) column and a sigrnoidal gradient starting at 86.5% acetonitrile + acetic acid (0.1%) and ending at 100 % acetonitrile + acetic acid (0.1%) over 50 mins with a flow rate of 1.0 ml/min using a Waters 600 controller. Fatty acids were detected using both UV absorbance at 192 nm (Waters model 2487) and evaporative light scatter (Waters model 2420). Fatty acid composition and structures were confirmed at the MSU Mass Spectrometry facility by GC/MS (wwwbchmsuedu/facilities/massspec/index.htnrl). Fatty acid standards for RP- 44 HPLC were obtained from Nu-Chek Prep (Elysian, MN). Mead acid (20:3,n-9) was obtained from Sigma-Aldrich (St. Louis, MO). Statistical Analysis. Statistical analysis involved Student’s t-test; p values were calculated using Microsoft-excel t-test for 2 samples with unequal variance. 45 Table 2.1 Primers used to clone Elovls and desaturases from mouse or rat liver. Enzyme Accession No. Primer Sequence Elovll BC006735 Sense:ATGGAGGCTGTTGTCAACTTG Antisense: TCAGTTGGCCTTGACCTTGGT Elov12 NM_019423 Sense:ATGGGCGGCCGCATGGAGCAGCTGAAGGCCTTT Antisense: TTATTGAGCCTTCTTGTCCGT Elovl3 AF 054504 Sense: ATGGACACATCCATGAATTTC Antisense: TCATTGGCTCTTGGATGCAAC Elov15 NM_1343 82 Sense: ATGGAACATTTCGATGCGTCA Antisense: TCAATCCTTCCGCTGCTTCCT Elovl6 AY053453 Sense: ATGAACATGTCAGTGTTGACT Antisense: TCTAGACTACTCAGCCTTCGTGGCTTTCTT ASD NM_146094 Sense: ATGGCTCCCGACCCGGTGCCG Antisense: CTATTGGTGAAGGTAAGCGTC A6D BC057189 Sense: AGTCGACATGGGGAAGGGAGGTAACCAG Antisense: TCATTTATGGAGGTAAGCATC A9D NM_009127 Sense: ATGCCGGCCCACATGCTCCAA Antisense: TCAGCTACTCTTGTGACTCCC 46 2.3. Results 2.3.1. Tissue-specific expression of rat fatty acid elongases. Northern and RT-PCR approaches were used to determine the profile of fatty acid elongase expression in several rat tissues (Fig. 2.1). Elovll was well expressed in lung, brain, kidney and heart, but is barely detectable in liver, brown adipose tissue and skin. Because northern analysis of hepatic Elov12 indicates this transcript is expressed at low levels (see Fig. 2.2), a RT-PCR approach was used to examine its expression in various rat tissues. Elov12 is expressed in liver, lung, brain and kidney; Elov12 expression was not detected other tissues. Elovl3 was only detected in skin. Elov15 expression was readily detected in all tissues examined. Elovl6 mRNA is also expressed at low levels in rat liver (Fig. 2.2). RT-PCR was used to show that Elovl6 is expressed in all tissues; its highest expression is in brain. The tissue distribution of Elovll [155], Elov12 [155], Elovl3 [140,155], Elov15 [119] and Elovl6 [117,119] is similar to previous reports. Rat liver expresses 4 distinct fatty acid elongases, Elovll, Elov12, Elov15 and Elovl6. The relative abundance of the elongase mRN As in livers of adult male rats fed Teklad chow diets ad lib is: Elov15 > Elovll = Elov12 = Elovl6 (Fig. 2.1 and 2.2). 2.3.2. Dietary Effects on Hepatic Elongase and Desaturase Gene Expression. To examine the effects of diet on hepatic elongase expression, adult male rats were meal fed high carbohydrate diets supplemented with olive oil (10% w/w), fish oil (10%w/w) or the PPARa agonist, WY14,643 (0.1% w/w) for 7 days. Because elongases 47 Elongase Liver Lung Skin Heart Method 1 NB 2 Retina Specific Out Figure 2. 1. Tissue specific expression of rat fatty acid elongases. RNA was extracted from rat liver, lung, brain, brown adipose tissue (BAT), kidney, skin and heart. These tissues were obtained from a 60 day old male rat maintained on a Teklad-Harlan chow. diet, ad lib. Elovll, Elovl3, and Elov15 mRNAs were detected by northern analysis using cDNAs cloned as described 1n Materials and Methods. Elov12 and Elovl6 were detected by RT—PCR (35 cycles). 48 Figure 2. 2. Effect of diet on rat hepatic elongase and desaturase expression. Male Spraque-Dawley rats (50 days of age) were meal-fed a high carbohydrate diet (HiCHO) supplemented with olive (10% w/w) [Olive], fish oil (10% w/w) [Fish] or olive plus WY14, 643 (0.1% w/w) [WY14, 643] for 7 days. Fasted animals were meal fed the HiCHO-olive oil diet as described above, but were fasted for 24 hrs prior to euthanasia. A: Northern blot of elongase and desaturase mRNAs. A rat liver standard (S) derived fiom a 90 day old rat fed Teklad chow (82403 was included in all northern blots for Elovll, Elov12, Elov15, Elovl6, A5, A6 and A desaturase mRN A measurement. Single transcripts were detected for each mRNA, except Elovl6. Two transcripts have been reported for Elovl6 [117]. In all cases, the size of the mRNA corresponded with the reported size of the transcript. A representative etlridium bromide stain of 18 S and 28 S mRNA documents equal loading of the RNA samples. Elovl3 was not detected in any treatment (not shown). B: Fold changes in fatty acid elongase, fatty acid desaturase and fatty acid synthase (F AS) mRNA abundance following dietary challenge. Top panel: Effect of refeeding fasted animals on mRNA expression. Results are expressed as the Mean : SD; N=3. *p50.05; fasted vs fed. Middle panel: Effect of fish oil feeding on mRNA expression. Results are expressed the Mean i SD,n=6/ group. *p30.05, olive vs fish. Bottom panel: Effect of WY14, 643 feeding on mRNA expression. Results are expressed as the Mean : SD,n=6/group. *p50.05; olive vs olive + WY14, 643. 49 2.2A S Fasted Olive EIOVI-1 Elovl-S Elovl-6 2.213. _ Fasted V8 Fed o 2.38 _A O l O .-‘ 01-0-0 Fold Change in mRNA Abundance o OM50) ElovI-1 EIovl-Z ElovI-S EIovI-G (150 660 d90 FAS 50 act in concert with desaturases for PUFA synthesis, we examined the hepatic expression of 4 elongases(Elov11, -2, -5 and -6), 3 desaturases (A5, A6 and A9) and fatty acid synthase (FAS). Our goal was to determine whether there is any evidence for coordinate regulation of pathways involved in fatty acid elongation, fatty acid desaturation and de novo lipogenesis (DNL). Figure 2.2 (A and B) illustrates the effect of these treatments on mRN A abundance of the various transcripts. Fasting and refeeding: Fasting suppresses the expression of enzymes involved in DNL, fatty acid desaturation [46,137,160] and at least one elongase, Elovl6 [119,134]. To determine if other hepatic fatty acid elongases are regulated by fasting, rats meal-fed a If high carbohydrate diet supplemented with olive oil (10 % w/w) were fasted for 24 hrs and refed the same diet for 4 hrs (Figs. 2.2). mRNAs encoding Elovll and Elov15 were induced marginally (1.5- and 1.9-fold) by refeeding fasted rats. Only Elovl6 mRNA displayed a robust (14-fold) induction in response to refeeding fasted rats. In contrast, all of the desaturase (A5, A6, A9) mRNAs, as well as mRNA FAS, were induced 5.2-, 2.6-, 18- and 6.5-fold, respectively. Unlike the desaturases, the elongases do not display a uniform response to fasting and refeeding. Fish oil Feeding. Feeding rats diets enriched in N3-PUFA, i.e., fish oil, inhibits DNL and the expression of all fatty acid desaturases [2,5,160]. While fish oil suppressed FAS and each desaturase mRN A by 250%, fish oil feeding had no significant effect on Elovll and Elov12 mRNA abundance. Only mRNAElovls and mRNAElovm were significantly (_>_50%) suppressed by fish oil (Fig. 2.2). Fish oil feeding does not uniformly suppress expression of all hepatic fatty acid elongases. 51 WY14,643 Feeding. All hepatic fatty acid desaturases are induced by PPARa agonists, like WY14,643 [134,160,161]. To determine if the elongases are regulated by WY14,643, rats were fed WY14,643 (0.1% w/w) in the high carbohydrate-olive oil diet for 7 days (Fig. 2.2). mRNAs encoding all of the desaturases and 3 of the elongases were induced by WY14, 643. Elovll and Elov15 were induced 2- and 2.5-fold; Elovl6 was induced nearly 4-fold. Elov12 mRNA was not affected by WY14,643 feeding. A5, A6 and A9 desaturase mRNAs were induced 4.1-, 2.3- and 4.8-fold respectively, while mRNApAs was not significantly affected by WY14,643 feeding. The effect of WY14,643 on both elongase and desaturase expression suggest PPARa agonist may induce long chain ‘ polyunsaturated synthesis. Dietary Effects on Hepatic Elongase Activity. We next determined if changes in hepatic elongase mRN A abundance is consistent with hepatic fatty acid elongase activity. Elongase assays used 7 substrates, i.e., 16:0-CoA, 18:0-CoA, 18:1,n-9-COA, 20:0-COA, 20:4,n-6-COA, 22:0-C0A and 24:0-COA, in separate assays (Fig. 2.3). Based on reports by others, these fatty acyl-CoAs likely serve as substrates for l or more of the hepatic elongases. Specifically, 16:0-C0A is a substrate for Elovll and Elovl6, but not Elov12 [117,134,155]. 18:1-CoA is not a substrate for Elovl6 [134], but is a substrate for other elongases." 20:4,n-6 is a substrate for Elov12 and Elov15, but not Elovl6 [117,119,134]. 20:0-C0A, 22:0-CoA and 24:0-COA are substrates for Elov11[155]. When compared to the olive oil-fed rats, hepatic microsomes isolated from fasted rats had a significantly lower (~50%) elongase activity when using 16:0-COA and 20:4- CoA as substrate. Elongation of other fatty acyl CoAs was not significantly affected by fasting and refeeding. This effect correlates with the suppression of Elovll , Elov15 and 52 -L “J I [1:1 Fast I Olive 1:: Fish 1:: WY14,643 in Chow] A CD CD I l r--—-4 A I ’2222222332: 0 Elongase Activity, Units N O) :zzzzz I 16:0-00A 18:0-00A 18:1-00A 20:0-CoA 20:4-CoA 22:0-COA 24:0-CoA Elongase Substrates C) Figure 2. 3. Effect of diet on hepatic fatty acid elongase activity. In vitro assays for elongase activity used microsomes isolated from livers of rats fasted or fed a high carbohydrate diet containing olive oil, fish oil or WY14, 643 as described in the Methods. Separate reactions were run using 16:0-CoA, 18:0-CoA, 18:1n9-COA, 20:0-COA, 20:4n6- CoA, 22: O-CoA and 24: 0- CoA as substrate. Results are expressed as Elongase Activity, Units (nmoles l4C-Malonyl CoA incorporated/mg protein), Mean + SD ,n=3/group. *p< 0. 05, Olive oil fed vs Fasted, Fish oil fed or WY14, 643 fed. 53 Elovl6 mRNA in starved animals (Fig. 2.2). A comparison of the elongase activity in microsomes isolated from olive and fish oil fed animals indicates that fish oil feeding suppressed (~60%) the elongation of both 16:0-C0A and 20:4-C0A. Fish oil suppressed only Elovl6 and Elov15 mRNA, respectively (Fig. 2.2). Feeding rats WY14,643 increased elongation of all saturated and monounsaturated fatty acyl-CoA substrates, except 16:0- CoA, 2- to 3-fold. Increased elongase activity following WY14,643 feeding correlates with increased hepatic abundance of mRNAs encoding Elovll , Elov15 and Elovl6 (Fig. 2.2). Increased Elovll expression in livers of WY14,643-fed rats is likely involved in the increased elongation activity toward saturated and monounsaturated fatty acids. Effect of fish oil and WY14,643 on hepatic and plasma fatty acid composition. We next determined if changes in elongase gene expression and elongase activity had any effect on hepatic or plasma lipid composition (Fig. 2.4). For this analysis, we focused on mead acid (20:3,n-9) levels, because: 1) 20:3,n-9 is not a dietary fat; its presence in cells is due to elongation and desaturation of 18:1,n-7 or 18:1,n-9 [162]; 2) dietary n-3 and n-6 PUFA suppress tissue levels of 20:3,n-9, by competing with 18:1,n-9 for A6 desaturase [163] and suppressing the expression of mRNAs encoding A5 and A‘5 desaturases. The contribution of elongases or WY14, 643 to 20:3,n-9 synthesis is unknown. Fish oil and WY14,643 affect tissue and plasma levels of 20:3,n-9 and 20:4,n-6 (Fig. 2.4). In olive oil-fed rats, the mole % of 20:3,n-9 and 20:4,n-6 in the plasma is 0.7 and 6.4. In liver, the mole % of 20:3,n-9 and 20:4,n-6 is 1.8 and 14.2. The ratio of 20:3,n- 9/20:4,n-6 in plasma and liver is 0.1 and 0.13. These values are in line with essential fatty acid sufficiency [164]. Rats fed the fish oil diet have a > 95% reduction in plasma and hepatic 20:3,n-9. In contrast, olive oil-WY14,643 fed animal have 4- and 5-fold increased 54 Plasma 10 - IDZO:3,n-9 I20:4,n-6| Liver 20" Fatty Acid, Mole °/o 15' 10' 75 Olive .Flshép Figure 2.4. Effect of diet on plasma and liver mead acid levels. Total lipids extracted from rat liver and plasma of animals fed olive oil, fish oil or WY14,643 were saponified, separated by RP-HPLC and quantified (Methods and Materials). The. identity of all fatty acids was confirmed by gas chromatography-mass spectrometry. Levels of mead acid (20:3,n-9) and arachidonic acid (20:4,n-6) were quantified and the results are presented as Mole % of the total saponified lipid fraction, Mean + SD, n= 3. The mole % of 20: 3 ,n-9 in olive oil vs fish oil- fed animals and m olive oil and olive oil + WY14, 643- fed animals was significantly different, *p<0. 01. 55 in 20:3,n-9 levels in plasma and liver, respectively. The 20:3,n-9/20:4,n-6 ratio in these animals is 0.8 and 0.6 in plasma and liver, a value found in essential fatty acid deficiency (EFAD). The elevation in 20:3,n-9 levels can be correlated to increased elongation of 18:1-CoA (Fig. 2.3) and elevated Elovll, Elov15, A5 and A6 desaturase mRNA abundance (Fig. 2.2) plus the increased dietary intake of 18:1,n-9 in the olive oil diet. 2.3.3. Developmental regulation of hepatic fatty acid elongase and desaturase gene expression. SREBP-lo, a key transcription factor for controlling hepatic lipid synthesis [78], has been implicated in the regulation of at least one fatty acid elongases (Elovl6) [117,134,139]. In an effort to determine if changes in SREBP-lo nuclear content correlate with elongase expression, we have used the model of early postnatal development. In this model, SREBP-1 is present in fetal hepatic nuclei (Fig. 2.5, upper panel). Shortly after birth, however, nuclear SREBP-1 levels fall and do not rise again until the animals are weaned at 21 days postpartum [159]. We examined the expression of hepatic elongases and desaturases in rat livers derived from fetal animals (18 days post coitum), at 10 days postpartum (suckling) and 30 days postpartum (weaned). Fatty acid synthase (F AS) was included for comparison (Fig. 2.5 lower panel). Expression of all 4 elongases (Elovll, -2, -5 and -6), all three desaturases (A5, A6, and A9) and FAS were detected in fetal liver. Elovll decreased to adult values after birth. Elov12 remains unchanged from 3 days prepartum to 30 days postpartum. Fetal Elovl 5 expression is <10% of the 30 day value and increased 5-fold by 10 days and 10-fold by 30 days postpartum. Elovl6 mRNA fell after birth to undetectable levels, but increased to adult values after weaning. The developmental regulation of Elovl6, A9 desaturase and F AS 56 A Immunoblot Days Postpartum Fetal 1 7 15 30 Nuclear SREBP-1 . . Nuclear SREBP-2 "' n. *1 mRNA Abundance [3 Fetal .10 Days Old I] 30 Days Old * * Relative mRNA Abundance W O -l N 00 h 01 O) Elovl-1 Elovl-2 Elovl-5 Elovl-6 d5 D dSD d9 D FAS Figure 2.- 5. DevelOpmental regulation of rat hepatic SREBP-l and -2 nuclear content and mRNA abundance of fatty acid elongase, fatty acid desaturase and fatty acid synthase. A. Immunoblot: Nuclear proteins isolated fiom livers at 18 days postcoitum (Fetal) , 1, 7, 15 and 30 days postpartum were prepared, separated by SDS- PAGE, transferred to nitrocellulose and probed with antibodies for SREBP-1 and SREBP-2 [159]. The inclusion of SREBP-2 shows that hepatic SREBP—l and SREBP-2 nuclear content is regulated differently during development. B_. mRN A Abundance: Rat liver mRNA from 18 days postcoitum (Fetal), 10 and 30 days postpartum was used to quantify levels of fatty acid elongases (Elovll, Elov12, Elov15, Elovl6), fatty acid desaturases (ASD, A6D and A9D) and fatty acid synthase (F AS). The results are represented as “Relative RNA Abundance” and are normalized to the level of expression in a 90 day old male rat. Mean : SD,number of animals (N)/group = 3. *p50.05, Fetal vs 10 or 30 days. 57 parallels changes in SREBP-1 nuclear content during early development. However, at no time in this analysis was Elovl6 mRN A found to be an abundant transcript. In contrast, mRNAs encoding Elov15, A5 and A6 desaturase are very low in fetal liver and increase after parturition. Developmental changes in Elov15, A5 and A6 desaturase do not parallel changes in SREBP-l nuclear content. At parturition, animals ingest a high fat milk diet where 65% of the calories are fat. The ingestion of the high fat milk diet is associated with the activation of PPARa- regulated transcripts [165,166]. Nuclear PPARa content changes little in livers of fetal, neonatal or weaned animals (Fig. 2.6). The PPARa-regulated transcript, CYP4A, is 5 induced (~10-fold) within 1 day of birth; both mRNAcyp4A and microsomal CYP4A protein increase >10-fold. The induction of mRNAcyp4A parallels the induction of mRNAElovls afier birth. Both A5 and A6 desaturase mRNAs rapidly increase after birth (not shown). This observation, coupled with the fact that mRNAEIovls is induced by WY14,643 (Fig. 2.2), suggest that PPARa and the high fat milk diets contribute to the postnatal increase in mRNA Elov15- Developmental regulation of hepatic fatty acid elongase activity. Fatty acid ' elongation activity was examined in microsomes isolated from fetal liver and livers of 10 and 30 days old male rats (Fig. 2.7). Fatty acid elongation activity for all substrates is low in fetal liver and increased 5- to 20-fold by 10 days postpartum. However, elongation of €22-24 saturated fatty acids is ~2 to 3-fold higher than €16-13 saturated fatty acids. Elongation activity for each substrate was induced significantly by 10 days postpartum. The level of elongase activity at 10 days postpartum was at or near adult (90 days) values. 58 Days Postpartum Fetal 3 7 10 15 18 21 90 IB: PPAR-alpha NB: CYP4A IB: CYP4A NB: Elovl-5 Figure 2. 6. Rapid induction of hepatic Elov15 and CYP4A mRNA following parturition. NB___ Elov15: mRNA was extracted from livers of fetal rats and from male rats 3, 7, 10, 15,18, 21 and 90 days old. A northern blot illustrating the induction of 1.310145 18 shown. IB CYP4A and IB PPARa: nuclear (PPARa) or microsomal (CYP4A) proteins were isolated from livers at 18 days postcoitum (fetal) and 3, 7, .10, 15, 18,21 and 30 days postpartum. Proteins were separated by SDS}PAGE, transferred-to nitrocellulose and probed with antibodies for PPARa or'CYP4A, Imm'uriob'ldt (1B) [159]. TotalRNA was extracted from rat livers at 18 days post coitum (fetal) and 1, 7, 15, 21 and 30 days postpartum (3 animals/group); S, RNA from 90 day old rat liver. RNA was separated and probed for CYP4A [75]. 59 of the substrates used, 2-3 times more 20:4-CoA was converted to its elongation product (22:4,n-6) than any other fatty acyl CoA tested. The low elongase activity in fetal livers correlates with the low level of expression of all hepatic elongases. Note that Elovll , Elov12 and Elovl6 are expressed at low levels in both fetal and adult livers (Figs. 2.1, 2.2 & 2.5). The postnatal increase in hepatic elongase activity correlates with only one elongase transcript, i.e., Elov15. I mRNAElovls was induced (5-fold) by 10 days postpartum (Fig. 2.5 and 2.6). Since no other elongase was significantly induced by 10 days postpartum, our studies suggest that Elov15 is the likely elongase responsible for a major fraction hepatic fatty acid elongation. If so, then Elov15 is capable of elongating a broad range of fatty acid substrates, including saturated (16:0, 18:0, 20:0, 22:0 & 24:0), monounsaturated (18:1n9) and polyunsaturated (20:4n6) fatty acids (Fig. 2.7). Elov15 mRNA is expressed at higher levels than all other elongases. If changes in elongase mRNA parallel changes in elongase protein and elongase activity, then much of the increase in hepatic fatty acid elongase activity that occurs during postnatal development can be attributed to Elov15. 2.4. Discussion Liver (Fig. 2.7), primary hepatocytes [53], Hek293 [54] and FTC-2b hepatoma cells display robust elongation activity toward C20 PUFA. C2022 PUFA fatty acids ‘ regulate the activity of fatty acid-regulated nuclear receptors, like PPARa and LXRa [53,54]. As such, changes in C20 PUFA elongation might impact PPARa and LXRa action. In an effort to determine which elongase might be responsible for C20 elongation, we examined the expression and activity of rat liver fatty acid elongases. These studies 60 .2 c . 3 4 ' Days Postpartum E Baum-mum .2 3 - ‘6 < 2 i 3’ 1 2 m 0 . 16:0-00A 18:0-00A 18:1-GOA 20:0-CoA 20:4—CoA 22:0-00A 24:0-CoA Elongase Substrates Figure 2. 7. Developmental regulation of hepatic fatty acid elongase activity. In vitro assays for elongase activity used microsomes isolated from livers of fetal rats (18 days post coitium), 10, 30 and 90 days postpartum. Separate reactions were run using 16:0-CoA, 18:0-CoA, 18:1,n-9-CoA, 20:0-CoA, 20:4,n-6-CoA, 22:0-COA and 24:0-CoA as substrate. Results are expressed as Elongase Activity, Units (”C-Malonyl CoA nmoles incorporated/mg protein), Mean j; SD,n=3/ group. Elongase activity in fetal liver was significantly lower than postpartum liver for all substrates examined,i:p5 0.05.552, “.;t a .. 61 have revealed new information on the regulation of elongase expression and which elongase likely contributes to C20 PUFA metabolism in liver. Of the 4 fatty acid elongases (Elovll, Elov12, Elov15 and Elov16) expressed in liver (Figs. 2.1 and 2.2), only Elovll, Elov12 and Elov15 use C20 fatty acid substrates [117,119,155]. The mRN As encoding Elovll and Elov12 are expressed at very low levels relative to mRNAErovls in adult rat liver. Moreover, Elovll and Elov12 mRNAs do not increase during postnatal development, a time in which the liver substantially increases its capacity for C20 PUFA elongation (Fig. 2.7). Elov15 is the only fatty acid elongase readily detected in primary hepatocytes. Based on this observation, we suggest that Elov15 is responsible for elongation of C20 PUFA to C22 PUFA in primary hepatocytes. Finding that Elov15 mRNA is expressed in broad array of tissues (Fig. 2.1) [119], regulated during postnatal development (Fig. 2.5), by dietary fat and PPARa agonist (WY14,643) (Fig. 2.2), suggest changes in Elov15 expression and activity may be important for the maintenance of cellular long chain fatty acids. Whether changes in Elov15 expression and activity affect fatty acid-regulated nuclear receptor function is an issue addressed in a later chapter. It is unclear if changes in elongase activity during postnatal development are due solely to changes in elongase expression. Elongases are condensing enzymes that interact with 3-keto acyl-CoA reductase, a dehydratase and trans 2,3-enoyl CoA reductase to elongate fatty acids [24,138,153]. The rate of fatty acid elongation is generally viewed as being determined by the activity of the elongase (condensing enzyme) and not the reductases or dehydratase. However, the low elongase activity in fetal liver may be due to deficient expression of the reductases and dehydratase. Additional studies will be required to examine the expression of these enzymes during postnatal development. These developmental and nutritional studies have implicated 2 transcription factors in control of elongase expression, i.e., PPARa and SREBP-1. During postnatal development two distinct patterns of fatty acid elongase and desaturase expression are evident. These two patterns of expression can be linked to changes in SREBP-l nuclear content or the activation of PPARa. Both SREBP-l and PPARa play major roles in whole body and hepatic lipid metabolism [51,78]. Changes in SREBP-1 nuclear content closely parallel changes in FAS, A9 desaturase and Elovl6 expression (Fig. 2.5). SREBP- 1 is expressed in fetal livers at levels near adult values. During the suckling period, however, SREBP-1 is absent from the nucleus. FAS, A9 desaturase and Elovl6 increase after weaning when SREBP-1 nuclear content increases. SREBP-1 nuclear abundance has been linked, directly or indirectly, to the transcriptional control of each of these genes [78,139,167]. The regulation of SREBP-1 nuclear content during postnatal development may be mediated by changes in proteolytic conversion of the SREBP-1c precursort(~125 kd) to the mature form (~65 kd) and its transport to the nucleus [159]. Ingestion of the high fat milk diet (~65% calories as fat) begins at parturition. The ingestion of the high fat milk diet, coupled with low blood insulin levels [168] likely contributes to this regulatory scheme. The expression of the other elongases, particularly Elov15, does not parallel changes in SREBP-1 nuclear content during postnatal development (Fig. 2.5). Treatment of primary hepatocytes with T1317, an LXR agonist, significantly elevates SREBP-1 nuclear content as well as several SREBP-l regulated transcripts [52]. T1317 only 63 modestly induced mRNAglovls (<3 0%) in primary hepatocytes (not shown). Unlike Elovl6 [117,134], Elov15 does not appear to be a target for either SREBP-1 or LXRa. Despite the finding that the postnatal induction of Elov15, A5 and A6'desaturase closely parallels the induction of PPARa-regulated transcripts, e.g., Cyp4A (Fig. 2.6) [75 ,158], the role PPARa plays in [144] this regulatory scheme is less clear. PPARa agonist, like WY14,643 (Fig. 2.2) and fibrates induce hepatic elongase activity toward a broad range of fatty acids. Our studies show that WY14,643 induced Elovll, -5 and -6 mRNA (Fig. 2.2). Studies with primary hepatocytes, however, have indicated that while PPARa target genes, like CYP4A and AOX, are well induced in primary hepatocytes [136,158], transcripts encoding elongases, e.g., Elov15, are only marginally (<25%) induced (not shown). The lack of a significant WY14,643 effect on Elov15 (or other elongases) expression in primary hepatocytes suggest these enzymes are not a direct target of PPARa. Clearly, many factors change during postnatal development, e.g., glucocorticoids, leptin, T3 [169,170], that could impact hepatic elongase expression. These and other factors were evaluated for their contribution to elOngase expression in the next chapter. ' Our studies reveal two regulatory patterns for elongase and desaturase expression during postnatal development. These different patterns of expression likely have physiological significance. Blood of pregnant female rats fed a Teklad chow diet contains levels of 20:4,n-6 and 22:6,n-3 at 10 and 5 mole %, respectively (not shown). In contrast, rat milk is deficient in 20:4,n-6 and 22:6,n-3, i.e., 0.5 and 0.1 mole % respectively. Analysis of hepatic fatty acid profiles in fetal, 10 and 30 day old liver shows that the mole % of 20:4,n-6 and 22:6,n-3 remains essentially unchanged, at 15-20 mole% (not 64 shown). Since neonates grow very rapidly, the only mechanism available to sustain the constant level of hepatic 20:4,n-6 and 22:6,n-3 in the absence of an extrahepatic source of €20-22 PUFA is to induce PUFA synthesis. The capacity of the neonatal liver to synthesize 20:4,n-6 and 22:6,n-3 from dietary precursors (18:2,n-6 and 18:3,-n3) is achieved, at least in part, by inducing mRNAs encoding A5 and A6 desaturase and Elov15 (Fig. 2.6 and 2.7). Fetal liver has high blood levels of 20:4,n-6 and 22:6,n-3 and low elongase activity, and low levels of mRNAs encoding Elov15, A5 and A6 desaturase. Adult animals fed fish oil (a rich source of 20:5,n-3, 22:5,n-3 and 22:6,n-3), have low 20:4,n-6 and 20:5,n-3 elongation activity (Fig. 2.5) and suppressed expression of mRNAs encoding Elov15, A5 and A6 desaturase (Fig. 2.2). Thus, changes in Elov15, A5 and A6 desaturase expression respond to changes in dietary PUFA composition and tissue requirements for C2o.22 PUFA. Elov12 has also been implicated in PUFA synthesis [118,127]. Yet mRNAglom remains unresponsive during postnatal development, following fasting/refeeding and fish oil treatment. Based on these findings, Elov15, and not Elov12, appears to play the major role, along with A5 and A6 desaturase, in the adaptiVe response of PUFA synthesis following changes in dietary lipid composition. This response is mediated, at least in part, through the regulation of the cellular abundance of the mRNAs encoding these enzymes. Additional studies will be needed to further define the roles Elov12 and Elov15 play in the control of hepatic PUFA synthesis. In this regard, raising rats on n3—PUF A deficient diets leads to changes in blood and tissue lipid profiles as well as diminished cognitive function [171]. Whether similar effects occur in null mutations of specific elongases or desaturases remains to be determined. 65 Feeding rats fish oil suppresses lipogenic gene expression [52,157,172] and mRNAglovls (250%) (Fig. 2.2). Primary hepatocytes treated with 20:5,n-3 showed ~50% reduction in Elov15 mRNA (not shown). Many of the effects of fish [oil on hepatic gene expression can be attributed to suppression of nuclear levels of SREBP-1c or activation of PPARa [14]. Our findings indicate that neither factor induces major changes in Elov15 expression in primary hepatocytes. Hepatic L-pyruvate kinase gene transcription is suppressed by 20:5,n-3 through mechanisms that are independent of PPARa and SREBP- lc [157,158]. The target for 20:5,n-3 action on L-PK is a region that binds a carbohydrate regulatory element binding protein (ChREBP), Mlx and HNF-4 [90,173]. Elov15 mRNA is unresponsive to changes in glucose treatment of primary hepatocytes suggesting that ' CHREBP and Mlx are not involved in Elov15 expression (not shown). The role of HNF-4 in Elov15 expression is under investigation. Finally, finding that WY14,643 elevated hepatic and plasma levels of mead acid (20:3,n-9) in olive oil-fed rats was a surprise (Fig. 2.4). Mead acid is a elongation and desaturation (A5 and A6 desaturase) product of vaccenic (18:1,n-7) and oleic (18:1,n-9) ' acid [162]. The effect of dietary n3 and n6 PUFA on tissue and plasma levels of 20:3‘,n'-9 has been well described [164]. It is generally accepted that much cf the PUFA-mediated suppression of 20:3,n-9 content in cells is due to competition of PUFA for A6 desaturase (n3 > n6 > n9) [163]. The elevated tissue content of €20-22 PUFA, coupled with the decline in both elongation and desaturation capacity (Fig. 2.2-2.4), likely accounts for the low levels of 20:3,n-9 in fish oil-fed animals. Contrary to the effects of fish oil, WY14,643 significantly increased hepatic and plasma levels of 20:3,n—9. The level of 18:2,n-6 in the olive oil diet (3.3mole %) is sufficient to prevent EF AD. Ingestion of a 66 18:1n9-enriched diet, however, coupled with the induction of hepatic Elovll, Elov15 and Elovl6 plus A5 and A6 desaturases by WY14,643, is apparently sufficient to increase 20:3,n-9 synthesis and its accumulation in tissues and plasma. The consequence of elevated 20:3,n-9 production is an increase in the hepatic 20:3,n-9/20:4,n-6 to 0.62, a value consistent with essential fatty acid deficiency (EFAD) [164]. Clearly, the elevated expression of key enzymes involved in PUF A synthesis, plus ingestion of a 18:1,n-9 enriched diet, is sufficient to shift the tissue balance of 20:3,n-9 and 20:4,n-6. 67 Chapter 3 Regulation of Hepatic Fatty Acid Elongase and Desaturase Expression in Diabetes and Obesity 3.1 Introduction The liver plays a central role in whole body lipid metabolism. Fatty acids are synthesized de novo from glucose. This pathway utilizes products from glycolysis and along with the two enzymes, acetyl CoA carboxylase and fatty acid synthase, generates palmitate (16:0). Insulin, triiodothyronine (T3), glucocorticoids and glucose induce, while C20 polyunsaturated fatty acids (PUFA), glucagon and epinephrine suppress de novo lipogenesis [14,137,174]. The liver also modifies fatty acid structure through metabolic pathways that include desaturation, elongation, mono-oxidation and peroxisomal [3- oxidation (chain shortening). Such modifications occur to fatty acids generated de novo, as well as fatty acids derived from the diet. These pathways are particularly critical for the generation of end products of PUFA synthesis. Arachidonic acid (20:4,n-6) and docosahexaenoic acid (22:6,n-3) are the main C20-22 PUFA accumulating in membranes of all tissues [31]. Together, these metabolic pathways play an important role in the maintenance of membrane lipid composition and lipid storage, the generation of precursors for signaling molecules, like eicosanoids, and the control of fatty acid- regulated transcription factors [14,52-54]. 68 Of the various pathways known to affect fatty acid structure, physiological control of fatty acid elongation remains poorly defined. We recently reported on the tissue- specific, nutritional and developmental regulation of fatty acid elongases in the rat [175]. The outcome of those studies suggested that hepatic elongase expression might be controlled by at least two transcription factors, peroxisome proliferator-activated receptor (PPAR; and sterol regulatory element binding protein-1 (SREBP-1). This report extends those findings by examining SREBP-1, PPARa, liver X receptor (LXR), carbohydrate regulatory element binding protein (ChREBP) and MAX-like factor X (MLX) control of elongase and desaturase expression. Our analysis also includes in. vivo studies to determine if changes in hepatic lipid composition induced by diabetes or obesity correlated with changes in elongase and desaturase expression. Overall, these studies establish a role for specific transcriptional regulatory networks in the control of hepatic desaturase and elongase gene expression and hepatic lipid composition. 3.2 Materials and Methods Animals. All procedures for the use and care of animals for laboratory research have been approved by the All University Committee for Animal Use and Care at Michigan State University. Streptggotocin-Induced Diabetes: Male Spraque-Dawley rats (200-250 g) (Charles River Laboratories, Kalamazoo, M1) were maintained on Harlan-Teklad laboratory chow (#8640) and water ad lib. Rats were injected, intraperitoneally with streptozotocin (7.5 mg/ 100 g. BW) and 3 ml of 25% glucose [176]. Three weeks later, blood glucose was measured on animals receiving no streptozotocin (control) or streptozotocin (diabetic, blood glucose 3120 mg/dl). Blood glucose was measured in isoflurane anesthetized rats 69 using a glucose meter (Freestyle Flash, Thera Sense, Inc, Alameda, CA). Control and diabetic rats were euthanized (isoflurane anesthesia and exsanguinations) for recovery of blood (plasma) and liver. High Fat Feeding of C5 7BL/6 Mice: Male C57BL/6 mice (Jackson Laboratories, Bar Harbor Maine), 2 months of age, were fed diets containing 10% lard (D12450B) or 60% lard (D12492), Research Diets, Inc.) ad lib for 10 weeks. Four days prior to euthanasia, mice were subjected to a glucose tolerance test. Briefly, mice were injected with glucose (2 g/kg, IP). Blood was withdrawn from the tail vein prior to and after glucose treatment. Blood glucose was measured using a hand-held glucose meter (Freestyle Flash, Thera Sense, Inc, Alameda, CA). Mice were euthanized using isoflurane and exsanguinated; blood and liver was recovered. Livers were used for RNA and lipid extraction. Lean amL Obese Mice. Livers from lean and obese C57BL/6 mice were obtained from Drs. Romsos and Claycomb (Department of Food Science and Human Nutrition, MSU). Male lean C57BL/6J-Lep0b/+ and obese (C57BL/6J-LepOb/0b) mice (B6.V-Lep ob/J' , #000632, Jackson Labs) were maintained on a Harlan-Teklad laboratory chow (#8640) diet and water ad lib. Livers were used for RNA, lipid and protein extraction. Wild type and PPARa [-/-Lmice. Homozygous wild type and PPARa-null (-/-) mice on an Sv/129 genetic background [177,178]were fed either a control diet or one containing WY14643 (at 50 or 500 ppm: Bio-Serv, Piscataway, NJ) for 1 week. Mice were euthanized, livers were removed and RNA was isolated for analysis of gene expression. 2, 3, 7, 8-tetracliorodibenzo-p-dioxin (T CDD) treated mice. Female C57BL/6 mice were treated by gavage with 0.1 ml of sesame oil for a nominal dose of 0 (vehicle control)lor 30 rig/kg body weight of TCDD. Mice were sacrificed 168 h afier dosing. Livers were 70 removed and RNA was isolated for analysis of gene expression and fatty acid composition. Primary Rat Hepatocytes: Male Spraque Dawley rats (Charles River Laboratories, Kalamazoo, M1) were maintained on Harlan-Teklad laboratory chow (#8640) and water ad lib. Rat primary hepatocytes were prepared from Teklad chow-fed (ad lib) male Spraque-Dawley rats, cultured on BioCoat (type I collagen) plates (Beckon Dickinson, Belford, MA.[179]. For RNA and protein extraction, cells were plated onto 100 mm type I collagen-coated plates (BD Bioscience, Bedford, MA) at 107 cells/plate in Williams E (Gibco/Invitrogen, Carlsbad CA), 10 mM lactate, 10 nM dexamethasone (Sigma, St. Louis), and 10 % fetal bovine serum (Gibco/Invitrogen). For adenoviral infection studies, cells were plated in the same media onto 6 well type 1 collagen-coated plates at 1.5 x 106 cells/well. The ratio of culture media to cell number was maintained constant for the difierent plating conditions. For treatments, hepatocytes were incubated in medium (\Vrlliams E + 10 nM dexamethasone without or with 100 nM insan and/or 25 mM glucose. . RNA extraction, northern analysis and quantitative real time-PCR (qR T -PCR). Total RNA was extracted from primary hepatocytes or liver samples and used for template for qRT-PCR or northern analysis as previously described [175]. SpeCific primers for each gene (Table 3.1) were designed using Primer Express software (Applied Biosystems, Foster City, CA). First strand cDNA was synthesized using the SuperScript II RNase H- Reverse Transcriptase (Invitrogen Carlsbad, CA). Synthesized cDNA was mixed with 2x SYBR Green PCR Master Mix (Applied Biosystems) and various sets of gene-specific forward and reverse primers and subjected to real-time PCR quantification using the ABI 71 PRISM 7700 Sequence Detection System (Applied Biosystems). All reactions were performed in triplicate. The relative amounts of mRNAs were calculated by using the comparative CT method (User Bulletin #2, Applied Biosystems). Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. Primers used for quantitative real time PCR are listed in Table 3.1. Lipid attraction and quantitation of hepatic fatty acids composition. Total lipid was extracted from liver in chloroforrnzmethanol (2: 1) plus 1 mM butylated hydroxytoluene [175]. 7-nonadecenoic acid (19:1) was added as a recovery standard at the time of extraction. Protein (Bio-Rad, Hercules, CA) was measured in extracts after the initial homogenization step. Total lipids were saponified, fractionated and quantified by reverse phase HPLC (RP-HPLC) using a YMC J-Sphere (ODS-H80) column and a gradient starting at 77.5% acetonitrile + acetic acid (0.1%) and ending at 100 % acetonitrile + acetic acid (0.1%) over 90 mins with a flow rate of 1.0 ml/min using a Waters 600 controller. Fatty acids were detected using both UV absorbance at 192 nm (Waters model 2487) and evaporative light scatter (Waters model 2420). Fatty acid composition and structures were confirmed at the MSU Mass Spectrometry facility by GC/MS (wwwzbchmsucdu/facilities/massspec/index.html). Fatty acid standards for RP-HPLC were obtained fiom Nu-Chek Prep (Elysian, MN). Immunoblotting. Liver microsomal and nuclear extracts were prepared as described previously [159,175]. Proteins (SO-100 11g) extracted fiom microsomal or nuclear fiactions were separated electrophoretically by SDS-polyacrylamide gel electrophoresis (NuPAGE 4-12% polyacrylamide Bis-Tris, Invitrogen) and transferred to nitrocellulose 72 Table 3. 1. Primers for quantitative reverse transcriptase-polymerase chain reaction Gene Forward Reverse . Rat Elovll CCCTACCTTTGGTGGAAGAA TCCAGATGAGGTGGATGATG Elov12 TTTGGCTGTCTCATCTTCCA GGGAAACCGTTCTTCACTTC Elov15 TACCACCATGCCACTATGCT GACGTGGATGAAGCTGTTGA Elovl6 CAACGGACCTGTCAGCAA GTGGTACCAGTGCAGGAAGA ASD TGGAGAGCAACTGGTI‘TGTG GTTGAAGGCTGACTGGTGAA A6D TGTCCACAAGTTTGTCATTGG ACACGTGCAGGCTCT'ITATG A9D ACA'I‘TCAATCTCGGGAGAACA CCATGCAGTCGATGAAGAAC LXRa TCAGCATCTTCTCTGCAGACCGG TCATTAGCATCCGTGGGAACA LXRB AAGCTGGTGAGCCTGCGC CGGCAGCTTCTTGTCCTG SREBP-1c GATTGCACATTTGAAGACATGCTT GTCCCAGGAAGGCTTCCAGAGA L-PK AGGAGTC'ITCCCCTTGCTCT ACCTGTCACCACAATCACCA Cyclophilin CTTCTTGCTGGTCTTGCCATTCCT GGATGGCAAGCATGTGGTCTTTG B-Actin ACTAT‘TGGCAACGAGCGGTT TGTCAGCAATGCCTGGGTACA Mouse Elovll CCCTACCTTTGGTGGAAGAA ATCCAGATGAGGTGGATGATG Elov12 ACGCTGGTCATCCTGTTCTT GCCACAATTAAGTGGGCTTT Elov15 GGTGGCTG'ITCTTCCAGATT CCCTTCAGGTGGTCTTTCC ElOVl6 ACAATGGACCTGTCAGCAAA GTACCAGTGCAGGAAGATCAGT ASD TGTGTGGGTGACACAGATGA GTTGAAGGCTGATTGGTGAA A6D CCACCGACATTTCCAACAC GGGCAGGTATTTCAGCTTC'IT A9D TCAACTTCACCACGTTCTTCA CTCCCGTCTCCAGTTCTCTT Cyclophilin CTTCTTGCTGGTCTTGCCATTCCT GGATGGCAAGCATGTGGTCTTTG B-Actin GACGGCCAGGTCATCACTAT CGGATGTCAACGTCACACT’I‘ Human Elovl 1 GCTGGCTGAGCACCTATACC TCAGCTCAATGAACTTGGAGAA Elov12 CCCTTCGG'ITGTCTCATC'IT CAGGTGGCTCTTGCATATCTT Elov15 GTGCACATI‘CCCTCTTGGTT TGGTCCTTCAGGTGGTCTI‘T Elovl6 CTAAGCAAAGCACCCGAACT GGCAACCATGTCT’ITGTAGGA ASD TTGGCCTGGATGATTACCTT CTGTGTCACCCACACAAACC A61) ATCCCTTTCTACGGCATCCT TAGGCCTCCTGGTCAATCTC A9D CACCCAGCTGTCAAAGAGAA GATGAAGCACATCATCAGCAA [3-Actin CTCTTCCAGCC'ITCC’ITCCT TGTI‘GGCGTACAGGTCTTI‘G 73 membranes. Membranes were incubated with antibodies for SREBP-1 (IgG-2A4, sc- 13551) (Santa Cruz Biotechnology, San Cruz, CA). HNF-4a (C-19), MLX (N-l7), anti- goat and anti-rabbit antibodies were obtained from Santa Cruz Biotechnology (Santa Cruz, CA). ChREBP antibody was obtained from (Novus Biologicals, Littleton, CO). Anti- mouse and anti-rabbit secondary antibodies were obtained from Bio-Rad (Hercules, CA); anti-goat antibodies were obtained from Santa Cruz Biotechnology. The SuperSignal West Pico chemiluminescence kit (Pierce) detection system was used. Recombinant adenovirus. Cloning of cDNA for Elov12, Elov15 and Elovl6 was described previously [175]. The coding region for each transcript was ligated into Ad-Easy XL adenoviral vector system (Stratagene), recombined in BJ 5183 cells and propagated in XL10 Gold ultra-competent cells. Ad-DNA was packaged into adenoviral particles Ad- 293 cells. The resultant adenovirus was amplified in l-IEK293 cells. Recombinant adenovirus expressing dominant negative MLX (Ad-anLm and the doxycycline inducible nuclear SREBP-1c (Ad-nSREBPl) were obtained from H. Towle, University of Minnesota, Minneapolis, MN [62,180]. Adenovirus was propagated and amplified Hek293 cells. Viral lysates were titered using the Adeno-X Rapid Titer Kit, Clontech. Confluent primary hepatocytes were infected (5-10 plaque forming units [PFU]/cell). Using Ad-GFP as a control for infection, greater than 80% of primary hepatocytes expressed functional protein at the 5-10 PFU/cell level. In vitro fatty acid elongation assay. Rat liver microsomes were isolated by differential centrifugation [159]. Elongation reactions were carried out with modifications to the procedure described by Moon et al [117]. Briefly, reaction mixtures contained 50 ug microsomal proteins in a total reaction volume of 100 pl. The reaction constituents are: 74 50 mM potassium phosphate buffer- pH 6.5, 5 11M rotenone (Sigma), 40 11M fatty acyl CoA (Avanti Polar Lipids, Alibaster, Al and Sigma, St. Louis, MO), 60 11M malonyl CoA (Sigma) [6.5 dpm/pmol] [2-14C]-malonyl CoA (Perkin Elmer), 1 mM NADPH (Sigma) 20 uM BSA (fatty acid free). Reactions (at 37°C) were initiated with the addition of NADPH. When fatty acids were used as substrate, NaOH neutralized fatty acid (40 uM) replaced fatty acyl CoA. CoASH (at 100 11M), MgC12 (1 mM) and ATP (1 mM) were added to the reaction to generate fatty acyl-CoA. Elongase reactions were terminated after 20 minutes with the addition of 100 pl 5 N KOH + 10% methanol; lipids were saponified for 1 hr at 65°C. The saponification reaction was acidified with 100 pl 5 N HCl; 100 pl ethanol was added to aid hexane extraction of fatty acids. Elongated fatty acids were collected by 2 independent extractions with hexane (800 pl). Hexane extracts were pooled and l4C-radioactivity was quantified by B-scintillation counting. Results are expressed as Elongase Activity Units, mnoles l4C-malonyl CoA incorporated/mg protein/20 mins. F ormation of reaction products was dependent on the presence of NADPH and the fatty acid CoA. Fatty acid elongation products were verified by reverse? phase-HPLC chromatography using a flow-through B-scintillation counter [53]. Statistical Analysis. Statistical analysis used Student’s t-test and ANOVA plus post hoc Tukey HSD test (http://faculty.vassar.edu/lowry/VassarStatshtml). 75 3.3 Results 3.3.1. Elongase and desaturase expression in rat and mouse liver. Our first objective was to compare fatty acid elongase and desaturase expression in rat and mouse (Fig. 3.1A). Of the 7 elongases identified in the genomes of these species, qRT-PCR analysis indicated that only 4 elongases are expressed in liver, Elovll, Elov12, Elov15 and Elovl6. Based on relative mRNA abundance, the hierarchy of elongase expression in rat and mouse liver is: Elov15 > Elovll = Elov12 = Elovl6. The hierarchy of desaturase expression is: A9D (stearoyl CoA desaturase-1, SCD-l) > Asdesaturase (A5 D) = A6 desaturase (A6D). Fatty acid elongase activity was assessed using 3 substrates. 16:0-CoA is a substrate for Elovll and Elovl6; 20:4-CoA is a substrate for Elov12 and Elov15; and 24:0- CoA is a substrate for Elovll [153]. Elongation of 16:0-CoA to 18:0-CoA was highest in mouse liver, while elongation of 24:0-COA to 26:0-CoA was comparable amongst speCies. Differences in elongation activity can be attributed to elongase subtype expression amongst species. 3.3.2. Role of PPARa in the control of hepatic elongase and desaturase expression. Feeding rats the PPARa agonist, WY14643, induces certain hepatic fatty acid elongases and desaturases and promotes changes in hepatic and plasma lipid composition [26]. Herein, we evaluate further the role PPARa plays in the control of hepatic elongase and desaturase expression. The effect of WY14643 on elongase and desaturase expression in wild type and PPARa null (-/-) mice was examined. Mice were fed a control diet or one containing WY14643 (at 50 or 500 mg WY14643/kg of diet) for l 76 Figure 3. 1. Fatty acid elongase and desaturase expression in rat, mouse and human liver. A. Liver from male Spraque-Dawley rats (2-3 months of age) and C57BL/6 mice (2-4 months of age) were used for RNA extraction and measurement of elongase and desaturase expression as described in Materials and Methods. Rats and mice were maintained on chow diets ad lib. mRN A abundance for each elongase and desaturase was measured by qRT-PCR. Results are expressed relative to an internal standard, Bactin (transcript/Bactin) mean : SD,n=4. B. Rat and mouse were extracted for microsomes to assay elongase activity using three separate substrates, 16:0-CoA, 20:4-CoA and 24:0-COA. Results are expressed as Elongase Activity (nmoles l4C-malonyl CoA assimilated into fatty acids/mg protein). Mean : SD,n=4. *p50.05 vs rat liver, ANOVA. 77 3.1A 0.41 * .° to on C1 rat I mouse ogzuifilflifl Elovl1 Elovl2 Elovl5 Elovls DGD D90 .° MP 01:» O L; 0| Transcript/Actin O N 0 d 3.18 A N -h 1 1 X’ D rat I mouse —L ‘ l | Elongase activity onhmmo l 16:0-GOA 20:4-CoA 24:0-CoA 78 week (Fig. 3.2). Of the 4 elongases expressed in mouse liver, WY14643 induced only Elov15 and Elovl6, 14- and 4-fold, respectively. Previous studies established that the PPARa agonist increased desaturase expression [134]. Our results indicate thatiAéD‘and A9D transcript levels were induced ~6-fold by WY14643, while A5 D was weakly induced. Elovll and Elov12 mRNA abundance was unresponsive to WY14643 treatment. The absence of a WY14643 effect on elongase and desaturase expression in PPARa (-/-) mice indicates that PPARa plays a role in controlling both elongase (Elov15 and Elovl6) and desaturase (A6D and A9D) expression. 3.3.3. Regulation of elongase and desaturase expression in primary rat hepatocytes. Insulin, SREBP-1c [134,181], LXR agonist (T0901317, T1317), glucose, ChREBP and MLX [62] control desaturase expression [9]. The following studies determined if these same transcriptional regulatory systems control elongase expression in rat primary hepatocytes. Regulation of elongases and desaturases by insan and LXR agonist. Insulin regulates lipid synthesis, at least in part, by controlling SREBP-l nuclear abundance [179,182]. LXR agonist stimulate lipogenesis through direct and indirect mechanisms [183]. LXR/RXR heterodimers bind LXRE in promoters of responsive lipogenic genes. LXR agonist also induce lipogenic gene expression through the induction of SREBP-1c gene transcription [182,183]. The effect of insulin and T1317 on hepatocyte elongase and desaturase expression was examined. In the absence of insulin or T1317, SREBP-1 nuclear abundance in hepatocytes is low (Fig. 3.3 insert). Treatment of rat primary hepatocytes with insulin or T1317 induced 79 16 12 (P [00 I50 0500 no 350 msoo Elovl-1 Elovl-2 Elovl-5 Elovl-6 DSD 06D D9D Figure 3. 2. The role of PPARa in the control of hepatic elongase and desaturase expression. Homozygous wild type and PPARa-null (-/-) mice on a Sv/129 genetic background [177,178] were fed either a control diet or one containing WY14643 (at 50 or 500 mg/kg of diet; Bioserv, Piscataway, NJ) for 1 week. Liver RNA was extracted and used as template for qRT-PCR. Results are reported as Fold Change in mRNA abundance (transcript/cyclophilin) for each enzyme, mean : SD,n=4. *p50.05, Students T-T est vs wild type on a control diet. 80 nuclear SREBP-1 ~4-fold, but had no effect on SREBP-2 nuclear abundance. T1317 (1 uM) had no significant effect on Elovll, -2 or -5 expression in rat primary hepatocytes and only modestly induced Elovl6, ~l .5-fold (Fig. 3.3). In contrast, all 3 desaturases were induced between 2- and 15-fold; A9D was most responsive. Insulin induced Elovl6 and A51) 51.5-fold, while A6D and A9D were induced >3-fold. Co-treatment with insulin and T1317 had no additive effect on SREBP-1 nuclear abundance or the expression of any elongase or desaturase. These studies suggest that the induction of Elovl6, ASD, A6D and A9D by insulin and T1317 likely involves control of SREBP-1 nuclear abundance. Although others have reported that insulin induces LXRa in primary hepatocytes [184], we found no evidence for an insulin effect on either LXRa or LXRa mRNA abundance (not shown). Glucocorticoids, T3 and leptin had no effect on elongase expression in primary rat hepatocytes. None of the hormones tested induced hepatic Elovl3, Elovl4 or Elovl7 (not shown). Effect of over expressed nuclear SREBP-1c on elongase and desaturase expression. To further evaluate the SREBP-1 control of elongase and desaturase expression, primary hepatocytes were infected with a recombinant adenovirus containing a doxycycline-inducible nuclear form of SREBP-1c [Ad-nSREBP-lc] [180]. These cells received no insulin or LXR agonist. Treatment of primary hepatocytes with doxycycline significantly induced the nuclear form of SREBP-1c (not shown) as well as the endogenous SREBP-1 transcript (Fig. 3.4). The endogenous SREBP-1c promoter contains a SRE and is activated by elevated levels of nuclear SREBP [182]. Over 81 Veh Ins T1317 Both H—ir-A-y—L-‘r-L-s InSREBP-1 | W] |nSREBP-2Ip~———~| lEIVehicle I Insulin 1:1 T1317 8 Both] Fold Change 3 a 01 Elovl-2 Elovl-5 Elovl-6 D5D D6D DQD Elovl-1 Figure 3. 3. Effect of insulin and LXR agonist (Tl317) on fatty acid elongase and desaturase expression in rat primary hepatocytes. Rat primary hepatocytes were maintained in Williams E medium containing 10 mM lactate, 10 nM dexamethasone . (DEX) with no insulin overnight. The next day, cells were switched to Williams E medium containing 25 mM glucose, 10 nM DEX (Vehicle) in the presence or absence of insulin (100 nM) or the LXR agonist, T0901317 [T13l7, Cayman Chemical Co. Ann Arbor, MI] (1 11M). After 24 hours of treatment, cells were harvested for RNA extraction and the measurement of nuclear SREBP-1 and SREBP-2 by irnmunoblot (insert, duplicate samples/treatment). Elongase and desaturase mRN A abundance was quantified by qRT-PCR. Results are expressed as the Fold Change in mRNA versus Vehicle (transcript/cyclophilin); mean : SD,n=4. These results are representative of 2 separate studies. *p_<_0.05 vs vehicle, ANOVA. 82 expressed nSREBP-lc induced transcripts encoding Elov12, Elovl6, A5 D, A6D and A9D ~3-fold at the maximal doxycycline dose. In a second study, Ad-nSREBP-lc infected cells were treated with insulin or T1317 (Fig. 3.4B). The goal of this study was to determine if insulin or T1317 had effects on elongase or desaturase expression independent of SREBP-1c. Over expressed SREBP- 1c (Ad-SREBP-lc) induced Elov12 (2.6-fold) and Elovl6 (6-fold), while ASD, A6D and A9D transcripts were induced 3.6-, 4.8- and 3.1—fold, respectively. Addition of insulin had no additional effect on the expression of any elongase or desaturase. Addition of T1317 induced Elovll 50%, while A9D was induced 4-fold. The expression of no other elongase or desaturase was affected by T1317. Based on these results, the induction of Elovl6, ASD, A6D by insulin and T1317 (Fig. 3.4A) is due to elevated SREBP-1 nuclear abundance. Only A9D expression is induced by elevated SREBP-1 nuclear abundance and independently by LXR agonist. Elovll was modestly responsive to LXR agonist only when SREBP-1 was over expressed. Elov15 mRNA abundance was unresponsive to changes in insulin, LXR agonist and SREBP-1 nuclear abundance (Fig. 3.4B). Glucose, ChREBP & MLX regulation of elongase and desaturase expression. Insulin stimulated glucose metabolism induces the translocation of ChREBP to the nucleus where ChREBP/MLX heterodimers bind to carbohydrate regulatory elements (ChoRE) in promoters of glucose responsive genes involved in glycolysis and lipogenesis [185,186]. The effect of glucose on elongase and desaturase expression in primary hepatocytes was examined. Primary hepatocytes were maintained in medium containing lactate (20 mM) and insulin (100 nM) or switched to medium containing 25 mM glucose 83 Figure 3. 4. Effect of over expressed nuclear SREBP-1c on elongase and desaturase expression. A. Primary rat hepatocytes in Williams E medium + 10 mM lactate + 10 nM DEX, but no insulin, were infected with a recombinant adenovirus expressing the nuclear form of SREBP-1c under the control of doxycycline [180]. Cells were switched to Williams E medium + 25 mM glucose with no insulin in the absence or presence of doxycycline (25, 200 or 250 ng/ml). Cells were harvested 24 hrs afterward for RNA extraction and measurement of elongase and desaturase mRN A abundance by qRT-PCR (transcript/cyclophilin). Endogenous SREBP-1 expression was quantified by northern analysis. Results are represented as Fold Induction by SREBP-1c, mean i SD,n=4. These results are representative of 2 separate studies. *p<0.05 vs vehicle, ANOVA. B. Primary rat hepatocytes in Williams E medium + 10 mM lactate + 10 nM DEX, but no insulin, were infected with a recombinant adenovirus expressing the nuclear form of SREBP-1c under the control of doxycycline [180]. Cells were switched to Williams E medium + 25 mM glucose with doxycycline at 250 ng/ml, in the absence or presence of insulin (100 nM) or T1317 (1 uM). Cells were harvested 24 hrs afterward for RNA extraction and measurement of elongase and desaturase mRN A abundance by qRT-PCR (transcript/cyclophilin). Results are represented as Fold Induction by insulin or T1317, mean : SD,n=3. These results are representative of 1 study. *p<0.05 vs vehicle, ANOVA. 84 9‘ A i? o E 8 ' If! 7 _ Doxcyclrne * a: 5 . DOIZ5D1OOI25O . , 0) fig >5 5 " a}: g 4 D ** * * * * E; 11':- 3 - 3:: *3 ' E»? 0 .3: o, 5‘ v‘j = 2 - =3: :3: ' i it '~: ‘3 a: - s: s a ’sr E 1 ES: .. n s: s E: is volllslzslsi iil ilsils o u. .'\ fir $3 f0 O Q Q 0 \04\ \°‘\\ \04\ \°‘\\ 0‘) 0% 0% <3» 0 Elovl-1 Elovl-2 Elovl-5 Elovl-6 DSD L-PK mRNA Abundance Relative to Glucose Treatment A L O I Figure 3. 5. Effect of glucose on elongase and desaturase expression. Primary rat hepatocytes in Williams E medium + 10 mM lactate + 10 nM DEX + 10 nM insulin were switched to Williams E medium + 25 mM glucose + 10 nM DEX + 10 nM insulin. After 24 hrs of treatment cells were harvested for extraction of nuclear proteins and RNA. The nuclear abundance of ChREBP and MLX was measured by irnmunoblotting at the times indicated [59] (Insert). Elongase, desaturase and L-pyruvate kinase (L-PK) mRNA i - abundance was quantified by qRT-PCR (transcript/cyclophilin). White bars, cells maintained in lactate; black bars, cells switched to glucose. , In a second experiment, primary hepatocytes in Williams E medium + 10 mM lactate + 10 nM DEX + 10 nM insulin were infected with recombinant adenovirus expressing luciferase (Luc, gray bars) or dominant negative MLX (dn-MLX, checked bars). After 24 hours of treatment, cells were switched to Williams E medium + 25 mM glucose + 10 nM DEX + 10 nM insulin. After 24 hrs of glucose treatment, cells were harvested for RNA extraction and elongase, desaturase and L-pyruvate kinase (L-PK) mRN A expression was quantified by qRT-PCR (transcript/cyclophilin). Results are represented as mRN A Abundance, Relative to Glucose Treatment, mean i SD,n=4. These results are representative of 2 separate studies. *p<0.001 vs glucose treated cells, ANO VA. 87 specificity was examined using saturate (16:0), monounsaturated (16:1,n-7), and polyunsaturated (18:3,n-6, 20:5,n-3 and 22:5,n—3) fatty acids. Ad-Luc infected cells served as a control for basal elongase activity using the various substrates. Ad-Elov12 infected hepatocytes elongate only 20:5,n-3 and 22:5,n-3 (Fig. 3.6). Ad-ElovlS infected cells elongated 16:1,n-7, 18:3,n-6 and 20:5,n-3. Ad-Elovl6 infected cells elongated only 16:0 and 16:1,n-7 (Fig. 3.6). 3 Expression of hepatic elongases and desaturases is controlled by PPARa, SREBP- ] and ChREBP/MLX (Fig. 3.2-3.5). To determine if changes in elongase and desaturase activity impact hepatocyte fatty acid composition, primary hepatocytes incubated with insulin (induces SREBP-1), PPARa agonists and glucose (induces ChREBP) were examined for the capacity to elongate and desaturate l4C-16:0. Primary hepatocytes were maintained in Williams E media with lactate and no insulin overnight. This treatment effectively lowers SREBP-1 and ChREBP nuclear abundance [59,179]. The next day cells were treated with lactate or glucose containing media in the absence or presence of insulin and WY14643. All cells received 100 uM l4C-16:01 After 24 hrs of treatment, cells were harvested for lipid extraction and analysis of elongation and desaturation products by RP-HPLC. Cells maintained in lactate media with no insulin or WY14643 do not desaturate 16:0 (Fig. 3.7B, C & D), but elongate 16:0 to 18:0. Addition of insulin or WY14643 to lactate-treated cells induces formation of 16:1,n-7, while only WY14643 induces formation of 18:1 (n-7/n-9). The combination of these treatments has no additive effect. In cells switched to glucose-containing media, 16:0 desaturation to 16:1,n-7 and the elongation and desaturation of 16:0 to 18:1,n-7/n-9 is induced when compared‘to the 88 D Ad-Luc D Ad-Elovl-Z I Ad-Elovl-5 I] Ad-Elovl-6 0) O I 01 T _L —L N N 01 O I I O I XXXXXXXXXXXXXXXX Elongase Activrty (nmolelmg protein) Klil lALALLIiLAKilLiILLI our Palmitate 16:1,n—7 18:3,n-6 20:5,n-3 22:5,n—3 Fatty Acid Substrate Figure 3. 6. Substrate specificity of hepatic fatty acid elongases. Recombinant adenovirus expressing luciferase (Luc) [white bar], Elov12 [checked bar], Elov15 [black bar] and Elovl6 [gray bar] were used to infect primary hepatocytes (5 PFU/cell). After 24 hrs, cells were harvested for elongase activity using various fatty acid substrates (see Methods and Materials). Results are expressed as Elongase Activity (nmoles 4C-malonyl CoA assimilated into fatty acid/mg protein). Results are represented as mean 1; SD, n=3. Results are representative of 2 separate studies. - . - 89 lactate-treated cells. The combination of glucose + insulin or glucose + WY14643 induced formation of both 16:1,n-7 and 18:1,n-7/n-9 synergistically. Glucose, insulin and WY14643 have no apparent effect on the elongation of 16:0 to 18:0. Since 18:0 is a substrate for A9D, measuring changes in 18:0 may not accurately reflect effects of these treatments on elongation activity. In an effort to reveal an effect on elongation, we examined the effect of glucose, insulin and WY14643 on the 18:1 to 16:1 ratio (Fig. 3.7E). This ratio would remain constant if these treatments did not regulate elongation. The most impressive effect on the 18:1/16:1 ratio is seen in cells maintained in lactate- containing medium supplemented with WY14643. Such studies indicate that hepatocyte levels of 18:1 (n-7/n-9) are controlled by both elongation and desaturation pathways. The elongation pathway is not a constitutive pathway. Cellular levels of 18:1 are not determined solely by regulating A9D expression. 3.3.5. Regulation of elongase and desaturase expression in animal models of metabolic disease. PPARa [188], SREBP-1 [189] and glucose metabolism, ChREBP & MLX [186] and LXR [190] play an important role in metabolic diseases like diabetes and obesity . Herein, we will determine if changes in hepatic lipid metabolism and composition induced by diabetes and obesity can be attributed to changes in elongase and desaturase expression. Three metabolic disorders were examined, streptozotocin-induced diabetes, glucose intolerance induced by high fat diets, obesity induced by leptin deficiency. Nuclear levels of SREBP-1, ChREBP, MLX and HNF-4a were monitored to correlate changes in nuclear content of these transcription factors with changes in elongase and desaturase expression. 90 14 Elovl-6 A’D 3.7A. C-16:0 ——>18:O —>18:1,n-9 A9D Elovl5 16:1,"..7 —18:1,n-7 Elovl-6 3.73. D Lactate I Glucose _ 10 - 0 g ‘- 8 ' .2 to a "' 6 ‘1’ 3 E o 4 0 co 0 ‘- 2 .\° 0 . Veh lnsulin WY14643 Both 3.76. CI Lactate I Glucose d ON-FOQO % Conversion of 16 0 to 181 Veh Insulin WY14643 Both 91 3.7D. El Lactate I Glucose '8 10 - E: 8 - i1; 5 ' 2° 4 ' 8:2 2 ' :2 0 L— Veh Insulin WY14643 Both 3.7E. El Lactate I Glucose 21.5 - z; 1 . “0.5 - l I ~ 0 33 o "‘ Veh Insulin WY14643 Both Figure 3. 7. Effects of insulin and WY14643 on monounsaturated fatty acid synthesis. A. The pathway for conversion of 16:0 to 16:1,n-7, 18:1,n-7 and 18:1,n-9 by Elov15, Elovl6 and A9D is diagrammed. B-E. Primary rat hepatocytes in cultured in Williams E + 10 nM Dex + 10 mM lactate overnight were maintained in the same medium or switched to Williams E + 10 nM Dex + 25 mM glucose in the absence or presence of insulin (100 nM) or WY14643 (50 uM). Cells received 100 uM l4C-l6:0 + 20 uM bovine serum albumin for 24 hrs. After 24 hrs of treatment, cells were extracted for total lipid and the lipid was saponified and the distribution of 14C-amongst 16:0, 16:1,n-7, 18:0 and 18:1,n-7 and n-9 was measured by RP-HPLC and a flow-through B- scintillation counter. Technical limitations preclude resolution of 18:1,n-7 and 18:1,n-9 by RP-I-IPLC. As such, the results are reported as 18:1. Results are reported as (B) % Conversion of l4C-16:0 to l4C-16:1; (C) % Conversion of l4C-16:0 to l4C-18:1; (D) % Conversion of l4C-l6:0 to l4C-l8:0; (E) ratio of l4C-18:1 to l4C-l6:l, an index of elongation. Results are the mean of duplicate sample. Results are representative of 2 separate studies. 92 Streptozotocin-Indueed Diabetes. Rats made diabetic using streptozotocin have elevated blood glucose (378 i 21 mg/dl) when compared to control animals (77.9 i 5.2 mg/dl). Liver nuclei from diabetic rats contained little detectable nuclear SREBP-1 and suppressed levels of MLX, but no significant change in ChREBP or HNF-4a (Fig. 3.8A). Diabetes suppressed the expression of lipogenic and glycolytic genes, e.g., fatty acid synthase and L-PK, by 370% [176], while expression of the gluconeogenic enzyme, phosphoenolpyruvate carboxykinase (Peka) was induced 3-fold (Fig. 3.8). The decline in the nuclear abundance of SREBP-1 and MLX correlated with a ~45% decline in Elovl6 and a >95% decline in A9D mRN A abundance. Other elongases and desaturases remained unaffected. Examination of hepatic lipid composition revealed a significant 30% decline in 16:0 in diabetic animals, but no change in other saturated, mono- or polytursaturated fatty acids. The decline in 16:0 is consistent with the decline in de novo lipogenesis in livers of diabetic rats [176]. Glucose-Intolerance Induced by High Fat Diets. High fat diets induce glucose intolerance, insulin resistance, fatty liver and altered hepatic metabolism [191,192]. To examine the effects of diet induced diabetes on elongaSe and desaturase expression, male C57BL/6 mice were fed a diet with 10% of the calories as fat (low fat, lard diet) or 60% of the calories as fat (high fat, lard diet). Animals fed the low fat diet are 29 g and have blood glucose and insulin levels within the normal range, 121 mg/dl and 0.5 ng/ml (Table 2). Animals fed the high fat diet are ~44 g, with elevated blood glucose (152 mg/dl) and insulin (3.9 ng/ml). When compared to the low fat fed group, high fat fed animals were glucose intolerant (Fig. 3.9A) and have fatty 93 e #3 6° \6 3.8A Control Diabetic «0 Value " Microsomal SREBP—1 0.7 3.8B N (a) h- 1 Fold Change O Elovl1 Elovl2 Elovl5 Elov|6 D5D 06D DQD Peka Figure 3. 8. Effect of streptozotocin-induced diabetes on hepatic elongase and desaturase expression. Male Spraque-Dawley rats were made diabetic using streptozotocin as described in Materials and Methods. Livers from control and diabetic animals were used for the isolation of nuclear and microsomal proteins for immunoblotting (A) and RNA extraction for qRT-PCR (B). [A] Effect of diabetes on SREBP-l (microsomal [pSREBP-l] and nuclear [nSREBP-1]), and nuclear ChREBP, MLX and HNF-4a abundance. Protein levels were measured by immunoblot (Materials and Methods). Duplicate samples for each treatment are shown. The effect of diabetes on the abundance of these proteins was quantified and expressed as Fold Change (mean,n=5) induced by diabetes. Statistical significance (p-value) was assessed by Student’s T-test. [B] Effect of diabetes on elongase, desaturase and phosphoenolpyruvate carboxykinase (Peka) expression. Transcripts encoding fatty acid elongases, fatty acid desaturases and cyclophilin were quantified by qRT-PCR, while Peka was quantified by northern blot analysis. Results are expressed as Fold Change in expression (transcript/cyclophilin) induced by diabetes. *p<0.05 vs control; N=5/group, Students T-test. 94 livers [191]. Nuclear levels of SREBP-l and MLX were suppressed 60% and 50% (Fig. 3.9B), while ChREBP and HNF-4u nuclear levels remained unchanged. Expression levels of Elovll and Elov12 were not significantly affected by high dietary fat (Fig. 3.9C). Elov15 and Elovl6 mRNA abundance, however, was suppressed by 50% and 75% with the high fat intake. The high fat diet had no effect on A5 D or A6D, but significantly (280%) suppressed A9D expression. High fat diets significantly suppressed fatty acid elongation of 16:0-CoA and 20:4-CoA, but not 24:0-CoA (Fig. 3.9D). Palmitic (16:0), stearic (18:0), oleic (18:1,n-9) and linoleic (18:2,n-6) acids represent the major fatty acids in both diets. Analysis of hepatic lipid composition indicates that 18:2,n-6 accumulates relative to 20:4,n-6 in livers of high fat fed animals (Fig. 3.9E). 18:2,n-6, an essential fatty acid, is converted to 20:4,n-6. Since neither diet contains 20:4,n-6, the appearance of 20:4,n-6 in the livers requires conversion from 18:2,n-6 to 20:4,n-6 by elongation (Elov12 and Elov15) and desaturation (A5 D and A6D). In the low fat diet, the ratio of 20:4,n-6 to 18:2,n-6 is 0.75. In the high fat diet the ratio is 0.3. Failure to convert 18:2,n-6 to 20:4,n-6 is consistent with a decline an Elov15 expression, a key enzyme involved in PUFA synthesis. Elongase and Desaturase Expression in Livers of Lean (Lep°b’+) and Obese (Lep°w°b) Mice. Defective leptin expression in C57BL/6J-Lep0b’0b mice leads to hyperphagia, hyperinsulinemia, insulin resistance and obesity [193]. When compared to the lean (LepOb/U littennates, obese animals are heavier and have elevated blood levels of glucose and insulin (Table 3.2). The livers of obese mice are massively engorged with lipid, 95 Figure 3. 9. Effect of high fat diets on hepatic elongase and desaturase expression. Male C57BL/6 mice were fed 10% or 60% lard diets for 10 weeks as described in (Materials and Methods) [191]. [A] After nine weeks on the low and high fat diets, mice were assessed for glucose tolerance (Materials and Methods). Blood glucose was measured at the times indicated: 10% Cal (as fat), solid line- filled circle, 60% Cal (as fat), dashed line-filled square. Results are expressed as Blood Glucose (mg/d1), mean : SD,n=8 in each group. [B]. Effect of dietary fat on SREBP (nricrosomal [pSREBP-l] and nuclear [nSREBP-1]), ChREBP, MLX and HNF-4a. After ten weeks on the diet, livers were recovered for nuclear and microsomal protein and RNA extraction as in Fig. 8. Nuclear and microsomal proteins were measured as described in Materials and Methods. Representative immunoblots from duplicate samples for each treatment are shown. The effect of dietary fat on the abundance of these proteins was quantified and expressed as Fold Change (mean,n=4 animals/group) induced by the high fat diet. Statistical significance (p-value) was assessed by Student ’s T-test. [C] Effect of dietary fat on elongase and desaturase expression. RNA was extracted and used for qRT- PCR analysis of elongase and desaturase expression. Results are expressed as Fold Change (transcript/cyclophilin), mean : SD,n=8. [D] Effect of dietary fat on elongase activity. Hepatic microsomal preparations were used for fatty acid elongase assays (Materials and Methods). Results are expressed as Elongase Activity, (nmoles l4C-malonyl CoA assimilated into fatty acids/mg protein). Mean :1; SD,n=8. *p50.05 vs 10% Calories as fat, Student’s T -Test. [B] Effect of dietary fat on hepatic lipid composition. Total lipids were extracted and saponified; fatty acid levels were quantified by RP-HPLC (Materials and Methods). Results are expressed as Fatty Acid Mole%, mean : SD,n=8/ group; *p50.05 vs 10% lard diet, Students T -test. [10% Calories as Fat (open bars) and 60% Calories as Fat (filled bars)]. 96 3.9A coo E o a) i“ 60A: Cal E 400- \ v \ a, / i w 8 300— 2 (D '8 200— a": 10% Cal § 0 20 40 60 80 100 Minutes after Glucose Administration 393 10% Cal 60% Cal Fold Change .° 0. .5 O Elovl-1Elovl-2 Elovl-5 Elovl-6 D5D D60 D90 97 010% Calories as Fat D 3 3 a F s a s I .m b a C o/o o 6 I * u .o. 5 o >H=>BO< emamcom 24:0-CoA 20:4-CoA 16:0-COA 010% Calories as Fat I 60% Calories as Fat 0 0 0 0 0 5 4 3 2 1 3. 225 .22 En". E s 3 0 meduuu n.:.m”NN Medan o-:.v5~ m.:.n5N oéfiou mean»? w-:.~"m_. 21—32. 3235 5-9—nor 3333.. 98 predominantly as neutral lipid. Hepatic nuclei derived from obese mice have a 2.6- and 2- fold increase in SREBP-1 and MLX nuclear abundance, but no change in ChREBP or HNF-4a when compared to the lean littermates. Expression levels of Elov15, Elovl6, A5 D, A6D and A9D are increased in livers of obese mice when compared to the lean littermates. Obesity also results in the induction of lipogenic gene expression (ACC, F AS) and PPARa target genes, e.g., Cyp4A and AOX (not shown). Fatty acid elongation of 16:0- CoA is increased 2-fold, while elongation of 20:4-CoA and 24:0-CoA was not affected (Fig. 3.10C). Fatty acid analysis of livers from lean and obese mice reveals a ~9-fold increase in total esterified fatty acid. Much of this increase is in the form of neutral lipid esterified with 18:1 (n-7 and n-9) (Fig. 3.10D). While monounsaturated fatty acid abundance increased, hepatic PUFA content, i.e., 18:2,n-6, 20:4,n-6 and 22:6,n-3, declined. This might appear inconsistent with the observed increase in fatty acid elongase activity and elongase and desaturase expression. LepOb/Ob mice, however, are hyperphagic; ingestion of excessive calories as carbohydrate elevates plasma insulin (Table 2) and enhances de novo lipogenesis and monounsaturated fatty acid synthesis. Induction of Elov15, Elovl6 and A9D expression by activated PPARa and elevated SREBP-1 and MLX nuclear content facilitates monounsaturated fatty acid (18:1,n-7 and 18:1,n-9) synthesis which is assimilated into neutral lipid. Elongation, as well as desaturation, are key steps for converting the end products of de novo lipogenesis to monounsaturated fatty acids. These studies indicate that fatty acid elongation is induced along with de novo lipogenesis and Ag-desaturation. Induction of these pathways contributes to the fatty liver phenotype ob/ob characteristic of Lep mice. 99 Figure 3. 10. Effect of leptin deficiency on hepatic elongase and desaturase expression. Male lean C57BL/6J-Lep0b * and obese (C57BL/6J-Lep0b/0b) mice (B6.V-Lep ob/J , #000632, Jackson Labs) were maintained on a Harlan-Teklad laboratory chow (#8640) diet and water ad lib. Livers from these mice were used for the isolation of nuclear and microsomal protein for immunoblotting (A) and RNA extraction for qRT-PCR (B). [A] Effect of obesity on SREBP-1 (microsomal [pSREBP-l] and nuclear [nSREBP-1]) and nuclear ChREBP, MLX and HNF-4a abundance. Protein levels were measured by immunoblot (Materials and Methods). Triplicate samples for each phenotype are shown. The effect of leptin deficiency on the abundance of these proteins was quantified and expressed as Fold Change (mean,n=4) induced by leptin deficiency. Statistical significance (p-value) was assessed by Student’s T-test. [B] Effect of leptin deficiency on elongase and desaturase expression. RNA was extracted and used for qRT-PCR analysis of elongase and desaturase expression. Results are expressed as Fold Change (transcript/cyclophilin), mean : SD,n=4. *p<0.001 vs Lean; Students T - test. [C] Effect of leptin deficiency on elongase activity. Hepatic nricrosomal preparations were used for fatty acid elongase assays (Materials and Methods). Results are expressed as Elongase Activity, (nmoles l4C-malonyl CoA assimilated into fatty acids/mg protein). Mean j; SD,nfl. *p_<_0.01 vs Lean, Student is T -Test. D. Effect of leptin deficiency on hepatic lipid composition. Total lipids were extracted and saponified; fatty acid levels were quantified by RP-HPLC (Materials and Methods). Results are expressed as Fatty Acid Mole%, mean i SD,n=4/group; *p50.01 vs Lean animals, Students T -test. [Lean (open bars) and Obese (filled bars)]. 100 é" o c b0 0‘ Q L Ob 3.10 A. can ese p-vaIue Nuclear MLX Nuclear HNF-4or 3.10 B. 1 Fold Change 0 N I5 0) a O I I Elovl-1 Elovl-2 Elovl-5 Elovl-6 D50 D60 D9D 101 3.10 C. 0 Lean I Obese 05050 211 530.5 9.5.05... 3_>_uu< occur—Om. 20:4-COA 24:0-COA 16:0-COA 3.10 D. n-:.o"NN n.:.m"NN Mir—£5.“ win—{SN mic.n5N D Lean I Obese mus—JEN mean": 0-....N5r «At—Lav OHNL MGM—m hue—€59 season. III-lip 00000000 7654321 3. 222. .23. been. 102 Elongase and Desaturase Expression in Livers of TCDD treated mice. TCDD is a toxic compound causing controversial human health effects by altering gene expression via the activation of the aryl hydrocarbon receptor (AhR), a member of the basic-helix-loop-helix-PAS (bHLH-PAS) family [194]. Hepatotoxicity is the severe result of TCDD exposure and is characterized by hepatomegaly accompanied by hepatocyte hypertrophy, fat accumulation, immune infiltration, necrosis, and alterations in liver enzymes [195]. Expression level of Elov15 is increased, whereas Elovl-2, Elovl-6 and A5 D are suppressed by 50% to 60% in livers of TCDD treated mice when compared to the control mice (Fig. 3.11 A). The TCDD treatment had no effect on Elovll, A6D or A9D expression. Moreover, TCDD treatment significantly decreases fatty acid elongation of 16:0-CoA, but not 20:4-COA and 24:0-COA (Fig. 3.11 B). This is due to the suppressed expression of Elovl6 by TCDD treatment. Fatty acid analysis of livers from TCDD treated mice reveals a ~2-fold increase in total esterified fatty acid. Much of this increase is due to the accumulation of 16:0, 18:1n— 9 and 18:2,n-6 (Fig. 3.11C). Conversion of 18:2,n-6 to 20:4,n-6 is suppressed by 50% in TCDD treated mice, which is not consistent with an increase in Elov15 expression (Fig. 3.11D). The decreased expression of A5 D may account for it. 3.4. Discussion F atty acid elongation and desaturation are two key metabolic routes for the synthesis of saturated, mono- and polyunsaturated fatty acids. Of these, fatty acid desaturases have received considerable attention for their regulation by hormones and nutrient and their capacity to generate specific unsaturated fatty acids. The outcome of 103 Figure 3. 11. Effect of TCDD treatment on hepatic elongase and desaturase expression. Male C57BL/6 mice mice were treated by gavage with 0.1 ml of sesame oil for a nominal dose of 0 (vehicle control) or 30 ug/kg body weight of TCDD for 168hr as described in (Materials and Methods) [200]. [A] Effect of TCDD on elongase and desaturase expression. RNA was extracted and used for qRT-PCR analysis of elongase and desaturase expression. Results are expressed as Fold Change (TCDD/V EH), mean : SD,n=4. [B] Effect of TCDD on elongase activity. Hepatic microsomal preparations were used for fatty acid elongase assays (Materials and Methods). Results are expressed as Elongase Activity, (nmoles l4C-malonyl CoA assimilated into fatty acids/mg protein). Mean : SD,n=4. *p50.05 vs Veh, Student ’3 T -Test. [C.D] Effect of TCDD on hepatic lipid composition. Total lipids were extracted and saponified; fatty acid levels were quantified by RP-HPLC (Materials and Methods). Results are expressed as Fatty Acid Mole%, mean i SD,n=4/group. 104 3.11A Fold change TCDD VS VEH 3.11B 3.11C l P N 0| 1 1 i 0 0| 1 9 0| A 1 l o- Elovl-1 Elovl-2 Elovl-5 Elovl-6 D9D elongation activity (nmollmg protein) AAIlinoleic acid O-‘NU‘UIONOOO llllllllll 0.9 1 0.8 t 0.7 r 0.6 t 0.5 ‘ 0.4 ‘ 0.3 ' 0.2 t 0.1 ‘ 16:0, veh 105 D5D D60 DVoh ITCDD 1 24:0, TCDD these studies indicates that desaturases are well-regulated enzymes that play an important role in cellular and whole body lipid composition [9,160]. One of these enzymes, A9D (stearoyl CoA desaturase-1), has emerged as a key enzyme in the control of whole body lipid composition [196]. In contrast to the desaturases, fatty acid elongase have only recently been recognized as proteins regulated at the pretranslational level [139,187,197]. Our previous studies indicated that fatty acid elongases are regulated by tissue-specific and nutritional factors and during postnatal development [26]. Those studies implicated certain transcription factors, like SREBP-1 and PPARa, as regulators of both elongase and desaturase expression. The current report extends those previous observations by evaluating the role of several hormones (insulin, T3, glucocorticoids and leptin), transcription factors (SREBP-1c, PPARa, LXR, ChREBP and MLX) and nutrients (glucose and fat) in the control of hepatic elongase and desaturase gene expression, fatty acid elongase activity and lipid composition. Finding the involvement of these factors in the control of elongase expression prompted studies to evaluate how these enzymes were regulated in metabolic disease. The outcome of these studies has provided new information on how changes in both elongase and desaturase expression in metabolic disease contributes to hepatic lipid composition. Seven fatty acid elongase subtypes (Elovll to Elovl7) have been identified in the genomes of the rat, mouse and human genomes (www.cnsembl.org). Of these, 4 elongase subtypes are expressed in rat & mouse liver (Fig. 3.1). The hierarchy for hepatic expression of these enzymes is sirrrilar in these two species: 106 Table 3. 2. Dietary composition and physiological parameters of C57BL/6 mice Low Fat versus High Fat Lean versus Obese Parameter Low Fat High Fat Chow Chow Calories as fat (%) 10 60 20.3 20.3 Calories as carbohydrate 70 20 55.4 55.4 (%) Calories as protein (%) 20 20 24.4 24.4 Body weight (g) 29.0 i 1.0 44.0 :t 23.7 d: 1.5 55.1 i 2.8 1.0 Fasting blood glucose 121.0 :1: 1.0 152.0 :t 1 12.0 i 11 266.0 i (mg/d1) 0.3 69.0 Plasma insulin (ng/ml) 0.5 :t 0.05 3.9 i 0.4 0.2 + 0.06 15.4 d: 1.04 Animals per group 8 8 4 4 107 Elov15>Elovl1=Elov12=Elovl6. Analysis of elongase activity indicates that mouse liver has the highest elongase activity. Elovll. Elovll is a low abundance elongase in liver of rat and mouse. Based on studies in yeast, Elovll elongates a broad array of saturated and monounsaturated fatty acids. Elovll expression, however, is not regulated by any physiological manipulation used so far in this or our previous study [26]. Thus, changes in hepatic lipid composition induced during postnatal development, fasting and refeeding, diabetes, obesity, dietary fat, LXR or PPARa. agonist can not be attributed to changes in Elovll activity. Hepatic Elovll appears to be expressed constitutively. Elov12. Elov12 is also a low abundance elongase in liver of rat and mouse. In contrast to other elongases, Elov12 has a very narrow substrate preference; it elongates C20 and C22 PUFA (Fig. 3.6). As such, Elov12 participates in the conversion of essential fatty acid precursors to end products of PUFA synthesis, i.e., 20:4,n-6 and 22:6,n-3. Like Elovll, Elov12 is not regulated by any factors examined in this report or our previous report. The exception to this is the induction of Elov12 mRNA following over expression of SREBP-1c (Fig. 3.3). Since insulin, LXR agonist and glucose fail to induce this transcript, we feel the induction of Elov12 by overexpressed SREBP-lo may have limited physiological significance in viva. Elov15. Elov15 is the most abundant elongase transcript in rat and mouse. It also is expressed in many tissues, induced during postnatal development and suppressed by feeding rats n-3 PUFA enriched diets [26]. Several hormones (insulin, T3, glucocorticoids and leptin) and transcription factors (SREBP-1c, LXR, ChREBP and MLX) have no impact on hepatic Elov15 expression. Only PPARa, n-3 PUFA-enriched 108 [26], high fat diets (Fig. 3.9) and obesity (Fig. 3.10) affect Elov15 expression. Regulation of Elov15 has physiological significance. Feeding rats a high carbohydrate diet supplemented with olive oil plus WY14643 significantly increased mead acid (20:3,n-9) production [26]. Mead acid is an elongation and desaturation product of 18:1,n-9, the predominant fatty acid in olive oil. It is our view that WY14643 induction of Elov15 contributes to the formation of 20:3,n-9. Elov15 also converts 16:1,n-7, but not 16:0, to an 18-carbon monounsaturated fatty acid (18:1,n-7) (Fig. 3.6) as well as elongating an intermediate (18:3,n-6) in the pathway for n-6 PUFA synthesis (20:4,n-6) (Fig. 3.6). Suppression of Elov15 in high fat-fed mice correlates with decreased hepatic 20:4,n- 6/18:2,n-6 ratio (Fig. 3.9). Enhanced Elov15 expression correlates with the elevated content of l8-carbon monounsaturated fatty acids in livers of obese rrrice (LepOM’b) (Fig. 3.10). Many PPARa-regulated transcripts, like acyl CoA oxidase and CYP4A, are ”/0” mice (not shown). Induction of Elov15 in livers of obese mice induced in livers of Lep is likely due to PPARa activation. Mice exposed to TCDD, leading to fatty liver, have elevated Elovl-5 expression and enzyme activity (Fig. 3.11). Despite the role Elov15 plays in PUFA synthesis and its elevation in livers of obese mice, hepatic lipids in obese animals are not enriched in PUFA. In fact, obese livers are depleted of PUFA relative to other fatty acids like 18:1 (Fig. 3.10). ASD, A6D and A9D are induced in livers of obese mice, but to differing extents (Fig. 3.10). A5 D and A6D are induced in obese liver as a result of elevated nuclear abundance of SREBP-1 and activation of PPARa. A9D is induced by these same transcription factors, but also by elevated nuclear ChREBP/MLX (Fig. 3.10). Thus, hyperphagia resulting from defective leptin production, coupled with the ingestion of the high carbohydrate diet stimulates de 109 novo lipogenesis and monounsaturated fatty acid synthesis. In this instance, Elov15 substrates, in particular 16:1,n-7 (Fig. 3.6) are end products of de novo lipogenesis and A9D. Elevated expression of Elov15, Elovl6 and A9D, coupled with enhanced production of end products of de novo lipogenesis yields elevates 18:1,n-7 and 18:1,n-9 production. Elovl6. Elovl6 is a low abundance enzyme in livers of rat and mouse. Like Elov12, Elovl6 has a narrow substrate preference, i.e., C12 and C16 saturated and unsaturated fatty acids (Fig. 3.6) [117]. In contrast to the other elongases, Elovl6 is regulated by multiple regulatory factors. Insulin and LXR agonist induce SREBP-1 which induce Elovl6 expression (Fig. 3.3 & 3.4). Insulin-induced glucose metabolism induces ChREBP nuclear content. ChREBP and MLX regulate glucose-regulated genes including L-PK, FAS and A9D. Elovl6 is amongst these glucose-regulated genes (Fig. 3.5). Activation of PPARa also induces Elovl6 (Fig. 3.6). Elovl6 is regulated during postnatal development, but unlike Elov15, Elovl6 expression falls at birth and is induced at weaning. Elovl6 expression during early postnatal development parallels SREBP-1 nuclear abundance [26]. Finding that both Elovl6 and ND are induced along with L-PK and FAS (Fig. 3.10) indicates that these enzymes play a role in the hepatic response to excess carbohydrate consumption. Excess carbohydrate is channeled to de novo lipogenesis via enhanced L-PK activity. Insulin-stimulated glucose metabolism induces ChREBP translocation into hepatic nuclei [186,198,199]. ChREBP and MLX heterodirnerize and bind ChoREs in promoters of responsive genes, like LPK, ACC and FAS. Insulin also increases SREBP- 1 nuclear abundance leading to increased promoter occupancy of SREBP-1 on SRE in target genes, e.g., ACC, FAS and A9D. Consistent with this scenario is the increased 110 nuclear abundance of SREBP-1 and MLX in livers derived from obese animals (Fig. 3.10). The end product of de novo lipogenesis, 16:0, is elongated (Elovl6) and desaturated (A9D) to yield 18:1, the fatty acid accumulating in livers of obese mice. In this metabolic scheme, there appears to be a tight coordination between glycolysis, de novo lipogenesis, fatty acid elongation (Elovl6) and desaturation (A9D) that involves 3 transcription factors, ChREBP, MLX and SREBP-lo. Although these studies provide a link between ChREBP, MLX, SREBP-l and PPARa in the control of elongase expression, the mechanism(s) for this control remains undefrned. Whether this control involves direct interaction of these transcription factors with regulatory elements in the promoters of the elongases or indirect control through other mechanisms will require detailed analysis of the promoters for Elov15 and Elovl6. These studies are beyond the scope of this report. In conclusion, we have established that specific hepatic fatty acid elongases, Elov15 and Elovl6 are regulated in liver by nutrients (glucose and fat), hormones (insulin) and nuclear receptor agonist, i.e., LXR and PPARa agonist. ChREBP, MLX, SREBP-1, PPARa and LXR control elongase, as well as desaturase expression. Only A9D is independently regulated by LXR. Metabolic diseases, like diabetes and obesity, induce changes in hepatic lipid composition by controlling the function of key transcription factors that impact elongase and desaturase expression. These studies support the notion that regulation of both fatty acid elongase and desaturase expression may play an important role in managing hepatic lipid composition in response to changes in dietary and hormonal status. 111 Chapter 4 Effect of Fatty Acid Elongases on Gene Expression and Lipid Metabolism in Rat Primary Hepatocytes 4.1. Introduction Fatty acid elongases catalyze the regulated and rate limiting step in long chain fatty acid elongation. Fatty acid elongases work in concert with desaturases to produce fatty acids longer than 16 carbons. The fate of these fatty acids is to serve as substrate for: 1) phospholipids and membranes synthesis; 2) eicosanoid synthesis (prostaglandins, thromboxanes and leukotrienes); 3) B-oxidation and energy metabolism; 4) triglycerides and lipid storage. Disturbance in any of these metabolic pathways is linked to metabolic disorders such as hepatic steatosis, hyperlipiderrria, inflammation and insulin resistance. Seven fatty acid elongase (Elovl) isoforms, each the product of a separate gene, have been cloned and characterized in mammals [115]. Evidence for the importance of Elovl in physiology comes from mutant, knockout and transgenic mice. For example, Quaking and J impy are two mouse mutant strains that have dramatically decreased Elovl- ] mRNA level [126]. Elovl3 is induced in brown adipose tissue following exposure of animals to the cold [116]. The water barrier function was impaired in Elovl3 null rrrice. Elovl6 is induced in transgenic mice over expressing SREBP-l [24,78,134]. Among the 7 isoforms, four expressed in rat and mouse liver (Elovl-l, -2, 1-5 and - 6); Elovl-5 is the most abundant based on its mRNA abundance. Elovl-5 utilizes a broad array of fatty acids as substrates (€16-20) for MU FA and PUFA synthesis [24,115,201]. It 112 is induced during postnatal development and suppressed by feeding rats n-3 PUFA enriched diets [175]. Recently, we reported PPARa agonist (WY14,643) and leptin- deficient obesity (LepOb/Ob ) induce Elovl-5 mRNA and enzyme activity leading to changes in hepatic and blood lipid composition [201]. In unpublished studies, we have also found that mice exposed to TCDD, leading to fatty liver, have elevated Elovl-5 expression and enzyme activity5 . C20 PUF A are the most potent fatty acids activating PPARa and its target genes [53]. Both Elovl-2 and Elovl-5 elongate C20 fatty acids. Compared to Elovl-5, however, Elovl-2 is expressed at lower levels in liver; its hepatic expression is constitutive in all physiological and pathological conditions tested. Elovl-2 also has a narrow substrate preference, €20.22 PUFA. As such, we hypothesize that changes in hepatic Elovl-2 or Elovl-5 might affect PUFA control PPARa-mediated gene expression. To test this hypothesis, we developed recombinant adenovirus harboring the Elovl-2 (Ad-Elovl-Z) or Elovl-5 (Ad-Elovl-S). We used recombinant adenovirus technology to over express Elovl-2 and Elovl-5 in rat primary hepatocytes. The outcome of these studies show that altered expression of these enzymes significantly impacts hepatocyte lipid compoSitiOn and PPARa-regulated gene expression. 4.2 Materials and Methods Recombinant adenovirus. Cloning of cDNA for Elovl-2 and Elovl-5 was described previously [201]. Cloning of cDNA for ACS4 is performed as described [175]. Primers of acyl CoA synthetase-4 (AC S4) corresponding to its open reading frame (Forward: atggcaaagagaataaaagctaagc; Reversezttatttgcccccatacatc) were designed based 0n Sequences obtained fi'om Genbank and the location of the open reading frame was 113 determined using DNA-Star. Rat liver was used as template. The coding region for each transcript was ligated into Ad-Easy XL adenoviral vector system (Stratagene), recombined in B] 5183 cells and propagated in XL10 Gold ultra-competent cells. Ad- DNA was packaged into adenoviral particles in Ad-293 cells (Strategene). The resultant adenovirus was titered using Adeno-XTM rapid titer kit (BD Biosciences, Palo Alto, CA) and used to infect rat primary hepatocytes. Rat primary hepatocyte isolation and adenovirus infection. Male Spraque Dawley rats (Charles River Laboratories, Kalamazoo, M1) were maintained on Harlan-Teklad laboratory chow (#8640) and water ad lib. Rat primary hepatocytes were prepared from Teklad chow-fed (ad lib) male Spraque-Dawley rats, cultured on BioCoat (type I collagen) plates (Beckon Dickinson, Belford, MA.[201]. For RNA and protein extraction, cells were plated onto 100 mm type I collagen-coated plates (BD Bioscience, Bedford, MA) at 107 cells/plate in Williams E (Gibco/Invitrogen, Carlsbad CA), 10 mM lactate, 10 nM dexamethasone (Sigma, St. Louis), and 10 % fetal bovine serum (Gibco/Invitrogen). For adenoviral infection studies, cells were plated in the same media onto 6 well type 1 collagen-coated plates at 1.5 x 106 cells/well. The ratio of culture media to cell number was maintained constant for the different plating conditions. For treatments, hepatocytes were incubated in medium (Mlliams E + 10 nM dexamethasone without or with 100 nM insulin and/or 25 mM glucose. Confluent primary hepatocytes were infected (5-10 plaque forming units [PF U]/cell). Using Ad-green fluorescent protein (Ad-GFP) as a control for infection, greater than 80% of primary hepatocytes expressed functional protein at'the 5- 10 PFU/cell level. The control virus used routinely for these studies, however, was Ad- 114 luciferase (Ad-Luc). Ad—Luc was obtained from C. Rhodes, Pacific Northwest Research Institute, Seattle, WA. Acyl-CoA synthetase 4 (ACS4) activity assay Acyl-CoA synthetase activity for ACS4 was determined using an isotopic method [202]. The assay mix for the isotopic method included 300 mM Tris—HCl, pH 7.4, 150 mM MgC12, 2.25mM glutatlrione, 20 mM ATP, 0.2 mM CoA, 200 [1M fatty acid, and 10 nCi [14C]fatty acid in 0.1% Triton X-100 in a total volume of 100 [11. The reaction was started by adding 5 pg postnuclear homogenate protein and the mixture was incubated for 5 min at 37 °C. Reactions were terminated with 0.2 ml of Dole's solution, isopropanol: heptane: 0.5 M H2 804 (40/10/1; v/v). water (0.15 ml) was added and unreacted palmitate was removed in the organic phase by extracting 2-3 times with 0.3 ml heptane. [14C]fatty acyl-CoA formed during the reaction was measured by scintillation counting. ‘ RNA extraction, northern analysis and quantitative real time-PCR (qR T -PCR). Total RNA was extracted from primary hepatocytes and used for northern blot and qRT-PCR analysis as previously described [74,201]. 32P[cDNA] far LPK, SREBPl, thMG-CoA Synthase or CYP4A2 were as described previously [74,179]. Hybridization was quantified by Phosphorlmager analysis (Molecular Dynamics). Specific primers for real time PCR for each gene (Table 4.1) were designed using Primer Express software (Applied Biosystems, Foster City, CA). First strand cDNA was synthesized using the SuperScript II RNase H- Reverse Transcriptase (Invitrogen Carlsbad, CA). Synthesized cDNA was rrrixed with 2x SYBR Green PCR Master Mix (Applied Bibsystems) and various sets of gene-specific forward and reverse primers and subjected to real-time PCR quantification using the ABI PRISM 7700 Sequence Detection System (Applied 115 Biosystems). All reactions were performed in triplicate. The relative amounts of mRNAs were calculated by using the comparative CT method (User Bulletin #2, Applied Biosystems). Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRNA. Primers used for quantitative real time PCR are listed in Table 4.1. Lipid extraction and quantitation of hepatic fatty acids composition. Total lipid was extracted from rat primary hepatocytes in chloroformzmethanol (2: 1) plus 1 mM butylated hydroxytoluene [175]. 7-nonadecenoic acid (19:1) was added as a recovery standard at the time of extraction. Protein (Bio-Rad, Hercules, CA) was measured in extracts after the initial homogenization step. Total lipids were saponified, fractionated and quantified by reverse phase HPLC (RP-HPLC) using a YMC J-Sphere (ODS-H80) column and a gradient starting at 77.5% acetonitrile + acetic acid (0.1%) and ending at 100 % acetonitrile + acetic acid (0.1%) over 90 mins with a flow rate of 1.0 ml/min using a Waters 600 controller. Fatty acids were detected using both UV absorbance at 192 nm (Waters model 2487) and evaporative light scatter (Waters model 2420). Fatty acid composition and structures were confrnned at the MSU Mass Spectrometry facility by GC/MS (wumzbchmsuedu/facilities/massspec/indcx.html). Fatty acid standards for RP- I-IPLC were obtained from Nu-Chek Prep (Elysian, MN). Immunoblotting. Total, cytosolic, microsomal and nuclear protein extracts from rat primary hepatocytes were prepared as described previously [159,175]. Proteins (50-100 pg) extracted from microsomal or nuclear fractions were separated electrophoretically by SDS-polyacrylamide gel electrophoresis (NuPAGE 4-12% polyacrylamide Bis-Tris, Invitrogen) and transferred to nitrocellulose membranes. Membranes were incubated with 116 Table 4. l.Primer sequences used for gene expression by northern blot and quantitative reverse transcription-PCR. Gene Forward Reverse INSIG l TGCAGATCCAGCGGAATGT CCAGGCGGAGGAGAAGATG INSIG 2A GACGGATGTGTTGAAGGATTTCT TGGACTGAAGCAGACCAATGTC INSIG ZB CCGGCAGAGCTCAGGATTT AACTGTGGACTGAAGCAGACCAA SCAP ATGAGGAGCTGTGGAGGAAA CCGTGGATTCAGGTGTAGTGT CYCLOPHILIN CTTCTTGCTGGTCTTGCCATTCCT GGATGGCAAGCATGTGGTCTTTG 117 antibodies for, SREBP-1 (IgG-2A4, sc-13551), SREBP-2(Santa Cruz Biotechnology, San Cruz, CA). Anti-mouse and anti-rabbit secondary antibodies were obtained from Bio-Rad (Hercules, CA). The SuperSignal West Pico chemiluminescence kit (Pierce) detection system was used. Statistical Analysis. Statistical analysis used Student’s t—test and AN OVA plus post hoc Tukey HSD test (http://faculty.vassar.edu/lowrv/VassarStats.html). 4.3 Result 4.3.1. Efficacy of Ad-Elov12 and Ad-ElovlS infection in rat primary hepatocytes. Rat primary hepatocytes were infected with Ad—Elole, Ad-ElovlS, Ad-Elovl6 Ad-Luc (Luciferase) or Ad-BGal (B-galactosidase) at 10 pfu/cell for 4 hrs. After infection, medium was replaced with fresh medium containing 14C-20:5,n-3, 14C-20:4,n-6 or 14C- 16:0 at 100 uM for an overnight incubation. The total lipid was extracted and saponified fatty acids were fractionated by RP-HPLC as described in Methods. The results showed that the cells infected with Ad-BGal or Ad-Elov12 and treated with 14(2 labeled 16:0 converted 25% of 16:0 to 18:0 (Fig 4.1 B). However, cells infected with Ad-Elovl6i converted 55% of 16:0 to 18:0 (Fig 4.1 B) and cells infected with Ad-ElovlS converted 35% of 16:0 to 18:0 (Fig. 4.1 C). Primary hepatocytes typically converted 40% of exogenous C20 to C22. Infecting cells with Ad-BGal, or Ad—Luc did not affect this reaction. The cells infected with Ad-Elovl-2 and treated with 14C labeled 20:5,n-3 converted 70% of 20:5,n—3 to 22:5,n-3 and 24:5,n-3 (Fig 4.1 A). The cells infected with Ad-Elovl-S and treated with 14C labeled 20:4,n-6 convert 60% of 20:4,n-6 to 22:4,n-6, 118 cpa' 7° Ad-BGal. _ 1...... |. 60 50 ‘° ' 4 its}; | ' . 30 20 10 o .....' hnl.ln J. ‘0 I‘ll .Ah ‘llA.tti‘ ...l.L..ltx“Jli 0:00 10:00 20:00 30:00 40:00 50:00:43 cps 30 Ad-Elov12 4 24:5,}: | 20 10 ljlllllllLlJllLLJ 0:00 10:00 20:00 30:00 40:00 50:00.13 c’.‘ 1 - BO . 1 Ad-Elovl6 119 i p O O 4.1B. NI 0 I A Inn-IAAA lanulun - . a . a . 0:00 10200 zoéoo 30:00 aoEoo 50:00 min in. main] lannlnnlallnnlllmn AllnllAn-lnn N O I 0:00 roéoo 20E00 30500 soioo 50:00.1: 0:00 10:00 20:00 30:00 40E00 soEooi-du 120 4.1C. 5“ E Ad-Luc 5" : 16:0 40‘, 30 7. :00 mins 2:) for. 30 {on 4050) Won mi '1 50 : Ad-ElovlS 10 € .1 30 t otbo ' rot'oo ' 2050c 30:00 40:03 ‘ SOiOOmin 121 4.1D. Cps: 120‘? 110‘; 1003 3"; 2034.06 . _ Ad-Luc 80? . . . . 70‘} 60 3 50‘; 30“: 2234,06 20.; . . . . . . . 10“ ll GEM-m4 —1 L -.}L._._a_ Lil“, cgtoo F 10:00 ' 20:00 ' 30:00 ' 40:00 ' 50:000in 0- 80‘: 70': 60.3 . . . . .Ad-Elovls 50" 10*} ll . l | , , 0:00 - 10:00 f20500 f 30200 ' 40:00 50:00min Figure 4. 1. Fatty acid analysis of hepatocytes overexpressed Elov12, Elov15, Elovl6, BGal and Luc. Primary hepatocytes were infected with 10 pfu/cell Ad-Elov12, Ad-ElovlS, Ad-Elovl6, Ad-BGal and Ad-Luc respectively. Then, either ”(2-16:0 or 1“c-2o:5n-3 or l4C-20z4n6 (at 250 uM, 1.7 Ci/mole) were treated to these 4 group of cells. Total lipids were saponified, fractionated, and quantified by RP-HPLC. Distribution of l4C-16:0 (B and C) and 14C-20 (A and D) derivatives were fractionated by RP-HPLC. The level of radioactivity was quantified using a flow-through scintillation counter in line with the HPLC unit. 122 but not 24:4,n-6 (Fig. 4.1 D). This study shows that overexpression of Elov12 and Elov15 by recombinant adenovirus leads to increased conversion of their substrates to longer fatty acids. C20 and C22 are the substrates for the Elov12. C16 and C20 unsaturated fatty acids are the substrates for Elov15. This study indicates that recombinant adenovirus can be used to over express elongases in primary hepatocytes and change the lipid profile.— 4.3.2. Effect of Elov12 and Elov15 on fatty acid metabolism in rat primary hepatocytes. Further analysis of lipid profile in hepatocytes indicated that overexpression of either Elov12 or Elov15, reduced 20:5,n-3 level significantly, which is the most potent fatty acid activator of PPARa. Additionally, Elovl-2 increased 24:5,n-3 and Elovl-5 increased 22:5,n-3 level (Fig. 4.2 A and B). 4.3.3. Effect of Elov12 and Elov15 on gene expression in rat primary hepatocytes. Prior to the addition of fatty acids to primary hepatocytes, intracellular non- esterified unsaturated fatty acid levels are very low, representing 50.1% of the total fatty acyl chains in the cell. The relative abundance of putative PPARa ligands in the NEFA pool is 20:4,n-6 = 18:2,n-6 = 18:1,n-9 > 22:6,n-3 > 18:3,n-3/6 = 20:5,n-3. When compared to all PUFA in primary hepatocytes, 20:5,n-3 is the most active ligand for activating PPARa [136]. Since both Elovl-2 and Elovl-5 can convert C20 fatty acids'to C22 fatty acids, overexpression of Elovl-2 or Elovl-5 might attenuate 20:5,n-3 effects on PPARa-regulated gene transcript by rapidly reducing 20:5,n-3 mass in hepatocytes. 123 A. Total Fatty Acid '° mesh-3 50 33 . o l22.6.n-3 '6 40 E .2- ” m22:5.n-3 o < E 20 020:5.n-3 0 IL 10 I20:4.n-6 o . No Infectlon M-Luc Ad-Elovl-2 M-Elovl-O ‘0 ' B. Non-esterified Fatty Acid 241,.” 3‘ 2 n . I22:B.I‘|.-3 g a, _ _ ‘ y 1111111111111 (_f fl 0220»: II. 10 - l20:4.n»6 0 No lrl‘octbn M-Luc M-Ebvl-Z M—EbVI-S Figure 4. 2. Fatty acids analysis of rat primary hepatocytes overexpressing Elov12 and Elov15. Primary rat hepatocytes were infected with 10pfu/cell adenovirus expressing Elov12, Elov15 and luciferase (control) followed by 250uM 20: 5n3 treatment. Cells were , harvested at 24 hr after 20: 5n3 treatment. A. Total lipids were extracted and saponified ' and analyzed by RP-HPLC. Each fatty acid was expressed as percentage of total fatty acid amount. B: NEFA were separated from total lipid. Each fatty acid was expressed as... 0 percentage of total fatty acid amount. 0 124 To test this hypothesis, hepatocytes were infected with either 10 pfu/cell Ad- Elovl-2, ad-Elovl-S and Ad-acyl CoA synthetase-4 [Ad-ACS4 a control virus] Overnight and treated with 250 0M 20:5,n-3 for 24 hr. Total RNA was isolated and used for - northern analysis to examine the expression of PPARa target gene i.e., cytochrome P450- 4A (CYP4A), cytosolic thioesterase I (CTE-I) and mitochondrial HMG-CoA synthase 2 (HMGCSZ). The efficacy of Ad-ACS4 was indicated by enzyme assay as shown in Fig 4.4 A. The results show that overexpression of Elovl-2 significantly suppressed 20:5 ,n-3 induction on PPARa target genes CYP4A, CTE-I and HMGCS2, but did not affect 20:5,n-3 effects on the expression of mRNASREBp-1 or mRNApr (Fig. 4.3). Overexpression of Elovl-5 has the similar effect as Elovl-2 on PPARa target genes (Data not shown). 4.3.4. Effect of Elov12 and Elov15 on SREBPs nuclear content in rat primary hepatocytes. The nuclear abundance of SREBP-l, but not SREBP-2, is suppressed by PUFA. Of the C18-22 PUFA found in primary hepatocytes, 22:6,n-3 is the most potent suppressor. The mechanism for this control involves 22:6-mediated activation of SREBP- 1 26S proteasomal degradation [159]. Based on the differential effects of PUFA on SREBP-1 content, 1 next determined if changes in elongase activity affected SREBP-l nuclear abundance. The experiment was designed as the gene expression study. Instead of RNA analysis, however, nuclear and microsomal protein extracts were isolated fiom infected primary hepatocytes to measure SREBP-1 and SREBP-2 by immunoblot analysis. Elevated Elovl-2 activity in the absence of exogenous PUFA had no effect on SREBP-1 or SREBP-2 nuclear abundance. Addition of 20:5,n-3, however, enhanced the 125 suppression of SREBP-1 nuclear content, without effects on SREBP-2 nuclear content. Over expressing Elov15 or Elovl6, however, did not alter the 20:5,n-3 effect on SREBP-1 nuclear content (Fig. 4.4). Elovl6 did not utilize 20:5,n-3 as substrate. Although Elov15 converts 20:5,n-3 to 22:5,n-3, Elov15 does not convert 22:5,n-3 to 24:5,n-3. This study suggests that excessive formation of 24:5,n—3 impacts SREBP-1 nuclear abundance. Unfortunately, 24:5,n—3 is not commercially available, thus limiting a direct test of whether 24:5,n-3 or some other action of Elov12 over expression affects SREBP-1 nuclear abundance. We examined possible mechanisms for this effect of Elov12 over expression on 20:5-mediated suppression of SREBP-l nuclear abundance. As shown in Fig.4.3, infection of hepatocytes with Ad—Elov12 did not enhance the effect of 20:5,n-3 on mRNASREBm abundance. 22:6,n-3 is a robust suppressor of hepatocyte SREBP-l nuclear abundance [179]. The mechanism for this suppression involved 22:6-mediated 26S proteasomal degradation of nuclear SREBP-1 [179]. Treating Ad-Elov12 infected hepatocytes with 26S proteasomal inhibitors (MG132 or lactacystin) did not block the enhance 20:5,n-3 suppression of mature SREBP-1 (Data not shown). I next examined the effects of these treatments on proteins involved in proteolytic maturation of SREBP. Analysis of Insig-l and Insig-Z mRNA and SCAP protein revealed mRNA of Insigl and Insig2a were induced by overexpression of Elov12 in the cells treated with 20:5,n-3 compared to Ad-Luc group. There is no change of mRNA of Insin and SCAP by overexpression of Elov12 (Data not shown). However, SCAP protein was suppressed by 126 N N w uh 0| Cb u v u u 7 Enzyme actlvity Fold change to Rat Liver A fir O o 2 5 1o 20 Ad-ACS4 ( PFUIceII) D No Virus 1] ACS-4 I ELOVL-2 0 , , M CYP4A CTE1 HMG CoA SREBP-1c L-PK Synthase Fold Change in mRNA Abundance p: Figure 4. 3. Effect of Elov12 on hepatic gene expression. A. Efficacy of Ad-ACS4 in rat primary hepatocytes. Postrruclear fraction was prepared by differential centrifugation from rat liver and the rat primary hepatocytes infected with different dosage of Ad-ACS4. Fractions (Sug protein) were assayed for ACS activity. B. Primary rat hepatocytes were infected with 10pfu/cell Ad-Elov12 or Ad-ACS4 (control) followed by 250uM 20:5n3 treatment. RNA was harvested at 24 hours after treatment. Northern blot was performed to exam PPARa targeted genes (Cyp4A, CTE-l, HMG CoA Synthase) and SREBP-1c and L-PK gene expression. Mean : SD,n=3. These results are representative of 3 separate studies. 127 20:5,n—3 - + - + nSREBP-1 - nSREBP-2 pSREBP4 pSREBP-Z Figure 4. 4. Overexpression of Elov12 effect on 20:5,n-3 regulation of rat hepatic sterol regulatory element binding protein 1 (SREBP-l) and SREBP-2 nuclear content. Primary hepatocytes were prepared and infected with adenovirus expressing. Elov12, Elov15, Elovl6 and luciferase 24 hours. Followed by treatment of 250 uM 20:5,n- 3 described before. Nuclear and microsomal proteins were isolated from cells after 24 hours fatty acid treatment. Western blot was performed and probed with antibodies fOr SREBP-1 and SREBP-2. 128 overexpression of Elov12 followed by 20:5,n-3 treatment (Fig 4.5 C). Based on the outcome of these studies, elevated expression of Elov12 coupled with the addition of 20:5,n—3 conversion to 24:5,n-3 inhibits the proteolytic maturation of SREBP-1, but not SREPB-2 in rat primary hepatocytes. 4.4. Discussion My previous studies established that certain fatty acid elongases, like fatty acid desaturases, are regulated during development, by nutritional status and by hormones [175,201]. My goal in this study was to determine the effect of altered elongase expression on rat primary hepatocyte lipid metabolism. Since changes in hepatic lipid content are known to control two major transcriptional regulatory networks, 1 determined if altered elongase expression affected these networks, i.e., PPARa and SREBP-1. The outcome of these studies supports the hypothesis that fatty acid elongases affect both hepatocyte lipid content and gene expression. Fatty acid metabolism in the liver is tightly regulated. Fatty acids are oxidized to meet energy needs, or esterified to produce triglycerides, which are either stored in the liver or exported to peripheral tissue by VLDL particles. Two classes of transcription factors, namely, SREBPs and PPARs play a major role in lipid metabolism and energy homeostasis. Our studies suggest that overexpression of hepatic Elov12 or Elov15 can alter fatty acid structure, which will affect the composition of intracellular NEFA. As such, these enzymes contribute to the availability of fatty acid metabolites controlling SREBP-l and PPARa functions. Overexpression of both Elov12 and Elov15 enhance 20:5,n-3 conversion to 22:5 ,n-3 or 24:5,n-3, which leads to reduction of 20:5,n-3 and thus attenuates 20:5,n-3 mediated induction on PPARa regulated transcripts. Moreover, 129 0.05 — 0.04 - T 0.03 - Ad-anl 9 Ari-l I K‘. D -53A 0.02 — I +E°A 0.01 J Relative abndance lnsig1/cyclophilin 0.0035 - 0.003 - 1 0.0025 — 0.002 - El -BDA 0.0015 - . +EpA 0.001 — Ad-Eovl 2 Ad-LUC InsigZa/cyclophilin 0.0005 1 0 Relative abundance 9 O 0 a. 9" at" f. -r’. - + - + _—ll__"A- wwcsc'fl‘r Figure 4. 5. Effect of overexpression of Elov12 on mRNA of Insigs and SCAP protein. Primary rat hepatocytes were infected with 10pfu/cell Ad-Elov12 or Ad-Luc (control) followed by 250uM 20:5n3 treatment. RNA and microsome protein were harvested at 24 hours afier treatment. Real time PCR was performed to exam mRNA of insigl (A), insig2a (B) and SCAP protein level (C). These results are representative of 3 separate studies. 130 overexpression of Elov12, but not Elov12, can enhance 20:5,n-3 mediated suppression of SREBP-1c nuclear content. Over expressed Elov12: In liver, Elov12 is constitutively expressed at low levels. Its substrate preference is limited to €20.22 PUFA. Over expression of Elovl-2 enhanced the capacity of hepatocytes to convert 20:5,n-3 to 22:5,n-3 and 24:5,n-3. Interestingly, this does not lead to elevated 22:6,n-3 formation. The likely explanation for this is the suppression of A6-desaturase activity in cultured rat primary hepatocytes (unpublished observation). Over expression of Elov12 lowers total and non-esterified 20:5,n-3 levels in hepatocytes. A consequence of this is the attenuation of 20:5-mediated induction of PPARa target genes, i.e., CYP4A, CTEl and I-IMGCSZ. A second outcome of our analysis was frnding that over expressed Elov12 significantly enhanced the 20:5-mediated suppression of mature (nuclear) SREBP-1 in hepatocytes. The mechanism for this effect is attributed to effects on proteolytic maturation of SREBP-1. This interpretation is based on the following observations. First, over expressed Elov12 did not enhance 20:5,n-3 suppression of mRNASREBp-1 in primary hepatocyte. Second, 26S proteasomal inhibitors did not block the 20:5,n-3/Ad-Elov12 suppression of mature SREBP-1. Third, over expressed Elov12 suppressed hepatic levels of SCAP and induce INSIG 1 or 2. Taken together, these frnding support the notion that Elov12 alters 20:5,n—3 metabolism in such a fashion to impact SREBP—l maturation to the nuclear form. Over expressed Elov15: Like Elov12, over expression of Elov15 significantly lower hepatic levels of 20:5,n-3 (Fig. 4.1D) and attenuated 20:5,n-3 induction of PPARa target genes. Unlike Elov12, Elov15 does not elongate 20:5,n-3 to 24:5 ,n-3. Elov15 also 131 does not enhance 20:5-mediated suppression of SREBP-l nuclear abundance. Elov15, in contrast to Elov12, is the most abundant elongase in liver; it is also well regulated during post-natal develop, by diet, hormones drugs and toxins. As such, changes in Elov15 activity in these various physiological and pathophysiological states will likely affect hepatic lipid composition. Elov15 effects on gene expression, however, are likely to be not as widespread as those seen with Elov12. Elov15 effects may be restricted to the control of PPARa signaling. More studies will be required to verify this notion. In summary, fatty acid elongases have the capacity to significantly impact hepatic lipid composition. In doing so, elongase-mediated changes in hepatocyte lipid composition affect at least two transcriptional regulatory networks known to be controlled by exogenous fatty acids, viz. PPARa and SREBP-1. Such frndings reveal the importance of fatty acid metabolism on the control of these transcription factors. These studies, however, are in primary hepatocytes. Studies in the next chapter reveal effects of elongase over expression in vivo. These studies reveal even more effects of elongases on hepatic metabolism and gene expression. 132 Chapter 5 Effect of Elov15 on Hepatic Function and Lipid Metabolism in Vivo 5.1. Introduction- Seven fatty acid elongase(Elovl) isoforms, each the product of a separate gene, have been idendified in the mammalian genome; 7 of these isoforms have been cloned and characterized in mammals [115]. Evidence for the importance of Elovls in physiology comes from transgenic mice studies. For examples, Quaking and Jimpy are the two mouse mutant strains have dramatically decreased Elovl-l mRNA level [126]. Elovl-3 is induced in brown adipose tissue following exposure of animals to the cold [116]. The water barrier function was impaired in Elovl-3 null mice. Elovl-6 is induced in transgenic mice over expressing SREBP-1 [24,78,134]. Among these isoforms, there are four expressed in rat liver (Elovl-l, -2, -5 and -6), in which Elovl-5 is the most abundant; Elovl-5 utilizing a broad substrate array, €16-22, involved in PUF A synthesis [24,115,201]. It is induced during postnatal development and suppressed by feeding rats n-3 PUFA enriched diets [175]. Recently, we reported PPARa, high fat diets and obesity affect Elovl-5 expression [201]. Over expressing Elovl-5 in rat primary hepatocytes alters hepatic gene expression, at least in part, by controlling ligand availability for key transcriptional factors, like PPARa. 133 Even though individual Elovl isoforms have different substrate preferences, tissue specific expression, and are regulated differently [115,133,175,201,203], the significance of this diversity is unknown. The regulation of elongase paralleling the change of hepatic fatty acid composition under pathological conditions leads us to test the specific role of individual elongase in hepatic function and onset of chronic disease. Here, I over eXpress Elovl-5 in mouse liver by recombinant adenovirus and test the hypothesis that Elovl-5 will affect hepatic function by manipulating fatty acids structure. 5.2 Materials and Methods Recombinant adenovirus. Cloning of cDNA for Elovl-5 was described previously [201]. The coding region for Elov15 transcript was ligated into Ad-Easy XL adenoviral vector system (Stratagene), recombined in BJ 5183 cells and propagated in XL10 Gold ultra- competent cells. Ad-DNA was packaged into adenoviral particles Ad-293 cells. The resultant adenovirus was amplified in HEK293 cells and purified by cesium chloride gradient centrifuge. Animals. All animal procedures were done in accordance with Michigan State Universtiy Institutional Animal Care and Use committee approved guidelines. Six-week-old male C57BL/6 mice were purchased from Charles River, Kalamazoo and adapted to the environment for 1 week before study. All rrrice were housed in a specific pathogen-free environment with a 12-h light/dark cycle in a temperature-controlled environment. Mice had free access to water and regular chow diet. The adenovirus was intravenously administrated (via the retro-orbital sinus) into 3-4 groups animal: saline (n=8), Ad- LUC(n=8), Ad-Elovl-5(n=8). All treated animals were injected with 2 X 1011 virus particles/animal in a final volume of 200 pl sterile PBS + 1% sucrose; saline injected 134 mice received the PBS + 1% sucrose alone. Mice were given TekLad chow and water, ad lib, and were weighed daily. The 3rd day after injection, all mice were fasted overnight. The next day, 4 mice from each group were then refed with chow diet for 4 hrs before anesthesia. All animals were euthanized at noon — 2 PM on the 4th day post injection. Immunohistology: At euthansia, a sample of liver was taken for histology: a) formalin- fixed, paraffin-embedded for hematoxylin & eosin (H&E) stain and periodic acid-Schiff (PAS) staining to detect glycogen; a second sample was fixed in OCT, fi'ozen (liquid nitrogen/-80°C) and stained with Oil Red 0 (ORO) to detect lipid. Embedding, sectioning, staining and slide preparation was done in the Investigative HistoPatholOgy Laboratory Staff, Department of Physiology-MSU. Blood glucose measurement and plasma preparation: Blood was rapidly drawn from the vena cava (~1 ml) into a heparin syringe. Glucose measurement was performed at the time of blood collection using a hand-held glucose meter (One touch 11, Life Scan Inc.). The remaining blood was centrifuged at 800 rpm for 10min, and the plasma was stored in -80°C for subsequent assays for cholesterol, triglycerides and saponified fatty acid profile. Measurement of liver glycogen content: Liver (~0.1 g) was homogenized in PBS, adjusted to 0.5 M KOH, and incubated at 95°C for 1 hr. The homogenate was cooled, neutralized with 0.1X vol. 6% Nasta; glycogen was precipitated with 3 volumes of ethanol. The precipitate was collected by centrifugation, washed with 7 0% ethanol and resuspended in 1 ml water, adjusted to 50 mM Na-acetate, pH 5.0. Glycogen was hydrolyzed with amyloglucosidase and the released glucose measured using a Wako Glucose assay kit; Standards were glucose (200 mg/dl) and glycogen (1 mg/ml) [204] Absorbance was read in a spectrophotometer (DU 640; Beckrnan) at 340 nm wavelength. ‘ 135 In vitro fatty acid elongation assay. Rat liver microsomes were isolated by differential centrifugation [201]. Elongation reactions were carried out with modifications to the procedure described by Moon et al [117]. Briefly, reaction mixtures contained 50 pg microsomal proteins in a total reaction volume of 100 pl. The reaction constituents are: 50 mM potassium phosphate buffer- pH 6.5, 5 pM rotenone (Sigma), 40 pM fatty acyl CoA (Avanti Polar Lipids, Alibaster, Al and Sigma, St. Louis, MO), 60 pM malonyl CoA (Sigma) [6.5 dpm/pmol] [2-14C]-malonyl CoA (Perkin Elmer), 1 mM NADPH (Sigma) 20 pM BSA (fatty acid free). Reactions (at 37°C) were initiated with the addition of NADPH. When fatty acids were used as substrate,n-aOH neutralized fatty acid (40 pM) replaced fatty acyl CoA. CoASH (at 100 pM), MgC12 (1 mM) andiATP 0(1 mM) were added to the reaction to generate fatty acyl-CoA. Elongase reactions were terminated after 20 minutes with the addition of 100 pl 5 N KOH + 10% methanol; lipids were saponified for 1 hr at 65°C. The saponification reaction was acidified with 100 pl 5 N HCl; 100 pl ethanol was added to aid hexane extraction of fatty acids. Elongated fatty acids were collected by 2 independent extractions with hexane (800 p1). Hexane extracts were pooled and l4C-radioactivity was quantified by B-scintillation counting. ReSults are expressed as Elongase Activity Units,n-moles l4C-malonyl CoA incorporated/mg protein/20 mins. Formation of reaction products was dependent on the presence of NADPH and the fatty acid CoA. Fatty acid elongation products were verified by reverse- phase-HPLC chromatography using a flow-through B-scintillation counter. RNA extraction, northern analysis and quantitative real time-PCR (qRT-PCR). Total RNA was extracted from liver samples and used for template for qRT-PCR analysis as previously described [201]. Specific primers for each gene (Table 5.1) were designed 136 using Primer Express software (Applied Biosystems, Foster City, CA). First strand cDNA was synthesized using the SuperScript II RNase H- Reverse Transcriptase (Invitrogen Carlsbad, CA). Synthesized cDNA was mixed with 2x SYBR Green PCR Master Mix (Applied Biosystems) and various sets of gene-specific forward and reverse primers and subjected to real-time PCR quantification using the ABI PRISM 7700 Sequence Detection System (Applied Biosystems). All reactions were performed in triplicate. The relative amounts of mRN As were calculated by using the comparative CT method (User Bulletin #2, Applied Biosystems). Cyclophilin was used as a control and all results were normalized to the abundance of cyclophilin mRN A. Primers used for quantitative real time PCR are listed in Table 5.1. Lipid extraction and quantitation of hepatic fatty acids composition. Total lipid was extracted from liver in chloroformzmethanol (2: 1) plus 1 mM butylated hydroxytoluene [175]. 7-nonadecenoic acid ( 19:1) was added as a recovery standard at the time of extraction. Protein (Bio-Rad, Hercules, CA) was measured in extracts after the initial homogenization step. Total lipids were saponified, fractionated and quantified by reverse phase HPLC (RP-HPLC) using a YMC J-Sphere (ODS—H80) column and a gradient starting at 77.5% acetonitrile + acetic acid (0.1%) and ending at 100 % acetonitrile + acetic acid (0.1%) over 90 mins with a flow rate of 1.0 ml/min using a Waters 600 controller. Fatty acids were detected using both UV absorbance at 192 nm (Waters model 2487) and evaporative light scatter (Waters model 2420). Fatty acid composition and structures were confirmed at the MSU Mass Spectrometry facility by GC/MS (www.bch.msu.edu/facilities/massspec/index.html). Fatty acid standards for RP-HPLC were obtained from Nu-Chek Prep (Elysian, MN). 137 Table 5. l.Primer sequences used for gene expression by quantitative reverse transcription-PCR for mouse. Gene Forward Reverse ACC TGTCCGCACTGACTGTAACC ATTTCCATAGCCGACTTCCA F AS CAAATACAATGGCACCCTGA TGGCGAAGCCGTAGTTAGTT SREBP-1 AGGAGAACCTGACCCTACGA GGTAAGCGTCTCCACCACTT CYP4A10 TGTTTGACCCTTCCAGGTTT CAATCACCTI‘CAGCTCACTCA HMGCSZ CCTTGAACGAGTGGATGAGA CAGATGCTGTTTGGGTAGCA AOX-1 CCCAACTGTGACTTCCATCA ACGGATAGGGACAACAAAGG L-PK AGGAGTCTTCCCCTTGCTCT ACCTGTCACCACAATCACCA PEPCK AC ATTGCCTGGATGAAGTTTG GGCA'I'I'I‘GGATI‘TGTCTTCAC GLUT2 GAGGAAGTCAGGGCAAAGAA AAGAGCTGGATCACGGAGAC G6PC TCGTGGCTGGAGTCTTGTC GCGAAACCAAACAAGAAGATG GCK GAGATGGATGTGGTGGCAAT ACCAGCTCCACATTCTGCAT CHREBP CTGGGGACCTAAACAGGAGC GAAGCCACCCTATAGCTCCC A9 D TCAACTTCACCACGTTCTTCA CTCCCGTCTCCAGTTCTCTT A5 D TGTGTGGGTGACACAGATGA GTTGAAGGCTGATTGGTGAA A6 D CCACCGACATTTCCAACAC GGGCAGGTATTTCAGCTTCTT ELOVLS GGTGGCTGTTCTTCCAGATT CCCTTCAGGTGGTCTTTCC ELOVLZ ACGCTGGTCATCCTGTTCTT GCCACAATTAAGTGGGCTTT CYCLOPHILIN CTTCTTGCTGGTCTTGCCATTCCT GGATGGCAAGCATGTGGTCTTTG 138 Immunoblotting. Liver rrricrosomal and nuclear extracts were prepared as described previously [159,175]. Proteins (50-100 pg) extracted fi'om microsomal or nuclear fractions were separated electrophoretically by SDS-polyacrylamide gel electrophoresis (NuPAGE 4-12% polyacrylamide Bis-Tris, Invitrogen) and transferred to nitrocellulose membranes. Membranes were incubated with antibodies for ChREBP (Novus Biologicals, Littleton, CO); MLX(N-l7), SREBP-l (IgG-2A4, sc-l3551), SREBP-2, ERKl (so-94), and phosphor-ERK (SC-7383) (Santa Cruz Biotechnology, San Cruz, CA); GSKBB, phosphor-GSKBB (Ser9) and caspase 9 (Cell signaling technology, Beverly, MA). Anti- mouse, anti-goat and anti-rabbit secondary antibodies were obtained fiom Santa Cruz Biotechnology (Santa Cruz, CA). The SuperSignal West Pico chemiluminescence kit (Pierce) detection system was used. Statistical Analysis. Statistical analysis used Student’s t-test and AN OVA plus post hoc Tukey HSD test (htg)://facultv.vassar.edu/louryNassarStats.html). 5.3 Result 5.3.1. Generation of the mice overexpressing Elov15 in liver. C57BL/6 mice were injected with saline, Ad-Luc (control virus) and Ad-Elovl-S, respectively. Liver and kidney RNA were examined for Elovl-5 gene expression in these mice. Elovl-5 mRNA level was elevated ~30 fold compared to Ad-luciferase and saline groups (data not shown) in mouse liver. There is no significant difference of Elovl-5 expression in kidney, heart and spleen among the 3 treatments (data not shown). This outcome indicated that the adenovirus were sequestered by the liver, not other tissues. 139 N o I D Ad-Luc I Ad-EIovl—5 A (J! i Elongase Activity (nmoles/mg protein) or 8 16:0-COA 18:3-00A 20:4-COA Elongase Substrate Figure 5. 1. Effect of overexpressing Elovls on elongase activity in mice liver. Male C57BL/6 mice were injected with Ad—Luc and Ad-Elov15 and fed with chow diet for 4 days as described in (Materials and Methods). Hepatic rrricrosomal preparations were used for fatty acid elongase assays (Materials and Methods). Results are expressed as Elongase Activity, (nmoles 14C-malonyl CoA assimilated into fatty acids/mg protein). Mean 1: SD,n=8. *p50.01 vs Ad-Luc, Student’s T -T est. 140 Liver elongase assay showed that enzyme activity of Elovl-5 was increased by 2-3 fold as compared to Ad-Luc and saline groups ( Fig. 5.1). This level of Elovl-5 activity is ob/ob seen in animals fed the peroxisome proliferator, WY14643, or in livers of Lep mice [26,205]. 5.3.2. Metabolic characterization of the mice overexpressing Elov15 in liver. On a normal chow diet, all Ad injected mice lose body weight and have decreased food intake in the first day afier injection compared to the saline group (data not shown). Two days after injection, food intake resumes and body weight increases. By day 4, post injection, body weight and food intake between the saline and Ad-infected groups is not significantly different. All the Ad infected animals developed a modest, but insignificant increase in liver weight when compared to the saline group (data not shown). Livers of the Ad-Elovl-S group have small white lesions compared to Ad-Luc infected groups (Fig. 5.2 A). Surprisingly, the blood glucose measured at the time of euthansia decreased 30%- 40% in the Ad-Elovl-5 groups compared to the control group (Ad-luc and saline groups) in both fasting and refeeding state (Fig. 5.3 A). Hepatic glycogen stores, determined by Periodic acid shifi (PAS) staining, was significantly decreased in Ad-Elovl-S groups compared to Ad-Luc group (Fig. 5.2 B). Direct measure of hepatic glycogen content (Methods) confirmed the PAS staining results (Fig. 5.3 B). The fold change of hepatic glycogen levels during fast and refeeding of the Ad-Elovl-S infected mice was decreased 4 fold compared to Ad-Luc group. Hepatic lipid content, determined by oil red O staining (ORO), mirrored the quantitative hepatic triglyceride in all Ad groups (Fig. 5.2 B); Ad- 141 A. Ad-Luc Ad-Elovl5 B- Ad-Luc Ad-Elovl5 Figure 5. 2. Liver morphology and histology for Ad mice. A. Liver morphology of Ad injected mice. B. Liver histology of Ad mice. Liver sections from the mice overexpressing Luc and Elov15 were stained with hepatoxylin and eosin (H&E) and oil red O (ORO) and (PAS), magnification 20X. Arrows indicated necrosis in Ad-Elov15 liver. Sections are representive of several animals for each condition. Image in this figure is presented in color. 142 Elov15 had not major effect on hepatic neutral lipid content. Blood triglyceride levels, however, were significantly decreased in the fed state of Ad-Elovl-S infected animal. (Fig. 5.3 C). No change of blood cholesterol level was observed in Ad-Elovl-S injected mice (Data not shown). 5.3.3. Alteration of glucose metabolism in Ad-ElovlS injected mice. Stores of glycogen in the liver are considered the main buffer of blood glucose levels. Since glycogen level is suppressed in Ad-Elovl-S injected mice, we first hypothesized that overexpression of Elovl-5 altered hepatic glucose metabolism. Fasting induces glycogenolysis and gluconeogenesis, whereas refeeding induces glycolysis and glycogenesis. We first examined the mRNA expression and nuclear content of ChREBP, which is required for most glucose-regulated hepatic genes [55,62]. Results showed that mRNAChREBp was decreased in the refed group overexpressing Elovl-5, but not in Ad-Luc group (Fig. 5.4 A). Immunoblotting of ChREBP and MLX in fed mice showed a decline in nuclear ChREBP, but no change of nuclear MLX, the ChREBP heterodimer partner. (Fig. 5.4 B). L-PK, a target of ChREBP/MLX and a major glycolytic enzyme, was significantly suppressed in livers of Ad-ElovlS infected animals when compared to Ad- Luc infected animals (Fig. 5.5). A second ChREBP/MLX target gene was examined. Glut2 is a key hepatic glucose transporter. Like L-PK, Glut2 expression was also suppressed in Ad-Elovl-5 infected animals in both the fasted and fed state. The gluconeogenic enzymes, phosphoenolpyruvate carboxykinase (Peka) and glucose-6- phosphatase (G6Pase) were examined. Elovl-5 significantly suppressed mRNApcka and mRNAmsmc in both fasting and refeeding (Fig. 5.5). 143 Ad-Elov 15 Ad-Luc _ a _ _ ‘ 086420 1 12- 2:. .onooao coo-m 3A 3B 0 3 A 5 2 _ 0 2 4 5 1 A 0 1 Ll 5 2.3.50 £5320 0:30... 09.2.0 200. 0 Ad-Elov15 Ad-Luc 144 c» O __| 1 t—i Ad-Luc Ad-ElovlS N O O 1 A U" D 1 Triglyceride, mg/dl in" 8 O Figure. 5.3. Effect of overexpressing Elov15 on blood glucose, triglyceride and hepatic glycogen level. A. Blood glucose level was measured by blood glucose meter (One touch 11, Life Scan Inc.). The open bar represents the fast group; the filled bar represents the refeeding group; Mean j; SD,n=4. #P<0.05 vs fast group, anova. B. Liver glycogen level was measure as described in the method. Results are expressed as Fold Change (refeeding/fast), mean 1 SD, n=4. C. Effect of overexpressing Elov15 on blood triglyceride level. Blood triglyceride level was measured by Wako L—Type TG H test kit. The open bar represents the fast group; the filled bar represents the refeeding group; Mean :1; SD,n=4. #P<.01,##P<.05. 145 Glycogen synthase kinase-30 (Gsk-3B) is a serine/threonine kinase that plays a key role in controlling glycogen synthesis by regulating the phosphorylation status of glycogen synthase [206]. Gsk-3B is a highly-regulated kinase; its activity is controlled by its phosphorylation status. Insulin suppresses Gsk-3B activity by inducing its phosphorylation through the PIBK-Akt pathway [207]. Inhibition of GSK3|3 releases glycogen synthase from inhibition and stimulate glycogen synthesis. I examined total and phospho-Gsk3B in livers of fed Ad-infected animals only. Contrary to my expectation, the phosphorylation status of Gsk-3B (ser-9) was significantly increased in Ad-Elovl-5 injected when compared to the saline or Ad-Luc infected groups (Fig. 5.6). As such, Elovl-5 over expression does not suppress hepatic glycogen content by activating Gsk-3B. I also examined a second target of insulin action, Erk. Insulin rapidly, but transiently induces Erk phosphorylation in rat primary hepatocytes [83]. Over expressed Elovl-5 increased phospho-Erk with little effect on total Erk (Fig. 5.6). No adenovirus infection increased the formation of caspase 9 (37.5 kd), an indicator of intrinsic apoptosis [208](Fig. 5.6). Taken together, these results indicate that elevated hepatic activity of Elovl-5 mimics some, but not all of the effects of insulin: a) induces Gsk-3B phosphorylation; b) induces Erk phosphorylation; c) suppresses Peka gene expression. Unfortunately, however, there are contradictory outcomes; such as the suppression of hepatic glycogen content and expression levels for ChREBP, L-PK and Glut-2, with little effect on glucokinase. While my studies do not provide a complete explanation for the effects of over expressed Elovl-5 on hepatic glucose metabolism, these studies provide clear evidence that change in fatty acid elongase activity, alone, are sufficient to promote 146 0.3 7 0.25 J 0.2 — 0.15 — 0.1 — 0.05 — [3 fast I refed relative abundance, ChREBP/cyclophilin Ad- Luc Ad-Elovi-5 Figure. 5.4. Effect of overexpression of Elov15 on hepatic ChREBP gene expression and nuclear content. A. Effect of overexpression of Elov15 on hepatic ChREBP gene expression. RNA was extracted and used for qRT-PCR analysis of ChREBP gene. Results are expressed as Fold Change (transcript/cyclophilin), mean 1 SD, n=4. B. Effect of overexpression of Ad-ElovlS on nuclear ChREBP and MLX abundance. Livers from Ad-Luc and Ad-ElovlS injected mice were used for the isolation of nuclear proteins for immunoblotting. Duplicate samples for each treatment are shown. 147 major changes in hepatic glucose metabolism and key signaling pathways that impact hepatic glucose metabolism. 5.3.4. Decreased expression of genes involved in lipid metabolism in Ad- ElovlS injected mice. Lipid metabolism is a second major hepatic metabolic pathway that is affected by dietary lipid composition. Herein, I examined changes in hepatic expression of genes involved in hepatic lipid metabolism. Elevated expression of Elov15 (Ad-Elov15) in fasted mice suppressed expression of PPARa target genes, CYP4A and HMGCSZ levels, but not AOX-1, by 70% (Fig. 5.7). There was no change in SREBP-1 gene expression, SREBP-1 nuclear content or SREBPl-target genes, ACC and FAS, in Ad-ElovlS injected mice comparing Ad-Luc injected mice (Data not shown). However, these three genes responded nicely to the fast-refeeding. In addition, mRNA levels for SCD-l, the desaturase responsible for the production of monounsaturated fatty acids was significantly suppressed in liver of Ad-ElovlS injected mice compared with Ad-Luc injected mice in refed states (Fig. 5.7). SCDl expression is controlled by PPARa, SREBP-l and ChREBP/MLX [205]. The decline in SCDl expression in Ad-ElovlS infected animals is likely due to disruption of the PPARa and ChREBP/MLX regulatory pathways. 148 Wlin LPK/cyclophilin relative abundance 149 §§ 1 99.0.0 88 N L GBPC/cyclophilin § relative abundance O .1 Ad-Luc Ad-Elovl5 0.35 - .0 N or relative abundance PEPCK/cyclophilin o o 4: # 005 ~ 0 . l L Ad-Luc Ad—Elovl-5 Figure. 5.5. Effect of overexpressing Elov15 on gene expression involved in glucose metabolism. RNA was extracted and used for qRT-PCR analysis of gene involved in glucose metabolism. Results are expressed as Fold Change (transcript/cyclophilin), mean 1 SD, n=4. #P<0.01. 150 5.3.5. Effect of Elov15 overexpression in hepatic and plasma fatty acid profile. Elov15 utilizes a broad array of fatty acid substrates, €16.20, to impact both MUFA and PUFA synthesis. I hypothesized that overexpression of Elov15 can promote endogenous PUFA synthesis. To eliminate the contribution of dietary lipids from the absorptive phase, I examined hepatic and plasma fatty acid of fasted mice. The outcome of these studies provided unexpected results. The ratio of 20:4,n-6 to 18:2,n-6 is decreased in Ad-ElovlS injected mice (Fig. 5.8). 18:2,n-6, an essential fatty acid, is converted to 20:4,n-6 by elongation (Elov15) and desaturation (A5 D and A°D). There was no increase in 22:4,n-6 in livers of Ad-ElovlS infected animals. Since other enzymes involved in PUFA synthesis pathway, i.e., ASD and A°D desaturase, did not change, I speculate that over expressed Elov15 stimulated the conversion of 20:4, n-6 to 22:4,n-6 and its peroxisomal degradation. PUFA of 322 carbons are preferred substrates for peroxisomal oxidation [209]. Two interesting observations were made in my analysis of the plasma fatty acid profile. Mice receiving Ad-ElovlS had a higher (by 40%) level of PUFA in the plasma (Fig. 5.9). This is due to the accumulation of 18:2,n-6 in the plasma. Second, the level of 18:0, relative to 18:1, was increased (Fig. 5.9). 5.4. Discussion The data from these in vivo studies support the hypothesis of this thesis. Altered expression of an elongase, alone, is sufficient to change hepatic gene expression. The outcome of these studies, however, revealed a more complicated change in hepatic 151 Saline Ad-LUC l Ad-ElOVl-5 stk-3 B Gsk-3 B pErk1/2 Erkl/2 Figure. 5.6 Effect of overexpression of Elov15 on regulating GSK30 and ERK. Total protein was extracted from liver as methods described. Protein levels were measured by immunoblot (Materials and Methods). Duplicate samples for each treatment are shown. 152 metabolism affecting both glucose and lipid metabolism. As such, these studies may have revealed novel linkages between fatty acid elongation and other facets of hepatic metabolism. Several reports in the literature have demonstrated that infection of adenovirus into the retro-orbital sinus of mice results in >99% of the virus infecting liver [210,21 1], and as expected, we did not observe Elov15 expression in other tissues, including kidney, heart, or spleen (data not shown). Overexpression of Elov15 in mouse liver, by the recombinant adenoviral approach, elevated elongase activity 3-fold when compared to liver of animals infected with the Ad-Luc control or control (saline injected) animals. °°/°° obese mice This level of expression is seen in other physiological states, e.g., Lep [205], TCDD-treated mice (Fig 3.11 A) and WY14,643 treated rats [26]. As such, the change in elongase activity is within a physiological range. Several key observations were made. First, there was ~40% decrease in blood glucose level in fasted and fed Ad-ElovlS infected mice. The likely explanation for this is the decline in hepatic glycogen content and expression of genes involved in gluconeogenesis (PEPCK and Glc-6-P), coupled with a decline in genes involved in glucose transport (Glut2) and glycolysis (LPK). The outcome of these results implicated some change in hepatic insulin signaling. Analysis of two downstream targets of insulin action, namely, Gsk3B and Erk indicated that elevated Elovl-5 activity as associated with increased phosphorylation of these proteins. This same effect is observed in acute treatment of hepatocytes with insulin [83]. As such, elevated Elov15 activity appears to mimic insulin action. Defects in liver and muscle glycogen synthesis are major factors contributing to postprandrial hyperglycemia in patients with type 2 diabetes [212]. Our 153 relative abundance CYP4A/cyclophilin O 00 l 0399 ”01008 [111 0.15 l 0.1 + SOD/cyclophilin relatlve abundance P or? as SCD-l. Ad-Luc 154 Results are it ,i Figure. 5.7. Effect of overexpressing Elov15 on PPARa target gene expression. RNA was extracted and used for qRT-PCR analysis of PPARa target gene, Cyp4A and HMGCSZ as well (transcript/cyclophilin), mean 1 SD, n=4. #p<.01, *p<.05. expressed as Fold Change 80 — 70 — as 60 _ l18:1,n-9 ° 4 §_ 50 B Stearate E 40 — to E 16:1,n-7 i? 30 — . E E1 Palmitate 20 — 10 ~ 0 t . Ad-Luc Aid-Elov15 45 _ I 22.6,n'3 40 _ I 22:5,n—3 35 _ n 22:4,n—6 g: 30 — I 20:5,03 5. 25 — 1:1 20:4,n-6 E 8 20 l I 20:3,n—9 r? f 15 — I 20:3,n—6 10 - 1:118:3,n-6 5 ' I 18:3,n—3 0 . . D18:2,n—6 Ad-Luc Ad-Elovl5 Figure. 5.8. Effect of overexpressing Elov15 on hepatic lipid composition in the fasted mice. Total lipids were extracted and saponified; fatty acid levels were quantified by RP-HPLC (Materials and Methods). Results are expressed as Fatty Acid Mole%, mean 1 SD, n=4/group. 155 80- 7o — 60 — .\° I 18:1,n-9 g 50 — 'o‘ I Stearate .g 40 _ I 16:1,n-7 g 30 — u_ 1: Palmitate 20 — 1o — O l l Ad-Luc Ad-Elovl5 4o - I22:6.n-3 T 35 - ' f l22:5.n—3 30 — U22:4,n—6 =5 . . 5° 25 _ .1 ,' I20.5,n-3 - D20:4,n—6 ‘7: 20 - I20:3.n-9 E 15 ~ 1.1. I20:3.n—6 1o — U18:3,n—6 5 _ I18:3,n—3 0 w [11812.n-6 Ad-Luc Ad-Elovl5 %— Figure. 5.9. Effect of overexpressing Elov15 on plasma lipid composition in the fasted mice. Total lipids were extracted and saponified; fatty acid levels were quantified by RP-HPLC (Materials and Methods). Results are expressed as Fatty Acid Mole%, mean : SD, n=4/ group. 156 results suggest that elevated Elov15 lowers blood glucose levels. We observed in high fat- fed animals, both glucose-intolerance and lower hepatic Elov15 expression [205]. Perhaps alterations in hepatic Elov15 levels might improve blood glucose levels in high-fat-fed mice. More analysis will be required to fully understand the underlying mechanism(s) by which elevated Elov15 impacts hepatic glucose metabolism. A second key observation is the effect of elevated hepatic Elov15 activity on hepatic and plasma lipid composition. While overexpressed Elov15 did not significantly impact SREBP-1, and its target genes F AS and ACC during fast and refeeding, over expressed Elov15 did impact the expression of several PPARa target genes, CYP4A and HMGCS2. The effect of over expressed Elov15 was most apparent in the fasted state. Although total hepatic lipid content did not change, the type of fatty acids seen in V liver and plasma did change. In liver, overexpressed Elov15 resulted in a reduction of 20- 22 carbon PUF A. We speculate this is due to enhanced degradation in the peroxisome. The oxidation reaction of fatty acyl-CoA is the initial and rate-limiting step of the peroxisomal beta-oxidation pathway. AOXs and pristanoyl-CoA oxidase (PCOX) can oxidize the CoA esters of straight-chain fatty acids. Interestingly, the expression of AOX- 1, which is responsible for peroxisomal B-oxidation, was not affected by Elovl-5 overexpression. However, we need additional data on how overexpression of Elov15 affects other subtypes of AOX and PCOX. Hepatic content of 18:1,n-9 increased in livers of mice receiving Ad-ElovlS. 18:1 is the preferred substrate for triglyceride and cholesterol ester synthesis [27]. The ratio of 18:1 to 18:0 has been implicated in the regulation of cell growth and differentiation through effects on membrane fluidity and 157 signal transduction [10]. With this outcome, 1 expected to see an elevation in plasma triglyceride content. Overexpressed Elov15 had three effects on plasma lipids; a) decreased triglyceride levels; b) decreased ratio of 18:1,n-9 to 18:0; and c) change in PUFA composition, mainly due to the accumulation of 18:2,n-6. Since 18:2,n-6 is only available from the diet, we speculate the accumulation of 18:2,n-6 in plasma is due to the decreased uptake of this fatty acid by liver or the increased efflux of 18:2,n-6 from the liver if the uptakcd 18:2,n-6 is not converted to its CoA form rapidly. While my studies do not fully explain the mechanisms for these observations, they point out that change in hepatic lipid metabolism lead to changes in blood lipid composition. In studies not shown here, animals infected with Ad-Elov12 show a remarkable decline in hepatic 20:4,n-6 levels. Whereas, in animals infected with Ad-A°-desaturase show a significant increase in plasma 20:4,n-6 levels (data not shown). Taken together, these studies highlight the fact that hepatic PUF A synthesis has a major effect on blood lipids and likely will significantly impact lipid composition in other tissues. Since both fatty acid and glucose metabolism are altered by Elov15 overexpression, this led us to question how the early disruption of the fatty acid balance in the liver or plasma may influence subsequent abnormalities in glucose metabolism. Skeletal muscle also plays a major role in the development of metabolic disorders, through the regulation of insulin-induced glucose disposal. The increased PUF A amoumt in plasma may change the fatty acid composition in the membrane of skeletal muscle, which may modify insulin signaling pathway in skeletal muscle to impact blood glucose. More studies are needed to test this hypothesis. 158 In conclusion, the liver plays a central role in the regulation of whole body lipid and glucose homeostasis. My findings indicate that overexpression of Elov15, alone, can alter hepatic function which leads to changes in whole body and hepatic glucose and lipid metabolism. These results strongly suggest that fatty acid elongases play an important role in management of glucose and fatty acid metabolism. Whether control of hepatic Elov15 activity can be used to impact blood glucose and triglyceride in metabolic diseases remains an important unanswered question. 159 Chapter 6 Conclusions and Future Directions In the last two decades, a growing number of articles emphasized the role of fatty acid in the control of gene expression. Fatty acids can directly bind to and regulate the activity of some transcriptional factors, such as, PPAR family [51,53]. In this case, fatty acids act like hydrophobic hormones to control the activity or abundance of key transcriptional factors. In contrast, fatty acids also regulate other transcriptional factors through indirect mechanism by changing their nuclear abundance and activity, such as SREBP-1c [14]. Whatever mechanisms are involved in fatty acid regulation of gene expression, the structure of fatty acid is the key determinant for the regulation of transcriptional factors [14,53,159]. During fatty acid metabolism, several steps are involved in the modification of fatty acid structure, including elongation/desaturation, 0 oxidation (mitochondrial or peroxisomal), or assimilation into complex lipids [14]. This suggests the enMes in these metabolic pathways may influence gene expression by modifying fatty acid structures. My studies focus on the enzyme involved in the fatty acid elongation, which are called fatty acid elongase. Four fatty acid elongases (Elovll, 2, 5 and 6) and 3 desaturases (A5D, A6D and A9D) are expressed in liver. Among these enzymes, the desaturases are well studied, especially A9D. These enzymes are regulated by hormones, development and dietary lipids [5-10]. A9D knockout mice have shown low possibility to develop hepatic steatosis and improved insulin sensitivity in muscle [1 1-13]. As such, A9D had been recognized as 160 drug target for obesity treatment recently. Compared to the desaturases, we are still in the early stage of understanding how fatty acid elongases are regulated and what their roles are in physiology. My studies filled this gap with new information. In the first part of these studies, I examined the nutritional and developmental regulation of rat hepatic Elovl expression and correlated changes in elongase expression with elongase enzymatic activity. Of the four elongase subtypes expressed in rat liver, Elov15 is the most abundant elongase transcript. Rat hepatic Elov15 expression is regulated at the pretranslational level by dietary n3 PUFAs and PPARa agonist and during postnatal development. These same factors regulate hepatic Elovl activity. Changes in the expression of the elongases (and their activity) and desaturases are sufficient to induce changes in tissue and plasma levels of specific fatty acids (e.g., 20:3,n-9). Unlike A5, A°, and A9 desaturases, the elongases do not display a uniform response to fasting-refeeding, fish oil, or PPARa agonist. In this regard, changes in nuclear SREBP-l levels correlate with changes in Elovl-6 expression but not other elongases. Although PPARa agonists induce all desaturases and three elongases (Elovll, 5, and 6), the mechanism for this control remains unresolved. Next, we further show the evidence that Elov15 and Elovl6 are regulated in liver by nutrients (glucose and fat), hormones (insulin) and nuclear receptor agonist, i.e., LXR and PPARa agonist. From these studies, we conclude that ChREBP, MLX, SREBP-1, PPARa and LXR control elongase, as well as desaturase expression. Metabolic diseases, like diabetes and obesity, induce changes in hepatic lipid composition by controlling the function of key transcription factors that impact elongase and desaturase expression. These studies support the notion that regulation of fatty acid 161 elongase, as well as desaturases, may play an important role in managing hepatic lipid composition in response to changes in dietary and hormonal status. Individual Elovl isoforms have different substrate preferences, tissue specific expression, and are regulated differently [115,133,175,201,203]. The significance of this diversity is unknown. To get more information about the role of individual hepatic elongases, we amplify the individual elongase activity in hepatocytes and liver by recombinant adenovirus technology. The recombinant adenovirus approach elevated elongase expression and activity to 3 to 5 folds in rat primary hepatocytes. In addition, overexpression of Elov12 and Elov15 can suppress 20:5,n-3 PPARa regulated gene expression. I speculate this is due to the suppression of hepatocyte 20:5,n-3 levels and accumulation of 22:5,n-3 or 24:5,n—3. 22:5,n-3, and possibly 24:5,n-3, are weak activators of PPARa. Moreover, the overexpression of Elov12, but not Elov15, can enhance 20:5 ,n- 3 suppression of nSREBP-l content without affecting nSREBP-Z. This strongly supports the notion that fatty elongase have the capacity to alter gene expression by controlling cellular content of fatty acids that impact transcription factor activity or nuclear abundance. To broaden our view about the function of individual elongases in hepatic function and whole body lipid metabolism, 1 investigated the effect of overexpression of Elov15 in mice in vivo. Elov15 is the most abundant elongase expressed in rat liver. Moveover, it has broad substrate specificity. Overexpression of Elov15 in mouse liver was achieved by use of adenoviral intravenous transfer to obtain liver-specific overexpression. Significant reduction in blood glucose in both fasting and refeeding as well as deficiency of glycogen storage during refeeding in Elov15 overexpressed mice suggests Elov15 play 162 an important role in glucose homeostasis. The suppressed gene expression of PEPCK during fasting can partly explain this observation; it suggests that gluconeogenesis is suppressed. Effects on GSK30 and ERK phosphorylation status were observed by overexpressing Elovl-5. These results mimic insulin effects on these signaling pathways. Overexpression of Elov15 also significantly reduced PPAR 0. target gene expression while having no significant effect on SREBP-l and its targets genes. Moreover, overexpression of Elov15 alters the fatty acid composition both in liver and in plasma. Most important, overexpression of Elov15 decreased blood triglyceride level during refeeding. Taken together, these findings suggest that Elov15 is also an important factor controlling both hepatic and whole body lipid metabolism. In summary, this thesis provides novel information about hepatic fatty acid elongases, in particular Elov15. The results indicate that Elov15 plays an important role in hepatic function as well as whole body glucose and lipid metabolism. Abnormal expression of Elov15 in pathological conditions can account for the fatty acid profile in both liver and plasma. These studies also suggest Elov15 may be the potential target for treating common diseases such as hypertriglyceridemia, obesity, and insulin-resistant diabetes. In future, the use of microarray methods will help expand the effects of Elov15 overexpressed on liver function and may provide new clues to identify specific molecular events/pathways influenced by Elov15. Moreover, modulating Elov15 expression via siRNA knock down and adenoviral transduction will let us understand better and comprehensively the role of Elov15 plays in glucose and lipid metabolism. 163 To investigate the role of Elov15 on the onset of chronic disease, we can overexpress or knockdown Elov15 in diabetic or other pathological animal models, such as the Zucker rat, Lep°°/°° mouse or the high fat-fed C57BL/6 (glucose intolerant) mouse, to test if changes in Elov15 influence the onset and progression of metabolic disease. We can also use the same methods to test special role of other hepatic elongases. By this, we will have comprehensive understanding about fatty acid elongase family. The discovery of the effect of fatty acid elongase on glucose homeostasis may identify novel pathways to combat diabetes, benefiting millions of such patients worldwide. 164 APPENDIX PUBLICATIONS Journal Articles 1. Wang Y, Botolin D, Xu J, Christian B, Mitchell E, J ayaprakasam B,n-air M, Peters J, Busik J, Olson LK, Jump DB. Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and obesity. J Lipid Res. 47: 2028-2041 (2006) Botolin D, Wang Y, Christian B, Jump DB. Docosahexaneoic acid (22:6,n-3) regulates rat hepatocyte SREBP-1 nuclear abundance by Erk- and 26S proteasome-dependent pathways. J Lipid Res. 2006 Jan;47: 1 81-92 (2005). Jump, D.B.; Botolin, D.; Wang, Y.; Xu, J .; Christian, B. & Demeure, 0. Fatty Acid regulation of hepatic gene transcription. J Nutr, 2005, 135, 2503-2506 Wang Y, Botolin D, Christian B, Busik J, Xu J, Jump DB. Tissue- specific,nutritional, and developmental regulation of rat fatty acid elongases. J Lipid Res. 46:706—15 (2005). Wang Y, Botolin D, Christian B, Jump DB. Role of fatty acid elongases in , hepatic lipid and glucose metabolism. Manuscript in preparation for cell metabolism. Conference Abstracts 1. Regulation of rat hepatic elongase and desaturase gene expression. Yun Wang, Botolin D, Barbara Christian and Donald B. Jump.Regulation of hepatic elongase and desaturase gene expression. Poster presentation, EB 2006, San Francisco, California The impact of hepatic metabolism on fatty acid-regulated PPARa activity and SEEBP-l nuclear content. D.B. Jump and Y. Wang. FASEB Summer Conference: Nutrient Control of Gene Expression, July 30-Aug. 5, 2005, Tuscon, AZ . Gene regulation in response to polyunsaturated fatty acids. D.B. Jump and Y.Wang, and D. Botolin, J. Xu, 0. Demeure, J. Busik, W. Chen and B. Christian. International Conference on the Bioscience of Lipids, September 20-24, 2005 in Ajaccio, Corsica. 165 4. The impact of hepatic metabolism on fatty acid-regulated PPARa activity. D.B. Jump and Y. Wang. Fatty acids and cell signaling conference, Paris, France, Sept. 28, 2005. 166 [11 [21 [3] [4] l5] l6] [7] [8] [91 1101 [11] BIBLIOGRAPHY Jump, D.B., Clarke, S.D., Thelen, A. and Liirnatta, M. (1994) Coordinate regulation of glycolytic and lipogenic gene expression by polyunsaturated fatty acids. J Lipid Res 35, 1076-84. Jump, D.B., Clarke, S.D., MacDougald, O. and Thelen, A. (1993) Polyunsaturated fatty acids inhibit S 14 gene transcription in rat liver and cultured hepatocytes. Proc Natl Acad Sci U S A 90, 8454-8. MacDougald, O.A., Clarke, SD. and Jump, DB. (1992) Tissue specificity of $14 and fatty acid synthase in vitro transcription. Biochem Biophys Res Commun 182, 631-7. Armstrong, M.K., Blake, WI. and Clarke, SD. (1991) Arachidonic acid suppression of fatty acid synthase gene expression in cultured rat hepatocytes. Biochem Biophys Res Commun 177, 1056-61. Nakamura, M.T., Cho, HP. and Clarke, SD. (2000) Regulation of hepatic delta-6 desaturase expression and its role in the polyunsaturated fatty acid inhibition of fatty acid synthase gene expression in mice. J Nutr 130, 1561-5. Cho, H.P., Nakamura, M. and Clarke, SD. (1999) Cloning, expression, and fatty acid regulation of the human delta-5 desaturase. J Biol Chem 274, 37335-9. Cho, H.P., Nakamura, MT and Clarke, SD. (1999) Cloning, expression, and nutritional regulation of the mammalian Delta-6 desaturase. J Biol Chem 274, 471-7. Tang, C., Cho, H.P., Nakamura, MT. and Clarke, SD. (2003) Regulation of human delta-6 desaturase gene transcription: identification of a functional direct repeat-l element. J Lipid Res 44, 686-95. Ntambi, J .M. and Miyazaki, M. (2004) Regulation of stearoyl-CoA desaturases and role in metabolism. Prog Lipid Res 43, 91-104. Ntambi, J .M. (1999) Regulation of stearoyl-CoA desaturase by polyunsaturated fatty acids and cholesterol. J Lipid Res 40, 1549-58. Dobrzyn, A. and Ntambi, J .M. (2005) Stearoyl-CoA desaturase as a new drug target for obesity treatment. Obes Rev 6, 169-74. 167 [12] [13] [14] [15] [16] [17] [18] [191 [20] 121] [22] [23] [24] [25] Dobrzyn, A. and Ntambi, J .M. (2005) The role of stearoyl-CoA desaturase in the control of metabolism. Prostaglandins Leukot Essent Fatty Acids 73, 35-41. Dobrzyn, A. and Ntambi, J .M. (2004) The role of stearoyl-CoA desaturase in body weight regulation. Trends Cardiovasc Med 14, 77-81. Jrunp, DB. (2004) Fatty acid regulation of gene transcription. Crit Rev Clin Lab Sci 41, 41-78. Das, UN. (2006) Essential fatty acids: biochemistry, physiology and pathology. Biotechnol J 1, 420-39. Duplus, E., Glorian, M. and Forest, C. (2000) Fatty acid regulation of gene transcription. J Biol Chem 275, 30749-52. Jump, DB. (2002) Dietary polyunsaturated fatty acids and regulation of gene transcription. Curr Opin Lipidol 13, 155-64. Coleman, R.A., Lewin, T.M., Van Horn, CG. and Gonzalez-Baro, MR. (2002) Do long-chain acyl-CoA synthetases regulate fatty acid entry into synthetic versus degradative pathways? J Nutr 132, 2123-6. Kusakabe, T., Maeda, M., Hoshi, N., Sugino, T., Watanabe, K., Fukuda, T. and Suzuki, T. (2000) Fatty acid synthase is expressed mainly in adult hormone- sensitive cells or cells with high lipid metabolism and in proliferating fetal cells. J Histochem Cytochem 48, 613-22. Wakil, SJ. (1989) Fatty acid synthase, a proficient multifunctional enzyme. Biochemistry 28, 4523-30. ' Aprahamian, S.A., Arslanian, M.J. and Wakil, SJ. (1982) Comparative studies on the kinetic parameters and product analyses of chicken and rat liver and yeast fatty acid synthetase. Comp Biochem Physiol B 71, 577-82. Nugteren, DH. (1965) The enzymic chain elongation of fatty acids by rat-liver rrricrosomes. Biochim Biophys Acta 106, 280-90. Cinti, D.L., Cook, L., Nagi, MN. and Suneja, SK. (1992) The fatty acid chain elongation system of mammalian endoplasmic reticulum. Prog Lipid Res 31, 1-51. Moon, Y.A. and Horton, J .D. (2003) Identification of two mammalian reductases involved in the two-carbon fatty acyl elongation cascade. J Biol Chem 278, 7335- 43. Nagi, M.N., Cook, L., Ghesquier, D. and Cinti, D.L. (1986) Induction of rat liver mitochondrial fatty acid elongation by the administration of peroxisome 168 [26] [27] [28] [29] [30] [31] [32] [33] [34] [351 [36] proliferator di-(2-ethylhexyl)phthalate: absence of elongation activity in peroxisomes. Arch Biochem Biophys 248, 408-18. Wang, Y., Botolin, D., Christian, B., Busik, C., Xu, J. & Jump, DB. (2005) Tissue-specific, nutritional and developmental regulation of rat fatty acid elongases. J Lipid Res 46, 706-715. Buhman, K.K., Chen, HQ and Farese, R.V., Jr. (2001) The enzymes of neutral lipid synthesis. J Biol Chem 276, 40369-72. Kasai, T., Ohguchi, K., Nakashima, S., Ito, Y., Naganawa, T., Kondo, N. and Nozawa, Y. (1998) Increased activity of oleate-dependent type phospholipase D during actinomycin D-induced apoptosis in Jurkat T cells. J Irnmunol 161, 6469- 74. Khoo, D.E., Fermor, B., Miller, J ., Wood, 08., Apostolov, K., Barker, W., Williamson, RC. and Habib, NA. (1991) Manipulation of body fat composition with sterculic acid can inhibit mammary carcinomas in vivo. Br J Cancer 63, 97- 101. Menendez, J .A., del Mar Barbacid, M., Montero, S., Sevilla, E., Escrich, E., Solanas, M., Cortes-Funes, H. and Colomer, R. (2001) Effects of garnma- linolenic acid and oleic acid on paclitaxel cytotoxicity in human breast cancer cells. Eur J Cancer 37, 402-13. Sprecher, H. (2000) Metabolism of highly unsaturated n-3 and n-6 fatty acids. Biochim Biophys Acta 1486, 219-31. Cunnane, SC. (2001) Application of new methods and analytical approaches to research on polyunsaturated fatty acid homeostasis. Lipids 36, 975-9. Cunnane, S.C., Nadeau, CR. and Likhodii, SS. (2001) NMR and isotope ratio mass spectrometry studies of in vivo uptake and metabolism of polyunsaturates by the developing rat brain. J Mol Neurosci 16, 173-80; discussion 215-21 . Srrrith, K.J. and Skelton, H. (2002) Arachidonic acid-derived bioactive lipids: their role and the role for their inhibitors in dermatology. J Cutan Med Surg 6, 241-56. Terano, T., Salmon, J .A., Higgs, GA. and Moncada, S. (1986) Eicosapentaenoic acid as a modulator of inflammation. Effect on prostaglandin and leukotriene synthesis. Biochem Pharrnacol 35, 779-85. Calder, RC. and Grimble, RF. (2002) Polyunsaturated fatty acids, inflammation and immunity. Eur J Clin Nutr 56 Suppl 3, 814-9. 169 [37] [3 8] [39] [40] [41] [42] [431 [441 1451 1461 [47] [48] [49] Harris, W.S., Assaad, B. and Poston, WC. (2006) Tissue omega-6/omega-3 fatty acid ratio and risk for coronary artery disease. Am J Cardiol 98, 19i-26i. Harris, W.S., Park, Y. and Isley, W.L. (2003) Cardiovascular disease and long- chain omega-3 fatty acids. Curr Opin Lipidol 14, 9-14. Calder, PC. (2006) n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83, 15058-15193. Calder, RC. (2006) Polyunsaturated fatty acids and inflammation. Prostaglandins Leukot Essent Fatty Acids 75, 197-202. German, O.L., Insua, M.F., Gentili, C., Rotstein, NP. and Politi, LE. (2006) Docosahexaenoic acid prevents apoptosis of retina photoreceptors by activating the ERK/MAPK pathway. J Neurochem 98, 1507-20. Chen, W., Esselman, W.J., Jump, DB. and Busik, J.V. (2005) Anti-inflammatory effect of docosahexaenoic acid on cytokine-induced adhesion molecule expression in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci 46, 4342-7. Marszalek, J .R. and Lodish, HF. (2005) Docosahexaenoic acid, fatty acid- interacting proteins, and neuronal function: breastrnilk and fish are good for you. Annu Rev Cell Dev Biol 21, 633-57. Jump, D.B., Botolin, D., Wang, Y., Xu, J ., Christian, B. and Demeure, O. (2005) Fatty acid regulation of hepatic gene transcription. J Nutr 135, 2503-6. Jump, D.B., Thelen, A. and Mater, M. (1999) Dietary polyunsaturated fatty acids and hepatic gene expression. Lipids 34 Suppl, 8209-12. Jump, D.B., Thelen, A., Ren, B. and Mater, M. (1999) Multiple mechanisms for polyunsaturated fatty acid regulation of hepatic gene transcription. Prostaglandins Leukot Essent Fatty Acids 60, 345-9. Hertz, R., Magenheim, J ., Berrnan, I. and Bar-Tana, J. (1998) Fatty acyl-CoA thioesters are ligands of hepatic nuclear factor-4alpha. Nature 392, 512-6. Wisely, G.B., Miller, A.B., Davis, R.G., Thomquest, A.D., Jr., Johnson, R., Spitzer, T., Sefler, A., Shearer, B., Moore, J .T., Miller, A.B., Willson, T.M. and Williams, SP. (2002) Hepatocyte nuclear factor 4 is a transcription factor that constitutively binds fatty acids. Structure 10, 1225-34. de Urquiza, A.M., Liu, S., Sjoberg, M., Zetterstrom, R.H., Griffrths, W., Sjovall, J. and Perlrnann, T. (2000) Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science 290, 2140-4. 170 [50] [51] [521 [53] [54] [55] [56] [57] [581 1591 [601 Dhe-Paganon, S., Duda, K., Iwarnoto, M., Chi, Y.I. and Shoelson, SE. (2002) Crystal structure of the HNF4 alpha ligand binding domain in complex with endogenous fatty acid ligand. J Biol Chem 277, 37973-6. Desvergne, B. and Wahli, W. (1999) Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev 20, 649-88. Pawar, A., Botolin, D., Mangelsdorf, DJ. and Jump, DB. (2003) The role of liver X receptor-alpha in the fatty acid regulation of hepatic gene expression. J Biol Chem 27 8, 40736-43. Pawar, A. and Jump, DB. (2003) Unsaturated fatty acid regulation of peroxisome proliferator-activated receptor alpha activity in rat primary hepatocytes. J Biol Chem 278, 35931-9. Pawar, A., Xu, J ., Jerks, E., Mangelsdorf, DJ. and Jump, DB. (2002) Fatty acid regulation of liver X receptors (LXR) and peroxisome proliferator-activated receptor alpha (PPARalpha) in HEK293 cells. J Biol Chem 277, 39243-50. Xu, J ., Christian, B. and Jump, DB. (2006) Regulation of rat hepatic L-pyruvate kinase promoter composition and activity by glucose, n-3 polyunsaturated fatty acids, and peroxisome proliferator-activated receptor-alpha agonist. J Biol Chem 281, 18351-62. Jump, D.B., Botolin, D., Wang, Y., Xu, J ., Christian, B., and Demeure, O. (2005) Fatty acid regulation of gene transcription. J. Nutr 135, 2503-2506. Dentin, R., Benhamed, F., Pegorier, J .P., Foufelle, F., Viollet, B., Vaulont, S., Girard, J. and Postic, C. (2005) Polyunsaturated fatty acids suppress glycolytic and lipogenic genes through the inhibition of ChREBP nuclear protein translocation. J Clin Invest 115, 2843-54. Chakravarthy, M.V., Pan, Z., Zhu, Y., Tordjman, K., Schneider, J .G., Coleman, T., Turk, J. and Semenkovich, C.F. (2005) "New" hepatic fat activates PPARalpha to maintain glucose, lipid, and cholesterol homeostasis. Cell Metab 1, 309-22. Xu, J ., Christian, B., and Jump, DB. (2006) Regulation of rat hepatic L-pyruvate kinase promoter composition and activity by glucose, N-3 PUFA and peroxisome proliferator activated receptor-alpha agonist. J Biol Chem 281, In Press. Xu, J ., Cho, H., O'Malley, S., Park, J .H. and Clarke, SD. (2002) Dietary polyunsaturated fats regulate rat liver sterol regulatory element binding proteins-1 and -2 in three distinct stages and by different mechanisms. J Nutr 132, 3333-9. 171 [611 [62] [631 [64] [651 [661 [67] [68] 1691 [70] [71] Xu, J ., Teran-Garcia, M., Park, J .H., Nakamura, MT and Clarke, SD. (2001) Polyunsaturated fatty acids suppress hepatic sterol regulatory element-binding protein-1 expression by accelerating transcript decay. J Biol Chem 276, 9800-7. Ma, L., Tsatsos, N.G. and Towle, HQ (2005) Direct role of ChREBP.Mlx in regulating hepatic glucose-responsive genes. J Biol Chem 280, 12019-27. Simopoulos, AP. (2003) Essential fatty acids in health and chronic diseases. Forum Nutr 56, 67-70. Ravnskjaer, K., Boergesen, M., Rubi, B., Larsen, J .K., Nielsen, T., Fridriksson, J., Maechler, P. and Mandrup, S. (2005) Peroxisome proliferator-activated receptor alpha (PPARalpha) potentiates, whereas PPARgamma attenuates, glucose- stimulated insulin secretion in pancreatic beta-cells. Endocrinology 146, 3266-76. Guerre-Millo, M., Gervois, P., Raspe, E., Madsen, L., Poulain, P., Derudas, B., Herbert, J .M., Winegar, D.A., Willson, T.M., Fruchart, J .C., Berge, R.K. and Staels, B. (2000) Peroxisome proliferator-activated receptor alpha activators improve insulin sensitivity and reduce adiposity. J Biol Chem 275, 16638-42. Guerre-Millo, M., Rouault, C., Poulain, P., Andre, J ., Poitout, V., Peters, J .M., Gonzalez, F.J., Fruchart, J .C., Reach, G. and Staels, B. (2001) PPAR-alpha-null mice are protected fi'om high-fat diet-induced insulin resistance. Diabetes 50, 2809-14. Ye, J .M., Doyle, P.J., Iglesias, M.A., Watson, D.G., Cooney, G.J. and Kraegen, E.W. (2001) Peroxisome proliferator-activated receptor (PPAR)-alpha activation lowers muscle lipids and improves insulin sensitivity in high fat-fed rats: ‘ comparison with PPAR-gamma activation. Diabetes 50, 411-7. , Mascaro, C., Acosta, E., Ortiz, J .A., Marrero, P.F., Hegardt, PG. and Haro, D. (1998) Control of human muscle-type carnitine palmitoyltransferase I gene transcription by peroxisome proliferator-activated receptor. J Biol Chem 273, 8560-3. Varanasi, U., Chu, R., Huang, Q., Castellon, R., Yeldandi, A.V. and Reddy, J .K. (1996) Identification of a peroxisome proliferator-responsive element upstream of the human peroxisomal fatty acyl coenzyme A oxidase gene. J Biol Chem 271 , 2147-55. Benatti, P., Peluso, G., Nicolai, R. and Calvani, M. (2004) Polyunsaturated fatty acids: biochemical, nutritional and epigenetic properties. J Am Coll Nutr 23, 281- 302. Hermanson, 0., Glass, GK. and Rosenfeld, M.G. (2002) Nuclear receptor coregulators: multiple modes of modification. Trends Endocrinol Metab 13, 55-60. 172 [72] [731 [74] [75] [76] [77] [78] [79] [801 181] [82] Clarke, SD. and Jump, DB. (1994) Dietary polyunsaturated fatty acid regulation of gene transcription. Annu Rev Nutr 14, 83-98. Xu, H.E., Lambert, M.H., Montana, V.G., Plunket, K.D., Moore, L.B., Collins, J .L., Oplinger, J .A., Kliewer, S.A., Gampe, R.T., Jr., McKee, D.D., Moore, J .T. and Willson, TM. (2001) Structural determinants of ligand binding selectivity between the peroxisome proliferator-activated receptors. Proc Natl Acad Sci U S A 98, 13919-24. Mater, M.K., Thelen, A.P., Pan, DA. and Jump, DB. (1999) Sterol response element-binding protein 10 (SREBPlc) is involved in the polyunsaturated fatty acid suppression of hepatic Sl4 gene transcription. J Biol Chem 274, 32725-32. Ren, B., Thelen, A.P., Peters, J .M., Gonzalez, F .J . and Jump, DB. (1997) Polyunsaturated fatty acid suppression of hepatic fatty acid synthase and 814 gene expression does not require peroxisome proliferator-activated receptor alpha. J Biol Chem 272, 26827-32. Storlien, L.H., Hulbert, A.J. and Else, PL. (1998) Polyunsaturated fatty acids, membrane function and metabolic diseases such as diabetes and obesity. Curr Opin Clin Nutr Metab Care 1, 559-63. Brown, MS. and Goldstein, J .L. (1997) The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell 89, 331-40. Horton, J .D., Goldstein, J .L. and Brown, MS. (2002) SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 109,1125-31. Yang, T., Espenshade, P.J., Wright, M.E., Yabe, D., Gong, Y., Aebersold, R., Goldstein, J .L. and Brown, MS. (2002) Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell 110, 489-500. Yabe, D., Brown, MS. and Goldstein, J .L. (2002) Insig-2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element- binding proteins. Proc Natl Acad Sci U S A 99, 12753-8. Hannah, V.C., Ou, J ., Luong, A., Goldstein, J .L. and Brown, MS. (2001) Unsaturated fatty acids down-regulate srebp isoforms la and 1c by two mechanisms in HEK-293 cells. J Biol Chem 276, 4365-72. Xu, J ., Nakamura, M.T., Cho, HP. and Clarke, SD. (1999) Sterol regulatory element binding protein-1 expression is suppressed by dietary polyunsaturated 173 [83] [84] [85] [861 [87] [38] [89] [90] [911 [92] fatty acids. A mechanism for the coordinate suppression of lipogenic genes by polyunsaturated fats. J Biol Chem 274, 23577-83. Botolin, D., Wang, Y., Christian, B. and Jump, DB. (2006) Docosahexaneoic acid (22:6,n-3) regulates rat hepatocyte SREBP-1 nuclear abundance by Erk- and 26S proteasome-dependent pathways. J Lipid Res 47, 181-92. ' Ma, L., Robinson, L.N. and Towle, HQ (2006) ChREBP*Mlx is the principal mediator of glucose-induced gene expression in the liver. J Biol Chem 281, 28721-30. O'Callaghan, B.L., Koo, S.H., Wu, Y., Freake, HQ and Towle, HQ (2001) Glucose regulation of the acetyl-CoA carboxylase promoter P1 in rat hepatocytes. J Biol Chem 276, 16033-9. Koo, SH. and Towle, HQ (2000) Glucose regulation of mouse S(14) gene expression in hepatocytes. Involvement of a novel transcription factor complex. J Biol Chem 275, 5200-7. Ou, J ., Tu, H., Shan, B., Luk, A., DeBose-Boyd, R.A., Bashmakov, Y., Goldstein, J .L. and Brown, MS. (2001) Unsaturated fatty acids inhibit transcription of the sterol regulatory element-binding protein-1c (SREBP-1c) gene by antagonizing ligand-dependent activation of the LXR. Proc Natl Acad Sci U S A 98, 6027-32. DeBose-Boyd, R.A., Ou, J ., Goldstein, J .L. and Brown, MS. (2001) Expression of sterol regulatory element-binding protein 1c (SREBP-1c) mRNA in rat hepatoma cells requires endogenous LXR ligands. Proc Natl Acad Sci U S A 98, 1477-82. Repa, J .J ., Berge, K.E., Pomajzl, C., Richardson, J .A., Hobbs, H. and Mangelsdorf, DJ. (2002) Regulation of ATP-binding cassette sterol transporters ABCGS and ABCG8 by the liver X receptors alpha and beta.J Biol Chem 277, 18793-800. Liimatta, M., Towle, H.C., Clarke, S. and Jump, DB. (1994) Dietary polyunsaturated fatty acids interfere with the insulin/ glucose activation of L-type pyruvate kinase gene transcription. Mol Endocrinol 8, 1147-53. Browning, L.M., Krebs, J .D., Moore, C.S., Mishra, G.D., O'Connell M, A. and Jebb, SA. (2007) The impact of long chain n-3 polyunsaturated fatty acid supplementation on inflammation, insulin sensitivity and CVD risk in a group of overweight women with an inflammatory phenotype. Diabetes Obes Metab 9, 70- 80. Herbaut, C. (2006) [Omega-3 and health]. Rev Med Brux 27, S355-60. 174 [931 [94] [95] [96] [97] [98] [99] [100] [101] [102] [103] [104] [105] Harris, W.S. (2006) The omega-6/omega-3 ratio and cardiovascular disease risk: uses and abuses. Curr Atheroscler Rep 8, 453-9. Bigger, J .T., Jr. and El-Sherif, T. (2001) Polyunsaturated fatty acids and cardiovascular events: a fish tale. Circulation 103, 623-5. Kang, J .X. and Leaf, A. (2000) Prevention of fatal cardiac arrhythmias by polyunsaturated fatty acids. Am J Clin Nutr 71, 2028-7 S. AP, S. (1994) Fatty acids. In Goldberg (ed): "Functional foods". pp. pp355 —391,‘ New York, Chapman & Hall. Phillipson, B.E., Rothrock, D.W., Connor, W.E., Harris, W.S. and Illingworth, DR. (1985) Reduction of plasma lipids, lipoproteins, and apoproteins by dietary fish oils in patients with hypertriglyceridemia. N Engl J Med 312, 1210-6. Kris-Etherton, P.M., Harris, W.S. and Appel, L.J. (2003) Fish consumption, fiSh oil, omega-3 fatty acids, and cardiovascular disease. Arterioscler Thromb Vasc Biol 23, e20-30. Borkman, M., Storlien, L.H., Pan, D.A., Jenkins, A.B., Chisholm, DJ. and Campbell, L.V. (1993) The relation between insulin sensitivity and the fatty-acid composition of skeletal-muscle phospholipids. N Engl J Med 328, 238-44. Simopoulos, AP. (1994) Is insulin resistance influenced by dietary linoleic acid and trans fatty acids? Free Radic Biol Med 17, 367-72. Storlien, L.H., Kriketos, A.D., Jenkins, A.B., Baur, L.A., Pan, D.A., Tapsell, LC. and Calvert, GD. (1997) Does dietary fat influence insulin action? Ann N Y Acad Sci 827, 287-301. ' ’ Pan, D.A., Lillioja, S., Kriketos, A.D., Milner, M.R., Baur, L.A., Bogardus, C., Jenkins, AB. and Storlien, L.H. (1997) Skeletal muscle triglyceride levels are inversely related to insulin action. Diabetes 46, 983-8. Connor, W.E., Prince, M.J., Ullrnann, D., Riddle, M., Hatcher, L., Smith, RE. and Wilson, D. (1993) The hypotriglyceridemic effect of fish oil in adult-onset diabetes without adverse glucose control. Ann N Y Acad Sci 683, 337-40. Zhang, Y., Proenca, R., Maffei, M., Barone, M., Leopold, L. and Friedman, J .M. (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425-32. Rojo-Martinez, G., Soriguer, F.J., Gonzalez-Romero, S., Tinahones, F., Moreno, F ., de Adana, S.R., Garriga, M.J., Esteva, 1., Garcia-Ames, J ., Gomez-Zumaquero, 175 [106] [107] [103] [109] [110] [111] [112] [113] [114] [115] [116] J .M. and Garcia-Almeida, J .M. (2000) Serum leptin and habitual fatty acid dietary intake in patients with type 1 diabetes mellitus. Eur J Endocrinol 142, 263-8. Reseland, J .E., Haugen, F., Hollung, K., Solvoll, K., Halvorsen, B., Brude, I.R., Nenseter, M.S., Christiansen, E.N. and Drevon, CA. (2001) Reduction of leptin gene expression by dietary polyunsaturated fatty acids. J Lipid Res 42, 743-50. Henderson, B., Pettipher, ER. and Higgs, GA. (1987) Mediators of rheumatoid arthritis. Br Med Bull 43, 415-28. James, M.J., Gibson, RA. and Cleland, LG. (2000) Dietary polyunsaturated fatty acids and inflammatory mediator production. Am J Clin Nutr 71 , 343 S-8S. Meydani, SN. and Dinarello, CA. (1993) Influence of dietary fatty acids on cytokine production and its clinical implications. Nutr Clin Pract 8, 65-72. Welsch, CW. (1995) Review of the effects of dietary fat on experimental mammary gland turnorigenesis: role of lipid peroxidation. Free Radic Biol Med 18, 757-73. Pariza, M.W. (1987) Dietary fat, calorie restriction, ad libitum feeding, and cancer i risk. Nutr Rev 45, 1-7. Rose, DP. and Connolly, J .M. (1998) Influence of dietary linoleic acid on experimental human breast cancer cell metastasis in athyrrric nude mice. Int J Oncol 13, 1179-83. Hilakivi-Clarke, L., Stoica, A., Raygada, M. and Martin, MB. (1998) Consumption of a high-fat diet alters estrogen receptor content, protein kinase C activity, and mammary gland morphology in virgin and pregnant mice and female offspring. Cancer Res 58, 654-60. Tvrdik, P., Westerberg, R., Silve, S., Asadi, A., Jakobsson, A., Cannon, B., Loison, G. and J acobsson, A. (2000) Role of a new mammalian gene family in the biosynthesis of very long chain fatty acids and Sphingolipids. J Cell Biol 149, 707-18. Leonard, A.B., Pereira, S.L., Sprecher, H. and Huang, Y.S. (2004) Elongation of long-chain fatty acids. Prog Lipid Res 43, 36-54. Tvrdik, P., Asadi, A., Kozak, L.P., Nedergaard, J ., Cannon, B. and Jacobsson, A. (1997) Cig30, a mouse member of a novel membrane protein gene family, is involved in the recruitment of brown adipose tissue. J Biol Chem 272, 3173 8-46. 176 [117] [113] [119] [120] [121] [122] [123] [124] [125] [126] Moon, Y.A., Shah, N.A., Mohapatra, S., Warrington, J .A. and Horton, J .D. (2001) Identification of a mammalian long chain fatty acyl elongase regulated by sterol regulatory element-binding proteins. J Biol Chem 276, 45358-66. Leonard, A.B., Bobik, E.G., Dorado, J ., Kroeger, P.E., Chuang, L.T., Thurmond, J .M., Parker-Barnes, J .M., Das, T., Huang, Y.S. and Mukerji, P. (2000) Cloning of a human cDNA encoding a novel enzyme involved in the elongation of long- chain polyunsaturated fatty acids. Biochem J 350 Pt 3, 765-70. Inagaki, K., Aki, T., Fukuda, Y., Kawamoto, S., Shigeta, S., Ono, K. and Suzuki, 0. (2002) Identification and expression of a rat fatty acid elongase involved in the biosynthesis of C18 fatty acids. Biosci Biotechnol Biochem 66, 613-21. Zhang, K., Kniazeva, M., Han, M., Li, W., Yu, Z., Yang, Z., Li, Y., Metzker, M.L., Allikmets, R., Zack, D.J., Kakuk, L.E., Lagali, P.S., Wong, P.W., MacDonald, I.M., Sieving, P.A., Figueroa, D.J., Austin, C.P., Gould, R.J., Ayyagari, R and Petrukhin, K. (2001) A 5-bp deletion in ELOVL4 is associated with two related forms of autosomal dominant macular dystrophy. Nat Genet 27, 89-93. Oh, C.S., Toke, D.A., Mandala, S. and Martin, CE. (1997) EL02 and ELO3, homologues of the Saccharomyces cerevisiae ELOl gene, function in fatty acid elongation and are required for sphingolipid formation. J Biol Chem 272, 173 76- 84. Hanada, K., Nishijima, M., Kiso, M., Hasegawa, A., Fujita, S., Ogawa, T. and Akamatsu, Y. (1992) Sphingolipids are essential for the growth of Chinese hamster ovary cells. Restoration of the growth of a mutant defective in sphingoid base biosynthesis by exogenous sphingolipids. J Biol Chem 267, 23527-33. Simons, K. and Ikonen, E. (1997) Functional rafts in cell membranes. Nature 387, 569-72. Garcia-Arranz, M., Maldonado, A.M., Mazon, M.J. and Portillo, F. (1994) Transcriptional control of yeast plasma membrane H(+)-ATPase by glucose. Cloning and characterization of a new gene involved in this regulation. J Biol Chem 269, 18076-82. Zeidel, ML. (1996) Low permeabilities of apical membranes of barrier epithelia: what makes watertight membranes watertight? Am J Physiol 271, F243 -5. Suneja, S.K., Nagi, M.N., Cook, L. and Cinti, D.L. (1991) Decreased long-chain fatty acyl CoA elongation activity in quaking and jirnpy mouse brain: deficiency in one enzyme or multiple enzyme activities? J Neurochem 57, 140-6. 177 [127] Leonard, A.B., Kelder, B., Bobik, E.G., Chuang, L.T., Lewis, C.J., Kopchick, J .J ., Mukerji, P. and Huang, Y.S. (2002) Identification and expression of mammalian long-chain PUF A elongation enzymes. Lipids 37, 733—40. [128] Westerberg, R., Tvrdik, P., Unden, A.B., Mansson, J .E., Norlen, L., Jakobsson, A., Holleran, W.H., Elias, P.M., Asadi, A., Flodby, P., Toftgard, R., Capecchi, MR. and Jacobsson, A. (2004) Role for ELOVL3 and fatty acid chain length in development of hair and skin function. J Biol Chem 279, 5621-9. [129] Westerberg, R., Mansson, J .E., Golozoubova, V., Shabalina, I.G., Backlund,E.C., Tvrdik, P., Retterstol, K., Capecchi, MR. and Jacobsson, A. (2006) ELOVL3 is an important component for early onset of lipid recruitment in brown adipose tissue. J Biol Chem 281, 4958-68. [130] Jakobsson, A., J orgensen, J .A. and Jacobsson, A. (2005) Differential regulation of fatty acid elongation enzymes in brown adipocytes implies a unique role for Elovl3 during increased fatty acid oxidation. Am J Physiol Endocrinol Metab 289, E5 17-26. [131] Anzulovich, A., Mir, A., Brewer, M., Ferreyra, G., Vinson, C. and Baler, R (2006) Elovl3: a model gene to dissect homeostatic links between the circadian clock and nutritional status. J Lipid Res 47, 2690-700. [132] Bernstein, P.S., Tammur, J ., Singh, N., Hutchinson, A., Dixon, M., Pappas, C.M., Zabriskie, N.A., Zhang, K., Petrukhin, K., Leppert, M. and Allikmets, R. (2001) Diverse macular dystrophy phenotype caused by a novel complex mutation in the ELOVL4 gene. Invest Ophtlralmol Vis Sci 42, 3331-6. [133] Vasireddy, V., Vijayasarathy, C., Huang, J ., Wang, X.F., Jablonski, M.M., Petty, H.R., Sieving, PA. and Ayyagari, R. (2005) Stargardt-like macular dystrophy . protein ELOVL4 exerts a dominant negative effect by recruiting wild-typeprotein into aggresomes. Mol Vis 11, 665-76. [134] Matsuzaka, T., Shimano, H., Yahagi, N., Yoshikawa, T., Amemiya-Kudo, M., Hasty, A.H., Okazaki, H., Tamura, Y., Iizuka, Y., Ohashi, K., Osuga, J., Takahashi, A., Yato, S., Sone, H., Ishibashi, S. and Yamada, N. (2002) Cloning and characterization of a mammalian fatty acyl-CoA elongase as a lipogenic enzyme regulated by SREBPs. J Lipid Res 43, 911-20. [135] Gutierrez-Juarez, R., Pocai, A., Mulas, C., Ono, H., Bhanot, S., Monia, BR and Rossetti, L. (2006) Critical role of stearoyl-CoA desaturase-1 (SCDl) in the onset of diet-induced hepatic insulin resistance. J Clin Invest 116, 1686-95. [136] Pawar, A., and Jump, DB. (2003) Unsaturated fatty acid regulation of peroxisome proliferator activated receptor-alpha activity in primary rat hepatocytes. J Biol Chem 278, 35931-35939. ’ 178 Ir [137] [138] [139] [140] [141] [142] [143] [144] [145] [146] Hillgartrrer, F.B., Salati, L.M. and Goodridge, A.G. (1995) Physiological and molecular mechanisms involved in nutritional regulation of fatty acid synthesis. Physiol Rev 75, 47-76. Prasad, M.R., Nagi, M.N., Ghesquier, D., Cook, L. and Cinti, D.L. (1986) Evidence for multiple condensing enzymes in rat hepatic microsomes catalyzing the condensation of saturated, monounsaturated and polyunsaturated acyl coenzyme A. J Biol Chem 261, 8213-8217. Horton, J .D., Shah, N.A., Warrington, J .A., Anderson, N.N., Park, S.W., Brown, MS. and Goldstein, J .L. (2003) Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc Natl Acad Sci U S A 100, 12027-12032. Westerberg, R., Tvrdik, P., Unden, A-B., Mansson, J -E., Norlen, L., Jakobsson, A., Holleran, W.H., Elias, P.M., Asadi, A., Flodby, P., Toftgard, R., Capecchi, M.R., and Jacobsson, A. (2004) Role for ELOVL3 and fatty acid chain length in development of hair and skin function. J Biol Chem 279, 5621-5629. Lagali, P.S., Liu, J ., Ambasudhan, R., Kakuk, L.E., Bernstein, S.L., Seigel, G.M., Wong, P.W. and Ayyagari, R. (2003) Evolutionarily conserved ELOVL4 gene expression in the vertebrate retina. Invest Ophthalmol Vis Sci 44, 2841-50. Tvrdik, P., Asadi, A., Kozak, L.P., Nedergaard, J ., Cannon, B., and Jacobsson, A. (1997) Cig30, a mouse member of a novel membrane protein gene family, is involved in the recruitment of brown adipose tissue. J Biol Chem 272, 3173 8- 3 1746. Alava, M.A., Iturralde, M., Gonzalez, B. and Pineiro, A. (1999) Fatty acid desaturation: effect of alphafetoprotein on alpha-linolenic acid conversion by fetal rat hepatocytes. Prostaglandins Leukot Essent Fatty Acids 60, 209-15. Alegret, M., Cerqueda, E., Ferrando, R., Vazquez, M., Sanchez, R.M., Adzet, T., Merlos, M. and Laguna, J .C. (1995) Selective modification of rat hepatic microsomal fatty acid chain elongation and desaturation by fibrates: relationship with peroxisome proliferation. Br J Pharmacol 114, 1351-8. Asadi, A., Jorgensen, J ., and Jacobsson, A. (2002) Elovll and p55Cdc genes are localized in a tail-to-tail array and are co-expressin in prolifrating cells. J Biol Chem 277, 18494-18500. Chang, J .H., Lunt, D.K. and Smith, SE. (1992) Fatty acid composition and fatty acid elongase and stearoyl-CoA desaturase activities in tissues of steers fed high oleate sunflower seed. J Nutr 122, 2074-80. 179 I". [147] [148] [149] [150] [151] [152] [153] [154] [155] [156] [157] [158] Cinci, G., Guerranti, R., Pagani, R., Carlucci, F., Terzuoli, L., Rosi, F. and Marinello, E. (2000) Fatty acid composition of phospholipids, triglycerides and cholesterol in serum of castrated and estradiol treated rats. Life Sci 66, 1647-54. Clore, J .N., Li, L. and Rizzo, W.B. (2000) Effects of fructose and troglitazone on phospholipid fatty acid composition in rat skeletal muscle. Lipids 35, 1281-7. de Antueno, R.J., Knickle, L.C., Smith, H., Elliot, M.L., Allen, S.J., Nwaka, S. and Wintlrer, MD. (2001) Activity of human DeltaS and Delta6 desaturases on multiple n-3 and n-6 polyunsaturated fatty acids. F EBS Lett 509, 77-80. Fan, Y.Y. and Chapkin, RS. (1998) Importance of dietary gamma-linolenic acid in human health and nutrition. J Nutr 128, 1411-4. Keyes, S.R.a.C., D.L. (1980) Biochemical properties of cytochrome b5-dependent microsomal fatty acid elongation and identification of products. J Biol Chem 255, 11357-11364. Leonard, A.B., Bobik, E.G., Dorado, J ., Kroeger, P.E., Chuang, L.T., Thurmond, J .M., Parker-Parnes, J .M., Das, T., Huang, Y.S. and Mukerji, P. (2000) Cloning of a human cDNA encoding a novel enzyme involved in the elongation of PUF A. Biochem J 350, 765-770. Leonard, A.B., Pereira, S.L., Sprecher, H. and Huang, Y.S. (2004) Elongation of long-chain fatty acids. Prog Lipid Res. 43, 36-54. Suneja, S.K., Osei, P., Cook, L., Nagi, MN. and Cinti, D.L. (1990) Enzyme site- specific changes in hepatic microsomal fatty acid chain elongation in streptozotocin-induced diabetic rats. Biochim Biophys Acta 1042, 81-5. Tvrdik, P., Westerberg, R., Silve, S., Asadi, A., Jakobsson, A., Cannon, B., Loison, G., and Jacobsson, A. (2000) Role of a new mammalian gene family in the biosynthesis of very long fatty acids and Sphingolipids. J Cell Biol. 149, 707- 717. Walker, K.A. and Harwood, J .L. (1986) Evidence for separate elongation enzymes for very-long-chain-fatty-acid synthesis in potato (Solanum tuberosum). Biochem J 237, 41-6. Mater, M.K., Thelen, A.P., Pan, DA. and Jump, D.B. ( 1999) Sterol regulatory element-binding protein 1c (SREBPlc) is involved in the polyunsaturated fatty acid suppression of hepatic Sl4 gene transcription. J Biol Chem 274, 32725-32. Pan, D.A., Mater, M.K., Thelen, A.P., Peters, J .M., Gonzalez, F.J. and Jump, DB. (2000) Evidence against the peroxisome proliferator-activated receptor alpha 180 at [159] [160] [161] [162] [163] [164] [165] [166] [167] [168] [169] [170] (PPARalpha) as the mediator for polyunsaturated fatty acid suppression of hepatic L-pyruvate kinase gene transcription. J Lipid Res 41, 742-51. Botolin, D. and Jump, DB. (2003) Selective proteolytic processing of rat hepatic sterol regulatory element binding protein-1 (SREBP-l) and SREBP-2 during postnatal development. J Biol Chem 278, 6959-62. Nakamura, M.T.a.N., TX. (2004) Structure, function and dietary regulation of delta6, delta 5 and delta 9 desaturases. Annu. Rev. Nutr. 24, 345-376. Miller, CW. and Ntambi, J .M. (1996) Peroxisome proliferators induce mouse liver stearoyl-CoA desaturase 1 gene expression. Proc Natl Acad Sci U S A 93, 9443-8. Fulco, A.J. and Mead, J .F. (1959) Metabolism of essential fatty acids. VIII. Origin of 5,8,11-eicosatrienoic acid in the fat-deficient rat. J Biol Chem 234, 1411-6. Johnson, P.V. (1985) Dietary fat, eicosanoids and immunity. Adv. Lipid Res. 21, 103-141. Holman, RT. (1960) The ratio of trienoicztetraenoic acids in tissue lipids as a measure of essential fatty acid requirement. J Nutr 70, 405-410. Braissant, O.a.W., W. (1998) Differential expression of peroxisome‘proliferator- activated receptor-alpha, beta and gamma during rat embryonic development. Endocrinology 139, 2748-2754. Cook, W.S., Jain, S., Jia, Y., Cao, W-Q, Yeldandi, A.V., Reddy, J .K. and Rao, MS. (2001) Peroxisome proliferator-activated receptor alpha-responsive genes induced in the newborn, but not pematal liver of peroxisomal fatty acyl CoA oxidase null mice. Expt. Cell Res 268, 70-76. . Bennett, M.K., Lopez, J .M., Sanchez, H.B. and Osborne, T.F.,(1995) Sterol regulation of fatty acid synthase promoter. Coordinate feedback regulation of two major lipid pathways. J Biol Chem 270, 25578-83. Girard, J. (1990) Metabolic adaptations to change of nutrition at birth. Biol Neonate 58 Suppl 1, 3-15. Ahima, R.S., Prabakaran, D. and Flier, J .S. (1998) Postnatal leptin surge and regulation of circadian rhythm of leptin by feeding. Implications for energy homeostasis and neuroendocrine function. J Clin Invest 101, 1020-7. Girard, J ., Perdereau, D., Foufelle, F., Prip-Buus, C. and Ferre, P. (1994) Regulation of lipogenic enzyme gene expression by nutrients and hormones. ' Faseb J 8, 36-42. ' ‘ ' 181 [171] [172] [173] [174] [175] [176] [177] [178] [179] [180] [181] Catalan, J ., Moriguchi, T., Slotnick, B., Murthy, M., Greiner, RS. and Salem, N., Jr. (2002) Cognitive deficits in docosahexaenoic acid-deficient rats. Behav Neurosci 116, 1022-31. Mater, M.K., Thelen, AP. and Jump, DB. (1999) Arachidonic acid and PGE2 regulation of hepatic lipogenic gene expression. J Lipid Res 40, 1045-52. Stoeckman, A.K., Ma, L. and Towle, HQ (2004) Mlx is the functional ' heteromeric partner of the carbohydrate response element-binding protein in glucose regulation of lipogenic enzyme genes. J Biol Chem 279, 15662-9. Jump, DB. and Clarke, SD. (1999) Regulation of gene expression by dietary fat. Annu Rev Nutr 19, 63-90. Wang, Y., Botolin, D., Christian, B., Busik, J ., Xu, J. and Jump, DB. (2005) Tissue-specific, nutritional, and developmental regulation of rat fatty acid elongases. J Lipid Res 46, 706-15. Jump, D.B., Bell, A., Lepar, G. and Hu, D. (1990) Insulin rapidly induces rat liver Sl4 gene transcription. Mol Endocrinol 4, 1655-60. Akiyama, T.E., Nicol, C.J., Fievet, C., Staels, B., Ward, J .M., Auwerx, J ., Lee, S.S., Gonzalez, F .J . and Peters, J .M. (2001) Peroxisome proliferator-activated receptor-alpha regulates lipid homeostasis, but is not associated with obesity: studies with congenic mouse lines. J Biol Chem 276, 39088-93. Lee, S. S., Pineau, T. ,,Drago J. Lee, E. J., Owens, J. W. Kroetz, D. L., Femandez- Salguero, P. M., Westphal, H. and Gonzalez, F. J. (1995) Targeted disruptiOn of the alpha isoforrn of the peroxisome proliferator-activated receptor gene in mice results in abolishment of the pleiotropic effects of peroxisome proliferators. Mol Cell Biol 15, 3012-22. . Botolin, D., Wang, Y.,Christian, B., and Jump, DB. (2006) Docosahexaneoic acid [22: 6, n-3] stimulates rat hepatic sterol regulatory element, binding protein- lc (SREBP-1c) degradation by an Erk-and 26S proteasome-dependent pathWay. J Lipid Res 47, 181-192. Stoeckman, AK. and Towle, HQ (2002) The role of SREBP-1c in nutritional regulation of lipogenic enzyme gene expression. J Biol Chem 277, 27029-35. Miyazaki, M., Dobrzyn, A., Man, W.C., Chu, K., Sarnpath, H., Kim, H.J. and Ntambi, J .M. (2004) Stearoyl-CoA desaturase 1 gene expression is necessary for fructose-mediated induction of lipogenic gene expression by sterol regulatory ‘ element-binding protein-lc-dependent and -independent mechanisms. J Biol Chem 279, 25164-71. 182 [182] [183] [184] [185] [186] [187] [188] [189] [190] [191] [192] Schultz, J .R., Tu, H., Luk, A., Repa, J .J ., Medina, J.C., Li, L., Schwendner, S., Wang, S., Thoolen, M., Mangelsdorf, D.J., Lustig, K.D. and Shan, B. (2000) Role of LXRs in control of lipogenesis. Genes Dev 14, 2831-8. Joseph, S.B., Laffitte, B.A., Patel, P.H., Watson, M.A., Matsukuma, K.E., Walczak, R. Collins, J. L., Osborne, T. F. and Tontonoz, P. (2002) Direct and indirect mechanisms for regulation of fatty acid synthase gene expression by liver X receptors. J Biol Chem 277, 11019- 25. Tobin, K.A., Ulven, S.M., Schuster, G.U., Steineger, H.H., Andresen, S.M., Gustafsson, J .A. and Nebb, HI. (2002) Liver X receptors as insulin-mediating factors in fatty acid and cholesterol biosynthesis. J Biol Chem 277. Kawaguchi, T., Takenoshita, M., Kabashirna, T. and Uyeda, K. (2001) Glucose and CAMP regulate the L-type pyruvate kinase gene by phosphorylation/dephosphorylation of the carbohydrate response element binding protein. Proc Natl Acad Sci U S A 98, 13710-5. Towle, HQ (2005) Glucose as a regulator of eukaryotic gene transcription. Trends Endocrinol Metab 16, 489-94. Jakobsson, A., Westerberg, R., and Jacobsson, A. (2006) Fatty acid elongases in mammals: Their regulation and role in metabolism. Progress in Lipid Research 45, 23 7-249. Chinetti-Gbaguidi, G., Fruchart, J .C. and Staels, B. (2005) Role of the PPAR family of nuclear receptors in the regulation of metabolic and cardiovascular homeostasis: new approaches to therapy. Curr Opin Pharmacol 5, 177-83. Kakuma, T., Lee, Y., Higa, M., Wang, Z., Pan, W., Shimomura, I. and Unger, " RH. (2000) Leptin, troglitazone, and the expression of sterol regulatory element _ binding proteins in liver and pancreatic islets. Proc Natl Acad Sci U S A 97, 8536-41. Kalaany, N.Y., Gauthier, K.C., Zavacki, A.M., Mammen, P.P., Kitazume, T., Peterson, J .A., Horton, J .D., Garry, D.J., Bianco, AC. and Mangelsdorf, DJ. (2005) LXRs regulate the balance between fat storage and oxidation. Cell Metab 1, 231-44. Jayaprakasam, B., Vareed, S.K., Olson, L.K. and Nair, M.G. (2005) Insulin secretion by bioactive anthocyanins and anthocyanidins present in fruits. J Agric Food Chem 53, 28-31. Van Heck, M., France, C.F., Compton, D.S., McLeod, R.L., Yunribe, N.P., Alton, K.B., Sybertz, E.J. and Davis, H.R., Jr. (1997) In vivo metabolism-based ‘ discovery of a potent chOIesterol absorption inhibitor, SCHS 823 5 , [in the rat and 183 rhesus monkey through the identification of the active metabolites of SCH48461. J Pharmacol Exp Ther 283, 157-63. [193] Halaas, J .L., Gajiwala, K.S., Maffei, M., Cohen, S.L., Chait, B.T., Rabinowitz, D., [194] [195] Lallone, R.L., Burley, SK. and Friedman, J .M. (1995) Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269, 543 -6. Denison, MS. and Heath-Pagliuso, S. (1998) The Ah receptor: a regulator of the biochemical and toxicological actions of structurally diverse chemicals. Bull Environ Contam Toxicol 61, 557-68. Poland, A. and Knutson, J .C. (1982) 2,3,7,8-tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanism of toxicity. Annu Rev Pharmacol Toxicol 22, 517-54. [196] Cohen, R, Miyazaki, M., Socci, N.D., Hagge-Greenberg, A., Liedtke, W., Soukas, [197] [198] [199] [200] [201] [202] A.A., Sharma, R., Hudgins, L.C., Ntambi, J .M. and Friedman, J .M. (2002) Role? for stearoyl-CoA desaturase-1 in leptin-mediated weight loss. Science 297, 240-3. Zhang, W., Patil, S., Chauhan, B., Guo, S., Powell, D.R., Le, J ., Klotsas, A., Matika, R., Xiao, X., Franks, R., Heidenreich, K.A., Sajan, M.P., Farese, R.V., Stolz, D.B., Tso, R, Koo, S.H., Montrniny, M. and Unterman, T.G. (2006) FoxOl Regulates Multiple Metabolic Pathways in the Liver: Effects on gluconeogenic, glycolytic, and lipogenic gene expression. J Biol Chem 281, 10105-17. Kabashima, T., Kawaguchi, T., Wadzinski, BE. and Uyeda, K. (2003) Xylulose 5-phosphate mediates glucose-induced lipogenesis by xylulose 5-phosphate- activated protein phosphatase in rat liver. Proc Natl Acad Sci U S A 100, 5107-12. Uyeda, K., Yamashita, H. and Kawaguchi, T. (2002) Carbohydrate responsive element-binding protein (ChREBP): a key regulator of glucose metabolism and , fat storage. Biochem Pharmacol 63, 2075-80. Boverhof, D.R., Burgoon, L.D., Tashiro, C., Chittirn, B., Harkema, J .R., Jump, DB. and Zacharewski, T.R. (2005) Temporal and dose-dependent hepatic gene expression patterns in mice provide new insights into TCDD-Mediated hepatotoxicity. Toxicol Sci 85, 1048-63. Wang, Y., Botolin, D., Xu, J ., Christian, B., Mitchell, E., Jayaprakasam, B., Nair, M.G., Peters, J .M., Busik, J .V., Olson, L.K. and Jump, DB. (2006) Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and obesity. J Lipid Res 47, 2028-41. Lewin, T.M., Van Horn, C.G., Krisans, SK. and Coleman, RA. (2002) Rat'liver acyl-CoA synthetase 4 is a peripheral-membrane protein located in two distinct 184 [203] [204] [205] [206] [207] [208] [209] [210] [211] [212] subcellular organelles, peroxisomes, and mitochondrial-associated membrane. Arch Biochem Biophys 404, 263-70. Jakobsson, A., Westerberg, R. and Jacobsson, A. (2006) Fatty acid elongases in mammals: their regulation and roles in metabolism. Prog Lipid Res 45, 237-49. Kim, Y.B., Peroni, O.D., Aschenbach, W.G., Minokoshi, Y., Kotani, K., Zisman, A., Kahn, C.R., Goodyear, L.J. and Kahn, BB. (2005) Muscle-specific deletion of the Glut4 glucose transporter alters multiple regulatory steps in glycogen metabolism. Mol Cell Biol 25, 9713-23. Wang, Y., Botolin, D., Xu, J inghua, Christian, B., Mitchell, E., Jayaprakasam, B., Nair, M., Peters, J .M., Busik, J ., Olson, L.K., and Jump, DB. (2006) Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and obesity. J. Lipid. Res. 47, 2028-2041. Kockeritz, L., Doble, B., Patel, S. and Woodgett, J .R. (2006) Glycogen synthase kinase-3--an overview of an over-achieving protein kinase. Curr Drug Targets 7, 1377-88. Cohen, P. (1999) The Croonian Lecture 1998. Identification of a protein kinase cascade of major importance in insulin signal transduction. Philos Trans R Soc Lond B Biol Sci 354, 485-95. Zacks, D.N., Zheng, Q.D., Han, Y., Bakhru, R. and Miller, J .W. (2004) FAS- mediated apoptosis and its relation to intrinsic pathway activation in an experimental model of retinal detachment. Invest Ophthalmol Vis Sci 45, 4563-9. Sprecher, H. and Chen, Q. (1999) P01yunsaturated fatty acid biosynthesis: a’ _ microsomal-peroxisomal process. Prostaglandins Leukot Essent Fatty Acids 60, 3 17-21 . Peeters, M.J., Patijn, G.A., Lieber, A., Meuse, L. and Kay, MA. (1996) i Adenovirus-mediated hepatic gene transfer in mice: comparison of intravascular and biliary administration. Hum Gene Ther 7, 1693-9. Lieber, A., Vrancken Peeters, M.J., Meuse, L., Fausto, N., Perkins, J. and Kay, MA. (1995) Adenovirus-mediated urokinase gene transfer induces liver regeneration and allows for efficient retrovirus transduction of hepatocytes in vivo. Proc Natl Acad Sci U S A 92, 6210-4. Cline, G.W., Johnson, K., Regittnig, W., Perret, P., Tozzo, E., Xiao, L., Damico, C. and Shulrnan, G.I. (2002) Effects of a novel glycogen synthase kinase-3 inhibitor on insulin-stimulated glucose metabolism in Zucker diabetic fatty (fa/fa) rats. Diabetes 51, 2903-10. 185 111]11111]]]1]11]]]1[1]]1 . --------