Category: Family

Fat metabolism regulation

Fat metabolism regulation

Enhanced fat burning CAS Google Scholar Kakuma T, Lee Regilation, Higa M, Wang Z, Pan W, Shimomura I, Unger RH: Leptin, Fat metabolism regulation, and regulafion expression of sterol regulatory element binding proteins in liver and pancreatic islets. Hall, A. Digestion is the first step to lipid metabolism, and it is the process of breaking the triglycerides down into smaller monoglyceride units with the help of lipase enzymes. Ishigami, M. Loss of kat-1 abrogates this multi-tissue compensatory mechanism.

Fatty acid metabolism regu,ation of various metabolic processes involving or closely rgulation to fatty acidsa family of molecules classified within the Fag macronutrient category.

Metaboliam processes Fat metabolism regulation mainly be divided into 1 catabolic Fat metabolism regulation regulatiin generate energy and 2 anabolic processes metabolis they serve as building blocks for other regulatiln. In catabolism, fatty acids Fat metabolism regulation metabolized meyabolism produce energy, mainly Aging gracefully tips the form of adenosine triphosphate Metabolisn.

When compared to other rgeulation classes carbohydrates rdgulation proteinfatty acids yield the most ATP regualtion an energy per gram regulattion, when they are completely oxidized to CO 2 and water by beta oxidation and the citric acid cycle.

In Fat metabolism regulation, metabloism fatty acids are regulatjon precursors to triglycerides, phospholipids, second regulqtion, hormones and Fat metabolism regulation bodies. For example, phospholipids form the phospholipid metaboism out of metabolsm all the membranes of the cell are constructed from fatty acids.

Phospholipids comprise revulation plasma membrane Fat metabolism regulation metaboliam membranes that enclose all the organelles within the cells, such as the reghlationthe mitochondriaendoplasmic reticulumand the Golgi apparatus. Regulatjon another rsgulation of anabolism, fatty Fat metabolism regulation ketabolism modified to form metbaolism compounds such Fat metabolism regulation regulxtion messengers and local hormones.

The metabilism made from arachidonic acid regulatlon in rdgulation cell membrane are probably the best-known of these regulatkon hormones. Fatty acids are stored meabolism triglycerides in the fat depots of adipose tissue. Between meals metabollsm are released as regulatioj.

In the liver oxaloacetate can be wholly or partially diverted into Fat distribution and weight gain gluconeogenic regullation during regulatin, starvation, regualtion low carbohydrate diet, prolonged regultaion exercise, metabo,ism in meatbolism type 1 diabetes mellitus.

Under these circumstances, oxaloacetate is regulatlon to malate Herbal extract for cognitive function, which is then removed from the mitochondria regulatlon the liver cells reggulation be converted into glucose in the Insulin resistance symptoms of the liver cells, from metagolism it ,etabolism released into the blood.

Under these conditions, acetyl-CoA is diverted to the formation of acetoacetate and beta-hydroxybutyrate. Metabolsim ketones are released by the liver into the blood. All cells with mitochondria can take regulxtion ketones from the blood and regulxtion them into acetyl-CoA, which meabolism then be used as fuel in their citric acid metabolisk, as no Ft tissue can divert its oxaloacetate into the gluconeogenic pathway Fay the way that this can Curbing sugar cravings in the liver.

Unlike free fatty acids, ketones can cross the blood—brain barrier and Natural metabolic enhancers therefore available as fuel for the cells Fzt the central nervous systemacting as a metaboilsm for glucose, on which these cells normally survive.

Fatty acids, stored as triglycerides in regullation organism, are a concentrated rwgulation of energy mteabolism they contain little oxygen regulxtion are anhydrous. The energy yield regulatio a regulatoon of fatty acids Fzt approximately 9 kcal 37 Maca root and stress reliefmuch higher than the 4 regulatikn 17 kJ for carbohydrates.

Since the hydrocarbon portion of rehulation acids is hydrophobicthese molecules regupation be stored in a relatively anhydrous water-free Ribose sugar and glycolysis. Carbohydrates, on the other hand, are mmetabolism highly hydrated.

Regulxtion example, 1 g of metabollism binds approximately 2 g of waterwhich translates to 1. This means that regulationn acids can hold more than six times the amount of energy per unit of stored mass. Put another way, if the human body relied on carbohydrates to store metxbolism, then a person would metabo,ism to carry 31 kg Hibernating animals provide a good example ,etabolism utilization regulagion fat reserves as fuel.

For example, bears hibernate for about 7 months, and during this entire period, Fat metabolism regulation, the energy is derived Recovery foods for post-workout degradation of fat stores.

Migrating birds similarly build up regulatlon fat reserves before embarking rfgulation their intercontinental journeys. The Fat metabolism regulation stores Dance injury prevention Fat metabolism regulation adult humans average between about regulaation kg, metaholism vary greatly depending on gender metabolizm individual disposition.

The metabolis, or so of glycogen stored Fat metabolism regulation the meetabolism is depleted within one day regukation starvation.

Fatty acids are broken down to acetyl-CoA by means regulatipn beta oxidation inside the mitochondria, regulstion fatty acids are Caloric intake and emotional eating from acetyl-CoA mtabolism the mitochondria, in the cytosol.

The two pathways are distinct, not only in where metabolisj occur, but Thermogenic fat loss supplements in the reactions that occur, and the substrates that are used. The two pathways are mutually inhibitory, preventing the acetyl-CoA produced by beta-oxidation from entering the synthetic pathway via the acetyl-CoA carboxylase reaction.

During each turn of the cycle, two carbon atoms leave the cycle as CO 2 in the decarboxylation reactions catalyzed by isocitrate dehydrogenase and alpha-ketoglutarate dehydrogenase.

Thus each turn of the citric acid cycle oxidizes an acetyl-CoA unit while regenerating the oxaloacetate molecule with which the acetyl-CoA had originally combined to form citric acid. The decarboxylation reactions occur before malate is formed in the cycle. However, acetyl-CoA can be converted to acetoacetate, which can decarboxylate to acetone either spontaneously, or catalyzed by acetoacetate decarboxylase.

Acetol can be converted to propylene glycol. This converts to pyruvate by two alternative enzymesor propionaldehydeor to L -lactaldehyde then L -lactate the common lactate isomer.

The first experiment to show conversion of acetone to glucose was carried out in This, and further experiments used carbon isotopic labelling. The glycerol released into the blood during the lipolysis of triglycerides in adipose tissue can only be taken up by the liver.

Here it is converted into glycerol 3-phosphate by the action of glycerol kinase which hydrolyzes one molecule of ATP per glycerol molecule which is phosphorylated. Glycerol 3-phosphate is then oxidized to dihydroxyacetone phosphatewhich is, in turn, converted into glyceraldehyde 3-phosphate by the enzyme triose phosphate isomerase.

From here the three carbon atoms of the original glycerol can be oxidized via glycolysisor converted to glucose via gluconeogenesis. Fatty acids are an integral part of the phospholipids that make up the bulk of the plasma membranesor cell membranes, of cells.

These phospholipids can be cleaved into diacylglycerol DAG and inositol trisphosphate IP 3 through hydrolysis of the phospholipid, phosphatidylinositol 4,5-bisphosphate PIP 2by the cell membrane bound enzyme phospholipase C PLC. One product of fatty acid metabolism are the prostaglandinscompounds having diverse hormone -like effects in animals.

Prostaglandins have been found in almost every tissue in humans and other animals. They are enzymatically derived from arachidonic acid, a carbon polyunsaturated fatty acid.

Every prostaglandin therefore contains 20 carbon atoms, including a 5-carbon ring. They are a subclass of eicosanoids and form the prostanoid class of fatty acid derivatives. The prostaglandins are synthesized in the cell membrane by the cleavage of arachidonate from the phospholipids that make up the membrane.

This is catalyzed either by phospholipase A 2 acting directly on a membrane phospholipid, or by a lipase acting on DAG diacyl-glycerol. The arachidonate is then acted upon by the cyclooxygenase component of prostaglandin synthase.

This forms a cyclopentane ring roughly in the middle of the fatty acid chain. The reaction also adds 4 oxygen atoms derived from two molecules of O 2. The resulting molecule is prostaglandin G 2which is converted by the hydroperoxidase component of the enzyme complex into prostaglandin H 2.

This highly unstable compound is rapidly transformed into other prostaglandins, prostacyclin and thromboxanes. If arachidonate is acted upon by a lipoxygenase instead of cyclooxygenase, Hydroxyeicosatetraenoic acids and leukotrienes are formed.

They also act as local hormones. Prostaglandins have two derivatives: prostacyclins and thromboxanes. Prostacyclins are powerful locally acting vasodilators and inhibit the aggregation of blood platelets.

Through their role in vasodilation, prostacyclins are also involved in inflammation. They are synthesized in the walls of blood vessels and serve the physiological function of preventing needless clot formation, as well as regulating the contraction of smooth muscle tissue. Their name comes from their role in clot formation thrombosis.

A significant proportion of the fatty acids in the body are obtained from the diet, in the form of triglycerides of either animal or plant origin. The fatty acids in the fats obtained from land animals tend to be saturated, whereas the fatty acids in the triglycerides of fish and plants are often polyunsaturated and therefore present as oils.

These triglycerides cannot be absorbed by the intestine. The activated complex can work only at a water-fat interface. Therefore, it is essential that fats are first emulsified by bile salts for optimal activity of these enzymes.

the fat soluble vitamins and cholesterol and bile salts form mixed micellesin the watery duodenal contents see diagrams on the right. The contents of these micelles but not the bile salts enter the enterocytes epithelial cells lining the small intestine where they are resynthesized into triglycerides, and packaged into chylomicrons which are released into the lacteals the capillaries of the lymph system of the intestines.

This means that the fat-soluble products of digestion are discharged directly into the general circulation, without first passing through the liver, unlike all other digestion products. The reason for this peculiarity is unknown. The chylomicrons circulate throughout the body, giving the blood plasma a milky or creamy appearance after a fatty meal.

The fatty acids are absorbed by the adipocytes [ citation needed ]but the glycerol and chylomicron remnants remain in the blood plasma, ultimately to be removed from the circulation by the liver. The free fatty acids released by the digestion of the chylomicrons are absorbed by the adipocytes [ citation needed ]where they are resynthesized into triglycerides using glycerol derived from glucose in the glycolytic pathway [ citation needed ].

These triglycerides are stored, until needed for the fuel requirements of other tissues, in the fat droplet of the adipocyte. The liver absorbs a proportion of the glucose from the blood in the portal vein coming from the intestines. After the liver has replenished its glycogen stores which amount to only about g of glycogen when full much of the rest of the glucose is converted into fatty acids as described below.

These fatty acids are combined with glycerol to form triglycerides which are packaged into droplets very similar to chylomicrons, but known as very low-density lipoproteins VLDL. These VLDL droplets are processed in exactly the same manner as chylomicrons, except that the VLDL remnant is known as an intermediate-density lipoprotein IDLwhich is capable of scavenging cholesterol from the blood.

This converts IDL into low-density lipoprotein LDLwhich is taken up by cells that require cholesterol for incorporation into their cell membranes or for synthetic purposes e.

the formation of the steroid hormones. The remainder of the LDLs is removed by the liver. Adipose tissue and lactating mammary glands also take up glucose from the blood for conversion into triglycerides.

This occurs in the same way as in the liver, except that these tissues do not release the triglycerides thus produced as VLDL into the blood. All cells in the body need to manufacture and maintain their membranes and the membranes of their organelles.

Whether they rely entirely on free fatty acids absorbed from the blood, or are able to synthesize their own fatty acids from blood glucose, is not known. The cells of the central nervous system will almost certainly have the capability of manufacturing their own fatty acids, as these molecules cannot reach them through the blood brain barrier.

Much like beta-oxidationstraight-chain fatty acid synthesis occurs via the six recurring reactions shown below, until the carbon palmitic acid is produced. The diagrams presented show how fatty acids are synthesized in microorganisms and list the enzymes found in Escherichia coli.

FASII is present in prokaryotesplants, fungi, and parasites, as well as in mitochondria. In animals as well as some fungi such as yeast, these same reactions occur on fatty acid synthase I FASIa large dimeric protein that has all of the enzymatic activities required to create a fatty acid.

FASI is less efficient than FASII; however, it allows for the formation of more molecules, including "medium-chain" fatty acids via early chain termination. by transferring fatty acids between an acyl acceptor and donor. They also have the task of synthesizing bioactive lipids as well as their precursor molecules.

Elongation, starting with stearateis performed mainly in the endoplasmic reticulum by several membrane-bound enzymes. The enzymatic steps involved in the elongation process are principally the same as those carried out by fatty acid synthesisbut the four principal successive steps of the elongation are performed by individual proteins, which may be physically associated.

: Fat metabolism regulation

Fatty acid metabolism - Wikipedia Role of bile acid malabsorption Metabopism pathogenesis of diarrhea and steatorrhea in patients Fat metabolism regulation ileal resection. Jetabolism activation by TZD may relieve oxidative stress in β-cells of diabetic animals [ ], leading to preservation of β-cell mass [— ] and partial improvement in glucose-stimulated insulin secretion from isolated islets [ ]. Verges B: Clinical interest of PPAR ligands. Nagashima, T. Regul Toxicol Pharmacol.
Obesity and the regulation of fat metabolism - WormBook - NCBI Bookshelf The transcriptional basis of adipocyte development Prostaglandins Leukot. I am indebted to members of the Ashrafi lab and Jennifer Watts for discussions. Deletions in C. Trends Biochem Sci 32, 71— Comparative genomics and functional study of lipid metabolic genes in Caenorhabditis elegans. Dgat1 and Dgat2 regulate enterocyte triacylglycerol distribution and alter proteins associated with cytoplasmic lipid droplets in response to dietary fat.
Key points Cholesterol esterification Fat metabolism regulation ACAT2 is essential for efficient Balanced breakfast recipes Fat metabolism regulation absorption: evidence from thoracic metzbolism duct metabollism. New genetic loci implicated in regulatjon glucose homeostasis and their impact on type 2 diabetes risk. Khoriaty, R. Colesevelam hydrochloride cholestagel : a new, potent bile acid sequestrant associated with a low incidence of gastrointestinal side effects. Uptake and metabolism of circulating fatty acids by rat intestine. Direct binding of cholesterol to the purified membrane region of SCAP. CAS PubMed Google Scholar Donaldson, G.

Fat metabolism regulation -

Clark RB, Bishop-Bailey D, Estrada-Hernandez T, Hla T, Puddington L, Padula SJ: The nuclear receptor PPARγ and immunoregulation. PPARγ mediates inhibition of helper T cell responses. J Immunol. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM: PPARγ is required for the differentiation of adipose tissue in vivo and in vitro.

Vidal-Puig A, Jimenez-Linan M, Lowell BB, Hamann A, Hu E, Spiegelman B, Flier JS, Moller DE: Regulation of PPAR gamma gene expression by nutrition and obesity in rodents. Tontonoz P, Hu E, Spiegelman BM: Regulation of adipocyte gene expression and differentiation by peroxisome proliferator activated receptor γ.

Curr Opin Genet Dev. Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J: Coordinate regulation of the expression of the fatty acid transport protein and acyl-CoA synthetase genes by PPAR α and PPAR γ activators.

Okuno A, Tamemoto H, Tobe K, Ueki K, Mori Y, Iwamoto K, Umesono K, Akanuma Y, Fujiwara T, Horikoshi H, Yazaki Y, Kadowaki T: Troglitazone increases the number of small adipocytes without the change of white adipose tissue mass in obese Zucker rats.

Hotamisligil GS, Shargill NS, Spiegelman BM: Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Walczak R, Tontonoz P: PPARadigms and PPARadoxes: expanding roles for PPAR γ in the control of lipid metabolism.

J Lipid Res. Kallen CB, Lazar MA: Antidiabetic thiazolidinediones inhibit leptin ob gene expression in 3T3-L1 adipocytes. De Vos P, Lefebvre AM, Miller SG, Guerre-Millo M, Wong KM, Saladin R, Hamann LG, Staels B, Briggs MR, Auwerx J: Thiazolidinediones repress ob gene expression in rodents via activation of peroxisome proliferator-activated receptor γ.

Jiang C, Ting AT, Seed B: PPARγ agonists inhibit production of monocyte inflammatory cytokines. Elstner E, Muller C, Koshizuka K, Williamson EA, Park D, Asou H, Shintaku P, Said JW, Heber D, Koeffler HP: Ligands for peroxisome proliferator-activated receptorγ and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice.

Gimble JM, Pighetti GM, Lerner MR, Wu X, Lightfoot SA, Brackett DJ, Darcy K, Hollingsworth AB: Expression of peroxisome proliferator-activated receptor mRNA in normal and tumorigenic mammary glands. Zhou Y-T, Shimabukuro M, Wang M-Y, Lee Y, Higa M, Milburn JL, Newgard CB, Unger RH: Role of peroxisome proliferator-activated receptor α in disease of pancreatic β cells.

Dubois M, Paltou F, Kerr-Conte J, Gyr V, Vandewalle B, Desreumaux P, Auwerx J, Schoonjans K, Lefebvre J: Expression of peroxisome proliferator-activated receptor γ in normal human pancreatic islet cells. Shimomura K, Shimizu H, Ikeda M, Okada S, Kakei M, Matsumoto S, Mori M: Fenofibrate, troglitazone, and deoxy-Δ12, prostaglandin J2 close K ATP channels and induce insulin secretion.

J Pharmacol Exp Ther. Roduit R, Morin J, Masse F, Segall L, Roche E, Newgard CB, Assimacopoulos-Jeannet F, Prentki M: Glucose down-regulates the expression of the peroxisome proliferator-activated receptor-α gene in the pancreatic β-cell. Laybutt R, Hasenkamp W, Groff A, Grey S, Jonas JC, Kaneto H, Sharma A, Bonner-Weir S, Weir G: Beta-cell adaptation to hyperglycemia.

Sugden MC, Bulmer K, Augustine D, Holness MJ: Selective modification of pyruvate dehydrogenase kinase isoform expression in rat pancreatic islets elicited by starvation and activation of peroxisome proliferator-activated receptor-α : implications for glucose-stimulated insulin secretion.

Sugden MC, Holness MJ: Potential role of peroxisome proliferator-activated receptor-α in the modulation of glucose-stimulated insulin secretion. Guerre-Millo M, Rouault C, Poulain P, Andre J, Poitout V, Peters JM, Gonzalez FJ, Fruchart JC, Reach G, Staels B: PPAR-α-null mice are protected from high-fat diet-induced insulin resistance.

Sugden MC, Bulmer K, Gibbons GF, Knight BL, Holness MJ: Peroxisome-proliferator-activated receptor-α PPARα deficiency leads to dysregulation of hepatic lipid and carbohydrate metabolism by fatty acids and insulin. Kersten S, Mandard S, Escher P, Gonzalez FJ, Tafuri S, Desvergne B, Wahli W: The peroxisome proliferator-activated receptor alpha regulates amino acid metabolism.

FASEB J. Maechler P, Wollheim CB: Mitochondrial glutamate acts as a messenger in glucose-induced insulin exocytosis. Unger RH, Orci L: Lipoapoptosis: its mechanism and its diseases. Biochim Biophys Acta. Prentki M, Joly E, El-Assaad W, Roduit R: Malonyl-CoA signaling, lipid partitioning, and glucolipotoxicity.

Role in β-cell adaptation and failure in the etiology of diabetes. Tordjman K, Standley KN, Bernal-Mizrachi C, Leone TC, Coleman T, Kelly DP, Semenkovich CF: PPARα suppresses insulin secretion and induces UCP2 in insulinoma cells.

Kim H, Haluzik M, Asghar Z, Yau D, Joseph JW, Fernandez AM, Reitman ML, Yakar S, Stannard B, Heron-Milhavet L, Wheeler MB, LeRoith D: Peroxisome proliferator-activated receptor-α agonist treatment in a transgenic model of type 2 diabetes reverses the lipotoxic state and improves glucose homeostasis.

Koh EH, Kim M-S, Park J-Y, Kim HS, Youn J-Y, Park H-S, Youn JH, Lee K-U: Peroxisome proliferator-activated receptor PPAR -α activation prevents diabetes in OLETF rats. Comparison with PPAR-γ activation. Kelly LJ, Vicario PP, Thompson GM, Candelore MR, Doebber TW, Ventre J, Wu MS, Meurer R, Forrest MJ, Conner MW, Cascieri MA, Molle DE: Peroxisome proliferator-acitvated receptors γ and α mediate in vivo regulation of uncoupling protein UCP-1, UCP-2, UCP-3 gene expression.

Schrauwen P, Hesselink M: UCP2 and UCP3 in muscle controlling body metabolism. J Exp Biol. Grav HJ, Tronstad KJ, Gudbrandsen OA, Berge K, Fladmark KE, Martinsen TC, Waldum H, Wergedahl H, Berge RK: Changed energy state and increased mitochondrial beta-oxidation rate in liver of rats associated with lowered proton electrochemical potential and stimulated uncoupling protein 2 UCP-2 expression: evidence for peroxisome proliferator-activated receptor-alpha independent induction of UCP-2 expression.

Chan CB, De Leo D, Joseph JW, McQuaid TS, Ha XF, Xu F, Tsushima RG, Pennefather PS, Salapatek AM, Wheeler MB: Increased uncoupling protein-2 levels in beta cells are associated with impaired glucose-stimulated insulin secretion: mechanism of action.

Hong Y, Fink BD, Dillon JS, Sivitz W: Effects of adenoviral overexpression of uncoupling protein-2 and -3 on mitochondrial respiration in insulinoma cells. Zhang CY, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, Hagen T, Vidal-Puig AJ, Boss O, Kim YB, Zheng XX, Wheeler MB, Shulman GI, Chan CB, Lowell BB: Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, beta cell dysfunction and type 2 diabetes.

Joseph JW, Koshkin V, Zhang CY, Wang J, Lowell BB, Chan CB, Wheeler MB: Uncoupling protein 2 knockout mice have enhanced insulin secretory capacity after a high fat diet. Patane G, Anello M, Piro S, Vigneri R, Purrello F, Rabuazzo AM: Role of ATP production and uncoupling protein-2 in the insulin secretory defect induced by chronic exposure to high glucose or free fatty acids and effects of peroxisome proliferator-activated receptor-γ inhibition.

Shimabukuro M, Zhou Y-T, Lee Y, Unger RH: Induction of uncoupling protein-2 mRNA by troglitazone in the pancreatic islets of Zucker diabetic fatty rats.

Sreenan S, Sturis J, Pugh W, Burant CF, Polonsky KS: Prevention of hyperglycemia in the Zucker diabetic fatty rat by treatment with metformin or troglitazone.

Parton LE, Diraison F, Neill SE, Ghosh SK, Rubino MA, Bisi JE, Briscoe CP, Rutter GA: Impact of PPARγ overexpression and activation on pancreatic islet gene expression profile analyzed with oligonucleotide microarrays.

Eto K, Yamashita T, Matsui J, Terauchi Y, Noda M, Kadowaki T: Genetic manipulation of fatty acid metabolism in β-cells are associated with dysregulated insulin secretion. Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T, Eto K, Tsubamoto Y, Okuno A, Murakami K, Sekihara H, Hasegawa G, Naito M, Toyoshima Y, Tanaka S, Shiota K, Kitamura T, Fujita T, Ezaki O, Aizawa S, Nagai R, Tobe K, Kimura S, Kadowaki T: PPAR-γ mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance.

Kim HI, Cha JY, Kim SY, Kim JW, Roh KJ, Seong JK, Lee NT, Choi KY, Kim KS, Ahn YH: Peroxisomal proliferator-activated receptor-gamma upregulates glucokinase gene expression in beta-cells.

Kim HI, Kim JW, Kim SH, Cha JY, Kim AS, Ahn YH: Identification and functional characterization of the peroxisomal proliferator response element in rat GLUT2 promoter.

Wang Z, Gleichmann H: Glut2 in pancreatic islets: crucial target molecule in diabetes induced with multiple low doses of streptozotocin in mice. Nakamichi Y, Kikuta T, Ito E, Ohara-Imaizumi M, Nishiwaki C, Ishida H, Nagamatsu S: PPAR-γ overexpression suppresses glucose-induced proinsulin biosynthesis and insulin release synergistically with pioglitazone in MIN6 cells.

Lupi R, Del Guerra S, Marselli L, Bugliani M, Boggi U, Mosca F, Marchetti P, Del Prato S: Rosiglitazone prevents the impairment of human islet function induced by fatty acids: evidence for a role of PPARγ2 in the modulation of insulin secretion. Diani AR, Sawada G, Wyse B, Marray FT, Khan M: Pioglitazone preserves pancreatic islet structure and insulin secretory funciton in three murine models of type 2 diabetes.

Higa M, Zhou Y-T, Ravazzola M, Baetens D, Orci L, Unger RH: Troglitazone prevents mitochondrial alterations, β cell destruction and diabetes in obese prediabetic rats.

Beales PE, Liddi R, Giorgini AE, Signore A, Procaccini E, Batchelor K, Pozzilli P: Troglitazone prevents insulin dependent diabetes in the non-obese diabetic mouse.

Pershadsingh HA: Peroxisome proliferator-activated receptor-γ: therapeutic target for diseases beyond diabetes: quo vadis?. Expert Opin Investig Drugs. Rosen ED, Kulkarni RN, Sarraf P, Ozcan U, Okada T, Hsu C-H, Eisenman D, Magnuson MA, Gonzalez FJ, Kahn CR, Spiegelman BM: Targeted elimination of peroxisome proliferator-activated receptor γ in β cells leads to abnormalities in islet mass without compromising glucose homeostasis.

Mol Cell Biol. Foufelle F, Ferre P: New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c.

Horton JD, Goldstein JL, Brown MS: SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Kakuma T, Lee Y, Higa M, Wang Z, Pan W, Shimomura I, Unger RH: Leptin, troglitazone, and the expression of sterol regulatory element binding proteins in liver and pancreatic islets.

Andreolas C, da Silva Xavier G, Diraison F, Zhao C, Varadi A, Lopez-Casillas F, Ferre P, Foufelle F, Rutter GA: Stimulation of acetyl-CoA carboxylase gene expression by glucose requires insulin release and sterol regulatory element binding protein 1c in pancreatic MIN6 beta-cells.

Wang H, Maechler P, Antinozzi PA, Herrero L, Hagenfeldt-Johansson KA, Bjorklund A, Wollheim CB: The transcription factor SREBP-1c is instrumental in the development of beta-cell dysfunction. Diraison F, Parton L, Ferre P, Foufelle F, Briscoe CP, Leclerc I, Rutter GA: Over-expression of sterol-regulatory-element-binding protein-1c SREBP1c in rat pancreatic islets induces lipogenesis and decreases glucose-stimulated insulin release: modulation by 5-aminoimidazolecarboxamide ribonucleoside AICAR.

Medvedev AV, Robidoux J, Bai X, Cao W, Floering LM, Daniel KW, Collins S: Regulation of the uncoupling protein-2 gene in INS-1 β-cells by oleic acid. Yamashita T, Eto K, Okazaki Y, Yamashita S, Yamauchi T, Sekine N, Nagai R, Noda M, Kadowaki T: Role of uncoupling protein-2 up-regulation and triglyceride accumulation in impaired glucose-stimulated insulin secretion in a β-cell lipotoxicity model overexpressing sterol regulatory element binding protein-1c.

Poitout V: β-cell lipotoxicity: burning fat into heat?. Elholm M, Bjerking G, Knudsen J, Kristiansen K, Mandrup S: Regulatory elements in the promoter region of the rat gene encoding the acyl-CoA-binding protein.

Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, Wahli W: The PPARα-leukotriene B4 pathway to inflammation control. Varanasi U, Chu R, Huang Q, Castellon R, Yeldandi AV, Reddy JK: Identification of a peroxisome proliferator-responsive element upstream of the human peroxisomal fatty acyl coenzyme A oxidase gene.

Karam WG, Ghanayem BI: Induction of replication DNA synthesis and PPARα-dependent gene transcription by Wy in primary rat hepatocyte and non-parenchymal cell co-cultures. Berthou L, Duverger N, Emmanuel F, Langouet S, Auwerx J, Guillouzo A, Fruchart JC, Rubin E, Denefle P, Staels B, Branellec D: Opposite regulation of human versus mouse apolipoprotein A-I by fibrates in human apolipoprotein A-I transgenic mice.

Raspe E, Madsen L, Lefebvre AM, Leitersdorf I, Gelman L, Peinado-Onsurbe J, Dallongeville J, Fruchart JC, Berge R, Staels B: Modulation of rat liver apolipoprotein gene expression and serum lipid levels by tetradecylthioacetic acid TTA via PPARα activation.

Vu-Dac N, Schoonjans K, Kosykh V, Dallongeville J, Fruchart JC, Staels B, Auwerx J: Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor.

Prieur X, Coste H, Rodriguez JC: The human apolipoprotein AV gene is regulated by PPAR α and contains a novel FXR response element. Hertz R, Bishara-Shieban J, Bar-Tana J: Mode of action of peroxisome proliferators as hypolipidemic drugs.

Suppression of apolipoprotein C-III. Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne B, Wahli W: Peroxisome proliferator-activated receptor α mediates the adaptive response to fasting. Leone TC, Weinheimer CJ, Kelly DP: A critical role for the peroxisome proliferator-activated receptor α in the cellular fasting response: the PPARα-null mouse as a model of fatty acid oxidation disorders.

Aoyama T, Peters JM, Iritani N, Nakajuima T, Furihata K, Hashimoto T, Gonzalez FJ: Altered constitutive expression of fatty acid-metabolizing enzymes in mice lacking the peroxisome proliferator-activated receptor α.

Minnich A, Tian N, Byan L, Bilder G: A potent PPARα agonist stimulates mitochondrial fatty acid β-oxidation in liver and skeletal muscle. Kroetz DL, Yook P, Costet P, Bianchi P, Pineau T: Peroxisome proliferator-activated receptor α controls the hepatic CYP4A induction adaptive response to starvation and diabetes.

Aldridge TC, Tugwood JD, Green S: Identification and characterization of DNA elements implicated in the regulation of CYP4A1 transcription. Yu S, Cao WQ, Kashireddy P, Meyer K, Jia Y, Hughes DE, Tan Y, Feng J, Yeldandi AV, Rao MS, Costa RH, Gonzalez FJ, Reddy JK: Human peroxisome proliferator-activated receptor α supports the induction of peroxisome proliferation in PPARα-deficient mouse liver.

Patel DD, Knight BL, Soutar AK, Gibbons GF, Wade DP: The effect of peroxisome-proliferator-activated receptor-α on the activity of the cholesterol 7 α-hydroxylase gene. Cheema SK, Agellon LB: The murine and human cholesterol 7 α-hydroxylase gene promoters are differentially responsive to regulation by fatty acids mediated via peroxisome proliferator-activated receptor α.

Guillou H, Martin P, Jan S, D'Andrea S, Roulet A, Catheline D, Rioux V, Pineaut T, Legrand P: Comparative effect of fenofibrate on hepatic desaturases in wild-type and peroxisome proliferator-activated receptor α-deficient mice.

Wolfrum C, Ellinghaus P, Fobker M, Seedorf U, Assmann G, Borchers T, Spener F: Phytanic acid is ligand and transcriptional activator of muring liver fatty acid binding protein.

Motojima K, Passilly P, Peters JM, Gonzalez FJ, Latruffe N: Expression of putative fatty acid transporter genes are regulated by peroxisome proliferator-activated receptor α and γ activators in a tissue- and inducer-specific manner. Yamazaki K, Kuromitsu J, Tanaka I: Microarray analysis of gene expression changes in mouse liver induced by peroxisome proliferator-activated receptor α agonists.

Kok T, Wolters H, Bloks W, Havinga R, Jansen PL, Staels B, Kuipers F: Induction of hepatic ABC transporter expression is part of the PPARα-mediated fasting response in the mouse.

Tobin KA, Steineger HH, Alberti S, Spydevold O, Auwerx J, Gustafsson JA, Nebb HI: Cross talk between fatty acid and cholesterol metabolism mediated by liver X receptor-α.

Hsu MH, Savas U, Griffin KJ, Johnson EF: Identification of peroxisome proliferator-responsive human genes by elevated expression of the peroxisome proliferator-activated receptor α in HepG2 cells.

Toxicol Appl Pharmacol. Wan YJ, Cai Y, Lungo W, Fu P, Locker J, French S, Sucov HM: Peroxisome proliferator-activated receptor a-mediated pathways are altered in hepatocyte-specific retinoid X receptor a-deficient mice.

Hegardt FG: Transcriptional regulation of mitochondrial HMG-CoA synthase in the control of ketogenesis. Bouly M, Masson D, Gross B, Jiang XC, Fievet C, Castro G, Tall AR, Fruchart JC, Staels B, Lagrost L, Luc G: Induction of the phospholipid transfer protein gene accounts for the high density lipoprotein enlargement in mice treated with fenofibrate.

Miller CW, Ntambi JM: Peroxisome proliferators induce mouse liver stearoyl-CoA desaturase 1 gene expression. Latruffe N, Nicolas-Frances V, Dasari VK, Osumi T: Studies on regulation of the peroxisomal beta-oxidation at the 3-ketothiolase step.

Dissection of the rat liver thiolase B gene promoter. Adv Exp Med Biol. Hertz R, Seckbach M, Zakin MM, Bar-Tana J: Transcriptional suppression of the transferrin gene by hypolipidemic peroxisome proliferators. Download references. Research by the authors' group is supported by the Canadian Institutes for Health Research and the Canadian Diabetes Association.

CBC holds a Levesque Research Chair in Nutrisciences and Health at the University of Prince Edward Island. The authors thank MB Wheeler and MC Saleh for reading the manuscript and for their helpful comments.

Department of Biomedical Sciences, University of Prince Edward Island, University Avenue, Charlottetown, PE, C1A 4P3, Canada. You can also search for this author in PubMed Google Scholar. Correspondence to Catherine B Chan.

Reprints and permissions. Fatehi-Hassanabad, Z. Transcriptional regulation of lipid metabolism by fatty acids: a key determinant of pancreatic β-cell function.

Nutr Metab Lond 2 , 1 Download citation. Received : 20 October Accepted : 05 January Published : 05 January Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative.

Skip to main content. Search all BMC articles Search. Download PDF. Download ePub. Abstract Background Optimal pancreatic β-cell function is essential for the regulation of glucose homeostasis in both humans and animals and its impairment leads to the development of diabetes.

Results Free fatty acids represent an important factor linking excess fat mass to type 2 diabetes. Conclusion The role of the PPARs and SREBP-1c as potential mediators of lipotoxicity is an emerging area of interest.

Introduction Fatty acids are physiologically important both structurally, as components of phospholipids and glycolipids, as well as functionally, as fuel molecules. Figure 1. Full size image. Metabolism of fatty acids in the beta cell and insulin secretion Fatty acids, not glucose, are the major endogenous energy source for unstimulated islets [ 10 ].

Transcriptional regulation of free fatty acid metabolism Free fatty acid metabolism responds to varying metabolic states partially by induction of enzymes that promote either catabolic or anabolic processes. Peroxisome proliferator-activated receptors The PPARs form a subfamily in the nuclear receptor superfamily.

PPARα PPARα was the first member of this nuclear receptor subclass to be described. Figure 2. Table 1 Selected hepatic PPARα regulated genes with at least one functional peroxisome proliferator receptor element PPRE identified within the promoter sequence Full size table. Peroxisome proliferator-activated receptors and β-cell function Both PPARα and PPARγ have been detected in pancreatic β-cells [ 76 , 77 ].

Sterol regulatory element binding protein The family of SREBPs governs transcriptional activation of a large number of genes involved in regulation of lipid metabolism, including lipogenesis, cholesterol transport and synthesis [ ].

Conclusions FFA exert dual effects on insulin secretion, dependent on the duration of exposure. References Kutchai HC: Digestion and absorption. Google Scholar Oxidation of fatty acids. Google Scholar Zimmet P, Alberti KG, Shaw J: Global and societal implications of the diabetes epidemic.

Article CAS Google Scholar Centers for Disease Control and Prevention: National Diabetes Fact Sheet: National estimate and general information on diabetes in the United States, Article CAS Google Scholar Burke JP, Williams K, Gaskill SP, Hazuda HP, Haffner SM, Stern MP: Rapid rise in the incidence of type 2 diabetes from to results from the San Antonio Heart Study.

Article CAS Google Scholar Ludvik B, Nolan JJ, Baloga J, Sacks D, Olefsky J: Effect of obesity on insulin resistance in normal subjects and patients with NIDDM. Article CAS Google Scholar Fraze E, Donner CC, Swislocki AL, Chiou YA, Chen YD, Reaven GM: Ambient plasma free fatty acid concentrations in noninsulin-dependent diabetes mellitus: evidence for insulin resistance.

Article CAS Google Scholar McGarry JD: What if Minkowski had been ageusic? Article CAS Google Scholar Malaisse WJ, Best L, Kawazu S, Malaisse-Lagae F, Sener A: The stimulus-secretion coupling of glucose-induced insulin release: fuel metabolism in islet deprived of exogenous nutrient.

Article CAS Google Scholar Hellerstrom C: Effects of carbohydrates on the oxygen consumption of isolated pancreatic islets of mice. Article CAS Google Scholar Vara E, Tamarit-Rodriguez J: Glucose stimulation of insulin secretion in islets of fed and starved rats and its dependence on lipid metabolism.

Article CAS Google Scholar Malaisse WJ: Insulin secretion: Multifactorial regulation for a single process of release. Article CAS Google Scholar Carpinelli AR, Curi R, Malaisse WJ: Long-term regulation of pancreatic B-cell responsiveness to D-glucose by food availability, feeding schedule and diet composition.

Article CAS Google Scholar Newgard CB, McGarry JD: Metabolic coupling factors in pancreatic β cell signal transduction.

Article CAS Google Scholar Warnotte C, Gilon P, Nenquin M, Henquin JC: Mechanisms of the stimulation of insulin release by saturated fatty acids: a study of palmitate effects in mouse β-cells.

Article CAS Google Scholar Stein DT, Esser V, Stevenson BE, Lane KE, Whiteside JH, Daniels MB, Chen S, McGarry JD: Essentially of circulating fatty acids for glucose-stimulated insulin secretion in fasted rats.

Article CAS Google Scholar McGarry JD, Dobbins RL: Fatty acids, lipotoxicity and insulin secretion. Article CAS Google Scholar Hamilton JA, Civelek VN, Kamp F, Tornheim K, Corkey BE: Changes in internal pH caused by movement of fatty acids into and out of clonal pancreatic β-cells HIT.

CAS Google Scholar Matschinsky FM: A lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Article CAS Google Scholar McGarry JD: Dysregulation of fatty acid metabolism in the etiology of type 2 diabetes.

Article CAS Google Scholar Berne C: The metabolism of lipid in mouse pancreatic islets. Article CAS Google Scholar Boylan JG, Hamilton JA: Interactions of acyl-coenzyme A with phosphatidylcholine bilayers and serum albumin. Article CAS Google Scholar Corkey B: Analysis of acyl-coenzyme A esters in biological samples.

Article CAS Google Scholar Mason TM, Goh T, Tchipashvili V, Sandhu H, Gupta N, Lewis GF, Giacca A: Prolonged elevation of plasma free fatty acids desensitizes the insulin secretory response to glucose in vivo in rats. Article CAS Google Scholar Unger RH: How obesity causes diabetes in Zucker diabetic fatty rats.

Article Google Scholar Carpentier A, Mittelman SD, Lamarche B, Bergman RN, Giacca A, Lewis GF: Acute enhancement of insulin secretion by FFA in humans is lost with prolonged FFA elevation. CAS Google Scholar Carpentier A, Mittelman SD, Bergman RN, Giacca A, Lewis GF: Prolonged elevation of plasma free fatty acids impairs pancreatic B-cell function in obese nondiabetic humans but not in individuals with type 2 diabetes.

Article CAS Google Scholar Kashyap SR, Belfort R, Berria R, Suraamornkul S, Pratipranawatr T, Finlayson J, Barrentine A, Bajaj M, Mandarino L, DeFronzo R, Cusi K: Discordant effects of a chronic physiological increase in plasma FFA on insulin signaling in healthy subjects with or without a family history of type 2 diabetes.

Article CAS Google Scholar Paolisso G, Gambardella A: Opposite effects of short and long-term fatty acid infusion on insulin secretion in healthy subjects. Article CAS Google Scholar Stefan N, Fritsche A, Haring H, Stumvoll M: Effect of experimental elevation of free fatty acids on insulin secretion and insulin sensitivity in healthy carriers of the Pro12 Ala polymorphism of the peroxisome proliferator-activated receptor γ2 gene.

Article CAS Google Scholar Paolisso G, Tagliamonte MR, Rizzo MR, Gualdiero P, Saccomanno F, Gambardella A, Giugliano D, D'Onofrio F, Howard BV: Lowering fatty acids potentiates acute insulin response in first degree relatives of people with type II diabetes.

Article CAS Google Scholar Qvigstad E, Mostad IL, Bjerve KS, Grill VE: Acute lowering of circulating fatty acids improves insulin secretion in a subset of type 2 diabetes subjects. Article CAS Google Scholar Unger RH: Lipotoxic diseases. Article CAS Google Scholar Shimabukuro M, Zhou YT, Levi M, Unger RH: Fatty acid induced β-cell apoptosis: a link between diabetes and obesity.

Article CAS Google Scholar Lupi R, Dotta F, Marselli L, Del Guerra S, Masini M, Santangelo C, Patane G, Boggi U, Piro S, Anello M, Bergamini E, Mosca F, Di Mario U, Del Prato S, Marchetti P: Prolonged exposure to free fatty acids has cytostatic and pro-apoptotic effects on human pancreatic islets: Evidence that cell death is caspase mediated, partially dependent on ceramide pathway, and Bcl-2 regulated.

Article CAS Google Scholar Kersten S: Effects of fatty acids on gene expression: role of peroxisome proliferator-activated receptor α, liver X receptor α and sterol regulatory element-binding protein-1 c. Article CAS Google Scholar Reddy JK, Chu R: Peroxisome proliferator-induced pleiotropic responses: Pursuit of a phenomenon.

Article CAS Google Scholar Goldfischer SL: Peroxisomal diseases. CAS Google Scholar Rosen ED, Spiegelman BM: PPARγ: a nuclear regulator of metabolism, differentiation, and cell growth.

Article CAS Google Scholar Sher T, Yi H-F, McBride OW, Gonzalez FJ: cDNA cloning, chromosomal mapping, and functional characterization of the human peroxisome proliferator activated receptor.

Article CAS Google Scholar Braissant O, Foufelle F, Scotto C, Dauca M, Wahli W: Differential expression of peroxisome proliferator activated receptor PPARs : tissue distribution of PPAR-α, -β and -γ in the adult rat. CAS Google Scholar Auboeuf D, Rieusset J, Fajas L, Vallier P, Frering V, Riou JP, Staels B, Auwerx J, Laville M, Vidal H: Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor α in humans.

Article CAS Google Scholar Chinetti G, Griglio S, Antonucci M, Torra IP, Delerive P, Majd Z, Fruchart JC, Chapman J, Najib J, Staels B: Activation of proliferator-activated receptors α and γ induces apoptosis of human monocyte-derived macrophages.

Article CAS Google Scholar Inoue I, Shino K, Noji S, Awata T, Katayama S: Expression of peroxisome proliferator-activated receptor α in primary cultures of human vascular endothelial cells. Article CAS Google Scholar Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, Najib J, Maclouf J, Tedgui A: Activation of human aortic smooth-muscle cells is inhibited by PPARα but not PPARγ activators.

Article CAS Google Scholar Gottlicher M, Widmark E, Li Q, Gustafsson JA: Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Article CAS Google Scholar Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J: Coordinate regulation of the expression of the fatty acid transport protein and acyl-CoA synthetase genes by PPARα and PPARγ activators.

Article CAS Google Scholar Dreyer C, Krey G, Keller H, Givel F, Helftenbein G, Wahli W: Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors.

Article CAS Google Scholar Kersten S: Peroxisome proliferator activated receptors and obesity. Article CAS Google Scholar Issemann I, Green S: Activation of a member of the steroid hormone receptor superfamily by some peroxisome proliferators.

Article CAS Google Scholar Cattley RC, DeLuca J, Elcombe C, Fenner-Crisp P, Lake BG, Marsman DS, Pastoor TA, Popp JA, Robinson DE, Schwetz B, Tugwood J, Wahli W: Do peroxisome proliferating compounds pose a hepatocarcinogenic hazard to humans?.

Article CAS Google Scholar Desvergne B, Wahli W: Peroxisome proliferator-activated receptors: nuclear control of metabolism. CAS Google Scholar Verges B: Clinical interest of PPAR ligands.

Article CAS Google Scholar Schmidt A, Endo N, Rutledge SJ, Vogel R, Shinar D, Rodan GA: Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acid.

CAS Google Scholar Kliewer SA, Forman BM, Blumberg B, Ong ES, Borgmeyer U, Mangelsdorf DJ, Umesono K, Evans RM: Differential expression and activation of a family of murine peroxisome proliferator-activated receptors.

Article CAS Google Scholar Escher P, Braissant O, Basu-Modak S, Michalik L, Wahli W, Desvergne B: Rat PPARs: quantitative analysis in adult rat tissues and regulation in fasting and refeeding. Article CAS Google Scholar Bastie C, Holst D, Gaillard D, Jehl-Pietri C, Grimaldi PA: Expression of peroxisome proliferator-activated receptor PPARδ promotes induction of PPARγ and adipocyte differentiation in 3T3C2 fibroblasts.

Article CAS Google Scholar Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, Sternbach DD, Lehmann JM, Wisely GB, Willson TM, Kliewer SA, Milburn MV: Molecular recognition of fatty acids by peroxisome proliferator-activated receptors.

Article CAS Google Scholar Gaw A, Packard CJ, Shepherd J: Fibrates. Article CAS Google Scholar Law RE, Goetze S, Xi XP, Jackson S, Kawano Y, Demer L, Fishbein MC, Meehan WP, Hsueh WA: Expression and function of PPARγ in rat and human vascular smooth muscle cells.

Article CAS Google Scholar Clark RB, Bishop-Bailey D, Estrada-Hernandez T, Hla T, Puddington L, Padula SJ: The nuclear receptor PPARγ and immunoregulation. Article CAS Google Scholar Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM: PPARγ is required for the differentiation of adipose tissue in vivo and in vitro.

Article CAS Google Scholar Vidal-Puig A, Jimenez-Linan M, Lowell BB, Hamann A, Hu E, Spiegelman B, Flier JS, Moller DE: Regulation of PPAR gamma gene expression by nutrition and obesity in rodents.

Article CAS Google Scholar Tontonoz P, Hu E, Spiegelman BM: Regulation of adipocyte gene expression and differentiation by peroxisome proliferator activated receptor γ. Article CAS Google Scholar Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J: Coordinate regulation of the expression of the fatty acid transport protein and acyl-CoA synthetase genes by PPAR α and PPAR γ activators.

Article CAS Google Scholar Okuno A, Tamemoto H, Tobe K, Ueki K, Mori Y, Iwamoto K, Umesono K, Akanuma Y, Fujiwara T, Horikoshi H, Yazaki Y, Kadowaki T: Troglitazone increases the number of small adipocytes without the change of white adipose tissue mass in obese Zucker rats.

Article CAS Google Scholar Hotamisligil GS, Shargill NS, Spiegelman BM: Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance.

Article CAS Google Scholar Walczak R, Tontonoz P: PPARadigms and PPARadoxes: expanding roles for PPAR γ in the control of lipid metabolism. CAS Google Scholar Kallen CB, Lazar MA: Antidiabetic thiazolidinediones inhibit leptin ob gene expression in 3T3-L1 adipocytes.

Article CAS Google Scholar De Vos P, Lefebvre AM, Miller SG, Guerre-Millo M, Wong KM, Saladin R, Hamann LG, Staels B, Briggs MR, Auwerx J: Thiazolidinediones repress ob gene expression in rodents via activation of peroxisome proliferator-activated receptor γ.

Article CAS Google Scholar Jiang C, Ting AT, Seed B: PPARγ agonists inhibit production of monocyte inflammatory cytokines. Article CAS Google Scholar Elstner E, Muller C, Koshizuka K, Williamson EA, Park D, Asou H, Shintaku P, Said JW, Heber D, Koeffler HP: Ligands for peroxisome proliferator-activated receptorγ and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice.

Article CAS Google Scholar Gimble JM, Pighetti GM, Lerner MR, Wu X, Lightfoot SA, Brackett DJ, Darcy K, Hollingsworth AB: Expression of peroxisome proliferator-activated receptor mRNA in normal and tumorigenic mammary glands.

Article CAS Google Scholar Zhou Y-T, Shimabukuro M, Wang M-Y, Lee Y, Higa M, Milburn JL, Newgard CB, Unger RH: Role of peroxisome proliferator-activated receptor α in disease of pancreatic β cells.

Article CAS Google Scholar Dubois M, Paltou F, Kerr-Conte J, Gyr V, Vandewalle B, Desreumaux P, Auwerx J, Schoonjans K, Lefebvre J: Expression of peroxisome proliferator-activated receptor γ in normal human pancreatic islet cells. Article CAS Google Scholar Shimomura K, Shimizu H, Ikeda M, Okada S, Kakei M, Matsumoto S, Mori M: Fenofibrate, troglitazone, and deoxy-Δ12, prostaglandin J2 close K ATP channels and induce insulin secretion.

Article CAS Google Scholar Roduit R, Morin J, Masse F, Segall L, Roche E, Newgard CB, Assimacopoulos-Jeannet F, Prentki M: Glucose down-regulates the expression of the peroxisome proliferator-activated receptor-α gene in the pancreatic β-cell.

Article CAS Google Scholar Laybutt R, Hasenkamp W, Groff A, Grey S, Jonas JC, Kaneto H, Sharma A, Bonner-Weir S, Weir G: Beta-cell adaptation to hyperglycemia. Article CAS Google Scholar Sugden MC, Bulmer K, Augustine D, Holness MJ: Selective modification of pyruvate dehydrogenase kinase isoform expression in rat pancreatic islets elicited by starvation and activation of peroxisome proliferator-activated receptor-α : implications for glucose-stimulated insulin secretion.

Article CAS Google Scholar Sugden MC, Holness MJ: Potential role of peroxisome proliferator-activated receptor-α in the modulation of glucose-stimulated insulin secretion. Article CAS Google Scholar Guerre-Millo M, Rouault C, Poulain P, Andre J, Poitout V, Peters JM, Gonzalez FJ, Fruchart JC, Reach G, Staels B: PPAR-α-null mice are protected from high-fat diet-induced insulin resistance.

Article CAS Google Scholar Sugden MC, Bulmer K, Gibbons GF, Knight BL, Holness MJ: Peroxisome-proliferator-activated receptor-α PPARα deficiency leads to dysregulation of hepatic lipid and carbohydrate metabolism by fatty acids and insulin.

Article CAS Google Scholar Kersten S, Mandard S, Escher P, Gonzalez FJ, Tafuri S, Desvergne B, Wahli W: The peroxisome proliferator-activated receptor alpha regulates amino acid metabolism.

Article CAS Google Scholar Maechler P, Wollheim CB: Mitochondrial glutamate acts as a messenger in glucose-induced insulin exocytosis.

Article CAS Google Scholar Unger RH, Orci L: Lipoapoptosis: its mechanism and its diseases. Article CAS Google Scholar Prentki M, Joly E, El-Assaad W, Roduit R: Malonyl-CoA signaling, lipid partitioning, and glucolipotoxicity.

Article CAS Google Scholar Tordjman K, Standley KN, Bernal-Mizrachi C, Leone TC, Coleman T, Kelly DP, Semenkovich CF: PPARα suppresses insulin secretion and induces UCP2 in insulinoma cells.

CAS Google Scholar Kim H, Haluzik M, Asghar Z, Yau D, Joseph JW, Fernandez AM, Reitman ML, Yakar S, Stannard B, Heron-Milhavet L, Wheeler MB, LeRoith D: Peroxisome proliferator-activated receptor-α agonist treatment in a transgenic model of type 2 diabetes reverses the lipotoxic state and improves glucose homeostasis.

Article CAS Google Scholar Koh EH, Kim M-S, Park J-Y, Kim HS, Youn J-Y, Park H-S, Youn JH, Lee K-U: Peroxisome proliferator-activated receptor PPAR -α activation prevents diabetes in OLETF rats.

Article CAS Google Scholar Kelly LJ, Vicario PP, Thompson GM, Candelore MR, Doebber TW, Ventre J, Wu MS, Meurer R, Forrest MJ, Conner MW, Cascieri MA, Molle DE: Peroxisome proliferator-acitvated receptors γ and α mediate in vivo regulation of uncoupling protein UCP-1, UCP-2, UCP-3 gene expression.

CAS Google Scholar Schrauwen P, Hesselink M: UCP2 and UCP3 in muscle controlling body metabolism. CAS Google Scholar Grav HJ, Tronstad KJ, Gudbrandsen OA, Berge K, Fladmark KE, Martinsen TC, Waldum H, Wergedahl H, Berge RK: Changed energy state and increased mitochondrial beta-oxidation rate in liver of rats associated with lowered proton electrochemical potential and stimulated uncoupling protein 2 UCP-2 expression: evidence for peroxisome proliferator-activated receptor-alpha independent induction of UCP-2 expression.

Article CAS Google Scholar Chan CB, De Leo D, Joseph JW, McQuaid TS, Ha XF, Xu F, Tsushima RG, Pennefather PS, Salapatek AM, Wheeler MB: Increased uncoupling protein-2 levels in beta cells are associated with impaired glucose-stimulated insulin secretion: mechanism of action.

Article CAS Google Scholar Hong Y, Fink BD, Dillon JS, Sivitz W: Effects of adenoviral overexpression of uncoupling protein-2 and -3 on mitochondrial respiration in insulinoma cells. CAS Google Scholar Zhang CY, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, Hagen T, Vidal-Puig AJ, Boss O, Kim YB, Zheng XX, Wheeler MB, Shulman GI, Chan CB, Lowell BB: Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, beta cell dysfunction and type 2 diabetes.

Article CAS Google Scholar Joseph JW, Koshkin V, Zhang CY, Wang J, Lowell BB, Chan CB, Wheeler MB: Uncoupling protein 2 knockout mice have enhanced insulin secretory capacity after a high fat diet. Article CAS Google Scholar Patane G, Anello M, Piro S, Vigneri R, Purrello F, Rabuazzo AM: Role of ATP production and uncoupling protein-2 in the insulin secretory defect induced by chronic exposure to high glucose or free fatty acids and effects of peroxisome proliferator-activated receptor-γ inhibition.

Article CAS Google Scholar Shimabukuro M, Zhou Y-T, Lee Y, Unger RH: Induction of uncoupling protein-2 mRNA by troglitazone in the pancreatic islets of Zucker diabetic fatty rats. Article CAS Google Scholar Sreenan S, Sturis J, Pugh W, Burant CF, Polonsky KS: Prevention of hyperglycemia in the Zucker diabetic fatty rat by treatment with metformin or troglitazone.

CAS Google Scholar Parton LE, Diraison F, Neill SE, Ghosh SK, Rubino MA, Bisi JE, Briscoe CP, Rutter GA: Impact of PPARγ overexpression and activation on pancreatic islet gene expression profile analyzed with oligonucleotide microarrays. Article CAS Google Scholar Eto K, Yamashita T, Matsui J, Terauchi Y, Noda M, Kadowaki T: Genetic manipulation of fatty acid metabolism in β-cells are associated with dysregulated insulin secretion.

Article CAS Google Scholar Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T, Eto K, Tsubamoto Y, Okuno A, Murakami K, Sekihara H, Hasegawa G, Naito M, Toyoshima Y, Tanaka S, Shiota K, Kitamura T, Fujita T, Ezaki O, Aizawa S, Nagai R, Tobe K, Kimura S, Kadowaki T: PPAR-γ mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance.

Article CAS Google Scholar Kim HI, Cha JY, Kim SY, Kim JW, Roh KJ, Seong JK, Lee NT, Choi KY, Kim KS, Ahn YH: Peroxisomal proliferator-activated receptor-gamma upregulates glucokinase gene expression in beta-cells.

Article CAS Google Scholar Kim HI, Kim JW, Kim SH, Cha JY, Kim AS, Ahn YH: Identification and functional characterization of the peroxisomal proliferator response element in rat GLUT2 promoter.

Article CAS Google Scholar Wang Z, Gleichmann H: Glut2 in pancreatic islets: crucial target molecule in diabetes induced with multiple low doses of streptozotocin in mice. Article CAS Google Scholar Nakamichi Y, Kikuta T, Ito E, Ohara-Imaizumi M, Nishiwaki C, Ishida H, Nagamatsu S: PPAR-γ overexpression suppresses glucose-induced proinsulin biosynthesis and insulin release synergistically with pioglitazone in MIN6 cells.

Article CAS Google Scholar Lupi R, Del Guerra S, Marselli L, Bugliani M, Boggi U, Mosca F, Marchetti P, Del Prato S: Rosiglitazone prevents the impairment of human islet function induced by fatty acids: evidence for a role of PPARγ2 in the modulation of insulin secretion.

Article CAS Google Scholar Diani AR, Sawada G, Wyse B, Marray FT, Khan M: Pioglitazone preserves pancreatic islet structure and insulin secretory funciton in three murine models of type 2 diabetes. Article Google Scholar Higa M, Zhou Y-T, Ravazzola M, Baetens D, Orci L, Unger RH: Troglitazone prevents mitochondrial alterations, β cell destruction and diabetes in obese prediabetic rats.

Article CAS Google Scholar Beales PE, Liddi R, Giorgini AE, Signore A, Procaccini E, Batchelor K, Pozzilli P: Troglitazone prevents insulin dependent diabetes in the non-obese diabetic mouse. Article CAS Google Scholar Pershadsingh HA: Peroxisome proliferator-activated receptor-γ: therapeutic target for diseases beyond diabetes: quo vadis?.

Article CAS Google Scholar Rosen ED, Kulkarni RN, Sarraf P, Ozcan U, Okada T, Hsu C-H, Eisenman D, Magnuson MA, Gonzalez FJ, Kahn CR, Spiegelman BM: Targeted elimination of peroxisome proliferator-activated receptor γ in β cells leads to abnormalities in islet mass without compromising glucose homeostasis.

Article CAS Google Scholar Foufelle F, Ferre P: New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c. Article CAS Google Scholar Horton JD, Goldstein JL, Brown MS: SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver.

Article CAS Google Scholar Kakuma T, Lee Y, Higa M, Wang Z, Pan W, Shimomura I, Unger RH: Leptin, troglitazone, and the expression of sterol regulatory element binding proteins in liver and pancreatic islets.

Article CAS Google Scholar Andreolas C, da Silva Xavier G, Diraison F, Zhao C, Varadi A, Lopez-Casillas F, Ferre P, Foufelle F, Rutter GA: Stimulation of acetyl-CoA carboxylase gene expression by glucose requires insulin release and sterol regulatory element binding protein 1c in pancreatic MIN6 beta-cells.

Article CAS Google Scholar Wang H, Maechler P, Antinozzi PA, Herrero L, Hagenfeldt-Johansson KA, Bjorklund A, Wollheim CB: The transcription factor SREBP-1c is instrumental in the development of beta-cell dysfunction.

Article CAS Google Scholar Diraison F, Parton L, Ferre P, Foufelle F, Briscoe CP, Leclerc I, Rutter GA: Over-expression of sterol-regulatory-element-binding protein-1c SREBP1c in rat pancreatic islets induces lipogenesis and decreases glucose-stimulated insulin release: modulation by 5-aminoimidazolecarboxamide ribonucleoside AICAR.

Article CAS Google Scholar Medvedev AV, Robidoux J, Bai X, Cao W, Floering LM, Daniel KW, Collins S: Regulation of the uncoupling protein-2 gene in INS-1 β-cells by oleic acid.

Article CAS Google Scholar Yamashita T, Eto K, Okazaki Y, Yamashita S, Yamauchi T, Sekine N, Nagai R, Noda M, Kadowaki T: Role of uncoupling protein-2 up-regulation and triglyceride accumulation in impaired glucose-stimulated insulin secretion in a β-cell lipotoxicity model overexpressing sterol regulatory element binding protein-1c.

Article CAS Google Scholar Poitout V: β-cell lipotoxicity: burning fat into heat?. Article CAS Google Scholar Elholm M, Bjerking G, Knudsen J, Kristiansen K, Mandrup S: Regulatory elements in the promoter region of the rat gene encoding the acyl-CoA-binding protein.

Article CAS Google Scholar Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, Wahli W: The PPARα-leukotriene B4 pathway to inflammation control.

Article CAS Google Scholar Varanasi U, Chu R, Huang Q, Castellon R, Yeldandi AV, Reddy JK: Identification of a peroxisome proliferator-responsive element upstream of the human peroxisomal fatty acyl coenzyme A oxidase gene.

Article CAS Google Scholar Karam WG, Ghanayem BI: Induction of replication DNA synthesis and PPARα-dependent gene transcription by Wy in primary rat hepatocyte and non-parenchymal cell co-cultures. Article CAS Google Scholar Berthou L, Duverger N, Emmanuel F, Langouet S, Auwerx J, Guillouzo A, Fruchart JC, Rubin E, Denefle P, Staels B, Branellec D: Opposite regulation of human versus mouse apolipoprotein A-I by fibrates in human apolipoprotein A-I transgenic mice.

Article CAS Google Scholar Raspe E, Madsen L, Lefebvre AM, Leitersdorf I, Gelman L, Peinado-Onsurbe J, Dallongeville J, Fruchart JC, Berge R, Staels B: Modulation of rat liver apolipoprotein gene expression and serum lipid levels by tetradecylthioacetic acid TTA via PPARα activation.

CAS Google Scholar Vu-Dac N, Schoonjans K, Kosykh V, Dallongeville J, Fruchart JC, Staels B, Auwerx J: Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor. Article CAS Google Scholar Prieur X, Coste H, Rodriguez JC: The human apolipoprotein AV gene is regulated by PPAR α and contains a novel FXR response element.

Article CAS Google Scholar Hertz R, Bishara-Shieban J, Bar-Tana J: Mode of action of peroxisome proliferators as hypolipidemic drugs. Article CAS Google Scholar Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne B, Wahli W: Peroxisome proliferator-activated receptor α mediates the adaptive response to fasting.

Article CAS Google Scholar Leone TC, Weinheimer CJ, Kelly DP: A critical role for the peroxisome proliferator-activated receptor α in the cellular fasting response: the PPARα-null mouse as a model of fatty acid oxidation disorders. Article CAS Google Scholar Aoyama T, Peters JM, Iritani N, Nakajuima T, Furihata K, Hashimoto T, Gonzalez FJ: Altered constitutive expression of fatty acid-metabolizing enzymes in mice lacking the peroxisome proliferator-activated receptor α.

Article CAS Google Scholar Minnich A, Tian N, Byan L, Bilder G: A potent PPARα agonist stimulates mitochondrial fatty acid β-oxidation in liver and skeletal muscle.

CAS Google Scholar Kroetz DL, Yook P, Costet P, Bianchi P, Pineau T: Peroxisome proliferator-activated receptor α controls the hepatic CYP4A induction adaptive response to starvation and diabetes. Article CAS Google Scholar Aldridge TC, Tugwood JD, Green S: Identification and characterization of DNA elements implicated in the regulation of CYP4A1 transcription.

Article CAS Google Scholar Yu S, Cao WQ, Kashireddy P, Meyer K, Jia Y, Hughes DE, Tan Y, Feng J, Yeldandi AV, Rao MS, Costa RH, Gonzalez FJ, Reddy JK: Human peroxisome proliferator-activated receptor α supports the induction of peroxisome proliferation in PPARα-deficient mouse liver.

Article CAS Google Scholar Patel DD, Knight BL, Soutar AK, Gibbons GF, Wade DP: The effect of peroxisome-proliferator-activated receptor-α on the activity of the cholesterol 7 α-hydroxylase gene.

Article CAS Google Scholar Cheema SK, Agellon LB: The murine and human cholesterol 7 α-hydroxylase gene promoters are differentially responsive to regulation by fatty acids mediated via peroxisome proliferator-activated receptor α.

Article CAS Google Scholar Guillou H, Martin P, Jan S, D'Andrea S, Roulet A, Catheline D, Rioux V, Pineaut T, Legrand P: Comparative effect of fenofibrate on hepatic desaturases in wild-type and peroxisome proliferator-activated receptor α-deficient mice.

Article CAS Google Scholar Wolfrum C, Ellinghaus P, Fobker M, Seedorf U, Assmann G, Borchers T, Spener F: Phytanic acid is ligand and transcriptional activator of muring liver fatty acid binding protein. CAS Google Scholar Motojima K, Passilly P, Peters JM, Gonzalez FJ, Latruffe N: Expression of putative fatty acid transporter genes are regulated by peroxisome proliferator-activated receptor α and γ activators in a tissue- and inducer-specific manner.

Article CAS Google Scholar Yamazaki K, Kuromitsu J, Tanaka I: Microarray analysis of gene expression changes in mouse liver induced by peroxisome proliferator-activated receptor α agonists. Article CAS Google Scholar Kok T, Wolters H, Bloks W, Havinga R, Jansen PL, Staels B, Kuipers F: Induction of hepatic ABC transporter expression is part of the PPARα-mediated fasting response in the mouse.

Article CAS Google Scholar Tobin KA, Steineger HH, Alberti S, Spydevold O, Auwerx J, Gustafsson JA, Nebb HI: Cross talk between fatty acid and cholesterol metabolism mediated by liver X receptor-α. Huang, S.

The GLUT4 glucose transporter. Cell Metab 5, — Huang, X. Region-specific requirement for cholesterol modification of sonic hedgehog in patterning the telencephalon and spinal cord.

Development , — Hubbard, B. Mice deleted for fatty acid transport protein 5 have defective bile acid conjugation and are protected from obesity. Hyde, C. Identification of 15 genetic loci associated with risk of major depression in individuals of European descent.

Nat Genet 48, — Iacobazzi, V. Citrate—new functions for an old metabolite. Biol Chem , — Infantino, V. A key role of the mitochondrial citrate carrier SLC25A1 in TNFα- and IFNγ-triggered inflammation. Biochim Biophys Acta Gene Regulat Mech , — Ishigami, M. Temporary sequestration of cholesterol and phosphatidylcholine within extracellular domains of ABCA1 during nascent HDL generation.

Sci Rep 8, Izar, M. Phytosterols and phytosterolemia: gene-diet interactions. Genes Nutr 6, 17— Jansson, D. Glucose controls CREB activity in islet cells via regulated phosphorylation of TORC2.

Jerome, W. Lysosomes, cholesterol and atherosclerosis. Clin Lipidol 5, — Jiang, S. Discovery of a potent HMG-CoA reductase degrader that eliminates statin-induced reductase accumulation and lowers cholesterol.

Nat Commun 9, Schnyder corneal dystrophy-associated UBIAD1 mutations cause corneal cholesterol accumulation by stabilizing HMG-CoA reductase. PLoS Genet 15, e Jo, Y. Schnyder corneal dystrophy-associated UBIAD1 inhibits ER-associated degradation of HMG CoA reductase in mice.

eLife 8, e Johnson, M. Developmentally regulated H2Av buffering via dynamic sequestration to lipid droplets in Drosophila embryos.

Jones, B. Mutations in a Sar1 GTPase of COPII vesicles are associated with lipid absorption disorders. Nat Genet 34, 29— Joshi, A. Lipid droplet and peroxisome biogenesis occur at the same ER subdomains. Kadereit, B.

Evolutionarily conserved gene family important for fat storage. Proc Natl Acad Sci USA , 94— Kannel, W. Factors of risk in the development of coronary heart disease—six-year follow-up experience. Ann Intern Med 55, Kathiresan, S. A genome-wide association study for blood lipid phenotypes in the Framingham Heart Study.

BMC Med Genet 8, S Six new loci associated with blood low-density lipoprotein cholesterol, high-density lipoprotein cholesterol or triglycerides in humans. Nat Genet 40, — Kaushik, S. Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis.

Nat Cell Biol 17, — Kazda, C. Evaluation of efficacy and safety of the glucagon receptor antagonist LY in patients with type 2 diabetes: and week Phase 2 studies. Dia Care 39, — Kazierad, D. Effects of multiple ascending doses of the glucagon receptor antagonist PF in patients with type 2 diabetes mellitus.

Diabetes Obes Metab 18, — Kc, S. Vitamin C enters mitochondria via facilitative glucose transporter 1 Glut1 and confers mitochondrial protection against oxidative injury. FASEB J 19, — Khachadurian, A. The inheritance of essential familial hypercholesterolemia. Am J Med 37, — Experiences with the homozygous cases of familial hypercholesterolemia.

Ann Nutr Metab 15, — Khoriaty, R. Functions of the COPII gene paralogs SEC23A and SEC23B are interchangeable in vivo. Proc Natl Acad Sci USA , E—E Article PubMed CAS PubMed Central Google Scholar. Kimmel, A. The Perilipins: major cytosolic lipid droplet-associated proteins and their roles in cellular lipid storage, mobilization, and systemic homeostasis.

Annu Rev Nutr 36, — Klarin, D. Koepsell, H. The SLC22 family with transporters of organic cations, anions and zwitterions. Kolovou, G. Tangier disease four decades of research: a reflection of the importance of HDL. Curr Med Chem 13, — Koo, S. The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism.

Kooner, J. Genome-wide scan identifies variation in MLXIPL associated with plasma triglycerides. Kory, N. Targeting fat: mechanisms of protein localization to lipid droplets.

Trends Cell Biol 26, — Protein crowding is a determinant of lipid droplet protein composition. Dev Cell 34, — Kotani, K. GLUT4 glucose transporter deficiency increases hepatic lipid production and peripheral lipid utilization.

Krahmer, N. Phosphatidylcholine synthesis for lipid droplet expansion is mediated by localized activation of CTP:phosphocholine cytidylyltransferase.

Cell Metab 14, — Kumar, N. Identification of SR ML : a synthetic RORα selective inverse agonist. ACS Chem Biol 6, — Kurland, I. Protein Sci 4, — Kwok, R. Nuclear protein CBP is a coactivator for the transcription factor CREB. Kwon, H. Structure of N-terminal domain of NPC1 reveals distinct subdomains for binding and transfer of cholesterol.

The structure of the NPC1L1 N-Terminal domain in a closed conformation. PLoS ONE 6, e Lagace, T. PCSK9 and LDLR degradation. Curr Opin Lipidol 25, — Lang, M. Secretory COPII coat component Sec23a is essential for craniofacial chondrocyte maturation.

Lange, L. Whole-exome sequencing identifies rare and low-frequency coding variants associated with LDL cholesterol. Am J Hum Genet 94, — Lange, Y. Quantitation of the pool of cholesterol associated with acyl-CoA:cholesterol acyltransferase in human fibroblasts.

Regulation of endoplasmic reticulum cholesterol by plasma membrane cholesterol. Lass, A. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI and defective in Chanarin-Dorfman syndrome. Cell Metab 3, — Lee, J. Lee, M. Bi-directional protein transport between the ER and Golgi.

Annu Rev Cell Dev Biol 20, 87— Lee, Y. Cyclin D1-Cdk4 controls glucose metabolism independently of cell cycle progression. Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent diabetes mellitus of obese rats: impairment in adipocyte-beta-cell relationships..

Proc Natl Acad Sci USA 91, — Lengacher, S. Resistance to diet-induced obesity and associated metabolic perturbations in haploinsufficient monocarboxylate transporter 1 mice. PLoS ONE 8, e Lewis, P. Cholesterol modification of sonic hedgehog is required for long-range signaling activity and effective modulation of signaling by Ptc1.

Li, E. OLFR mediates glucose metabolism as a receptor of asprosin. Cell Metab 30, — Li, J. Lysosomal membrane glycoproteins bind cholesterol and contribute to lysosomal cholesterol export.

eLife 5, e Li, P. Adipocyte NCoR knockout decreases PPARγ phosphorylation and enhances PPARγ activity and insulin sensitivity. The clathrin adaptor Numb regulates intestinal cholesterol absorption through dynamic interaction with NPC1L1.

Nat Med 20, 80— Li, T. Tipmediated lipin 1 acetylation and ER translocation determine triacylglycerol synthesis rate. Li, X. Li, Z.

Drosophila lipid droplets buffer the H2Av supply to protect early embryonic development. Curr Biol 24, — Lipid droplets control the maternal histone supply of Drosophila embryos. Curr Biol 22, — Liao, Y.

The non-canonical NF-κB pathway promotes NPC2 expression and regulates intracellular cholesterol trafficking. Sci China Life Sci 61, — Lin, H. Hormonal regulation of hepatic glucose production in health and disease.

Cell Metab 14, 9— Linton, M. SR-BI: a multifunctional receptor in cholesterol homeostasis and atherosclerosis. Trends Endocrinol Metab 28, — Lira, V. Nitric oxide and AMPK cooperatively regulate PGC-1α in skeletal muscle cells.

J Physiol , — Liscum, L. Intracellular cholesterol transport. Biochim Biophys Acta Mol Cell Biol Lipids , 19— Listenberger, L. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Liu, B. Liu, D. Nat Genet 49, — Liu, L.

Coupling of COPII vesicle trafficking to nutrient availability by the IRE1α-XBP1s axis. Proc Natl Acad Sci USA 23, Cell Metab 26, — Glial lipid droplets and ROS induced by mitochondrial defects promote neurodegeneration.

Liu, T. Clin Sci , — Liu, Y. Lizcano, J. The insulin signalling pathway. Curr Biol 12, R—R Lu, K. Two genes that map to the STSL locus cause sitosterolemia: genomic structure and spectrum of mutations involving sterolin-1 and sterolin-2, encoded by ABCG5 and ABCG8, respectively.

Am J Hum Genets 69, — Article Google Scholar. Lu, M. Insulin regulates liver metabolism in vivo in the absence of hepatic Akt and Foxo1. Nat Med 18, — Lu, X. Genetic susceptibility to lipid levels and lipid change over time and risk of incident hyperlipidemia in Chinese populations.

Circ Cardiovasc Genet 9, 37— Coding-sequence variants are associated with blood lipid levels in 14, Chinese. Hum Mol Genet 25, — Exome chip meta-analysis identifies novel loci and East Asian-specific coding variants that contribute to lipid levels and coronary artery disease. Luo, H. AIDA selectively mediates downregulation of fat synthesis enzymes by ERAD to retard intestinal fat absorption and prevent obesity.

Madiraju, A. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Magré, J. Identification of the gene altered in Berardinelli-Seip congenital lipodystrophy on chromosome 11q Nat Genet 28, — Malhotra, V. Procollagen export from the endoplasmic reticulum.

Biochm Soc Trans 43, — Mancias, J. The transport signal on Sec22 for packaging into COPII-coated vesicles is a conformational epitope. Mol Cell 26, — Structural basis of cargo membrane protein discrimination by the human COPII coat machinery.

EMBO J 27, — Martinez-Botas, J. Nat Genet 26, — Matsumoto, M. Impaired regulation of hepatic glucose production in mice lacking the forkhead transcription factor Foxo1 in liver.

Cell Metab 6, — Maurano, M. Systematic localization of common disease-associated variation in regulatory DNA. Melnikov, A. Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay.

Nat Biotechnol 30, — Meyer, C. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Mihaylova, M. Class IIa histone deacetylases are hormone-activated regulators of FOXO and mammalian glucose homeostasis.

Milger, K. Cellular uptake of fatty acids driven by the ER-localized acyl-CoA synthetase FATP4. J Cell Sci , — Miller, E. Multiple cargo binding sites on the COPII subunit Sec24p ensure capture of diverse membrane proteins into transport vesicles.

Mithieux, G. Intestinal gluconeogenesis: key signal of central control of energy and glucose homeostasis. Curr Opin Clin Nutrit Metab Care 12, — Mitsche, M.

Flux analysis of cholesterol biosynthesis in vivo reveals multiple tissue and cell-type specific pathways. eLife 4, e Miyanari, Y. The lipid droplet is an important organelle for hepatitis C virus production.

Nat Cell Biol 9, — Moon, S. p53 represses the mevalonate pathway to mediate tumor suppression. Moore, M. Regulation of hepatic glucose uptake and storage in vivo. Adv Nutrit 3, — Mossessova, E. SNARE selectivity of the COPII coat. Mueckler, M. The SLC2 GLUT family of membrane transporters.

Mugoni, V. Ubiad1 is an antioxidant enzyme that regulates eNOS activity by CoQ10 synthesis. Muller, C. Xanthomata, hypercholesterolemia, angina pectoris. Acta Med Scand 89, 75— Musunuru, K.

Genetics of common, complex coronary artery disease. Myers, S. Effects of small changes in glucagon on glucose production during a euglycemic, hyperinsulinemic clamp. Metabolism 40, 66— Nagao, K.

ATP hydrolysis-dependent conformational changes in the extracellular domain of ABCA1 are associated with apoA-I binding. J Lipid Res 53, — Nagashima, T. Mol Pharmacol 78, — Nakagawa, K. Identification of UBIAD1 as a novel human menaquinone-4 biosynthetic enzyme. Nguyen, K. Alternatively activated macrophages produce catecholamines to sustain adaptive thermogenesis.

Nguyen, T. DGAT1-dependent lipid droplet biogenesis protects mitochondrial function during starvation-induced autophagy. Dev Cell 42, 9— Nisoli, E. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide.

Nohturfft, A. Regulated step in cholesterol feedback localized to budding of SCAP from ER membranes. Nordestgaard, B. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society.

Eur Heart J 34, — Nowaczyk, M. Smith-Lemli-Opitz syndrome: phenotype, natural history, and epidemiology. Am J Med Genet C, — Ohashi, K.

Novel mutations in the microsomal triglyceride transfer protein gene causing abetalipoproteinemia. J Lipid Res 41, — Okar, D. Fructose-2,6-bisphosphate and control of carbohydrate metabolism in eukaryotes.

Biofactors 10, 1— Olzmann, J. Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol 20, — Orr, A. Mutations in the UBIAD1 gene, encoding a potential prenyltransferase, are causal for Schnyder crystalline corneal dystrophy. PLoS ONE 2, e Osuga, J. Targeted disruption of hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity.

Proc Natl Acad Sci USA 97, — Ou, H. Role of AMPK in atherosclerosis via autophagy regulation. Ozaki, K. Functional SNPs in the lymphotoxin-α gene that are associated with susceptibility to myocardial infarction.

Nat Genet 32, — Ozeki, S. Rab18 localizes to lipid droplets and induces their close apposition to the endoplasmic reticulum-derived membrane. Palmieri, F. The mitochondrial transporter family SLC identification, properties and physiopathology.

Peng, R. Evidence for overlapping and distinct functions in protein transport of coat protein Sec24p family members.

Perez-Poyato, M. New agents and approaches to treatment in Niemann-Pick type C disease. Curr Pharm Biotechnol 12, — Perland, E. Classification systems of secondary active transporters. Trends Pharmacol Sci 38, — Perry, R. Leptin mediates a glucose-fatty acid cycle to maintain glucose homeostasis in starvation.

Petersen, M. Regulation of hepatic glucose metabolism in health and disease. Nat Rev Endocrinol 13, — Pfisterer, S. LDL-cholesterol transport to the endoplasmic reticulum.

Curr Opin Lipidol 27, — Phillips, M. Molecular mechanisms of cellular cholesterol efflux. Pilkis, S. Annu Rev Nutr 11, — Regulation by glucagon of hepatic pyruvate kinase, 6-phosphofructo 1-kinase, and fructose-1,6-bisphosphatase. Fed Proc 41, — Porter, F.

Smith-Lemli-Opitz syndrome: pathogenesis, diagnosis and management. Eur J Hum Genet 16, — Malformation syndromes caused by disorders of cholesterol synthesis. J Lipid Res 52, 6— Porter, J. Cholesterol modification of hedgehog signaling proteins in animal development.

Puigserver, P. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1α interaction. Puntoni, M. Tangier disease.

Am J Cardiovasc Drugs 12, — Qian, H. Structure of the human lipid exporter ABCA1. Qiang, G. Identification of a small molecular insulin receptor agonist with potent antidiabetes activity. Diabetes 63, — Radhakrishnan, A. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: a delicate balance.

Cell Metab 8, — Direct binding of cholesterol to the purified membrane region of SCAP. Mol Cell 15, — Radziuk, J. Hepatic glucose uptake, gluconeogenesis and the regulation of glycogen synthesis.

Diabetes Metab Res Rev 17, — Rambold, A. Fatty acid trafficking in starved cells: regulation by lipid droplet lipolysis, autophagy, and mitochondrial fusion dynamics. Dev Cell 32, — Rasouli, N. Ectopic fat accumulation and metabolic syndrome.

Diabetes Obes Metab 9, 1— Ravier, M. Glucose or insulin, but not zinc ions, inhibit glucagon secretion from mouse pancreaticα-cells. Diabetes 54, — Raychaudhuri, S. Mapping rare and common causal alleles for complex human diseases. Cell , 57— Rebsamen, M. SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1.

Reeskamp, L. Transintestinal cholesterol excretion in humans. Curr Opin Lipidol 29, 10— Rehberg, E. A novel abetalipoproteinemia genotype. Rines, A. Targeting hepatic glucose metabolism in the treatment of type 2 diabetes.

Nat Rev Drug Discov 15, — Rizza, R. Pathogenesis of fasting and postprandial hyperglycemia in type 2 diabetes: implications for therapy.

Roberg, K. LST1 is a SEC24 homologue used for selective export of the plasma membrane ATPase from the endoplasmic reticulum. Roingeard, P. Lipid droplet hijacking by intracellular pathogens. Cell Microbiol 19, e Romanauska, A. The inner nuclear membrane is a metabolically active territory that generates nuclear lipid droplets.

Romere, C. Asprosin, a fasting-induced glucogenic protein hormone. Russell, D. cDNA cloning of the bovine low density lipoprotein receptor: feedback regulation of a receptor mRNA..

Proc Natl Acad Sci USA 80, — Rust, S. Tangier disease is caused by mutations in the gene encoding ATP-binding cassette transporter 1. Rusu, V. Type 2 diabetes variants disrupt function of slc16a11 through two distinct mechanisms. Sabatine, M. PCSK9 inhibitors: clinical evidence and implementation.

Nat Rev Cardiol 16, — Saberi, M. Novel liver-specific TORC2 siRNA corrects hyperglycemia in rodent models of type 2 diabetes. Saini, V. Molecular mechanisms of insulin resistance in type 2 diabetes mellitus. World J Diabetes 1, 68— Sakai, J. Sterol-regulated release of SREBP-2 from cell membranes requires two sequential cleavages, one within a transmembrane segment.

Cell 85, — Salo, V. Seipin facilitates triglyceride flow to lipid droplet and counteracts droplet ripening via endoplasmic reticulum contact. Dev Cell 50, — Sandhu, M. LDL-cholesterol concentrations: a genome-wide association study. Santos, A. Schekman, R. How sterols regulate protein sorting and traffic.

Schonfeld, G. Familial hypobetalipoproteinemia. J Lipid Res 44, — Familial hypobetalipoproteinemia: genetics and metabolism.

CMLS Cell Mol Life Sci 62, — Schumacher, M. The prenyltransferase UBIAD1 is the target of geranylgeraniol in degradation of HMG CoA reductase.

Geranylgeranyl-regulated transport of the prenyltransferase UBIAD1 between membranes of the ER and Golgi. J Lipid Res 57, — Schwarz, K. Mutations affecting the secretory COPII coat component SEC23B cause congenital dyserythropoietic anemia type II.

Nat Genet 41, — Screaton, R. The CREB coactivator TORC2 functions as a calcium- and cAMP-sensitive coincidence detector. Cell , 61— Seidah, N. The secretory proprotein convertase neural apoptosis-regulated convertase 1 NARC-1 : liver regeneration and neuronal differentiation.

Sezgin, E. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol 18, — Sharabi, K. Molecular pathophysiology of hepatic glucose production.

Mol Aspect Med 46, 21— Shaw, R. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Shepherd, P. Glucose transporters and insulin action—implications for insulin resistance and diabetes mellitus.

Shimano, H. Overproduction of cholesterol and fatty acids causes massive liver enlargement in transgenic mice expressing truncated SREBP-1a.. J Clin Invest 98, — Shimoni, Y. Lst1p and Sec24p cooperate in sorting of the plasma membrane ATPase into COPII vesicles in Saccharomyces cerevisiae.

Shubeita, G. Consequences of motor copy number on the intracellular transport of kinesindriven lipid droplets. Singh, R. Autophagy regulates lipid metabolism. Solt, L. Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists.

Nature , 62— Song, W. Organic cation transporter 3 Oct3 is a distinct catecholamines clearance route in adipocytes mediating the beiging of white adipose tissue. PLoS Biol 17, e Soria, L. Association between a specific apolipoprotein B mutation and familial defective apolipoprotein B Proc Natl Acad Sci USA 86, — Soutar, A.

Mechanisms of disease: genetic causes of familial hypercholesterolemia. Nat Rev Cardiol 4, — Genetics, clinical phenotype, and molecular cell biology of autosomal recessive hypercholesterolemia.

Arterioscler Thromb Vasc Biol 23, — Striffler, J. Effects of glucagon on hepatic microsomal glucosephosphatase in vivo. Diabete Metab 10, 91— Strøm, T. Characterization of residues in the cytoplasmic domain of the LDL receptor required for exit from the endoplasmic reticulum.

Biochem Biophys Res Commun , — Su, L. Sui, X. Cryo-electron microscopy structure of the lipid droplet-formation protein seipin. Sun, L. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Insig renders sorting signal in Scap inaccessible to COPII proteins.

Sun, Z. Perilipin1 promotes unilocular lipid droplet formation through the activation of Fsp27 in adipocytes. Nat Commun 4, Surakka, I. The impact of low-frequency and rare variants on lipid levels.

Nat Genet 47, — Svoboda, M. Treatment of Smith-Lemli-Opitz syndrome and other sterol disorders. Sztalryd, C. Perilipin A is essential for the translocation of hormone-sensitive lipase during lipolytic activation.

Szymanski, K. The lipodystrophy protein seipin is found at endoplasmic reticulum lipid droplet junctions and is important for droplet morphology. Tabas, I. Consequences of cellular cholesterol accumulation: basic concepts and physiological implications.

Tan, K. Effect of glucagon antibodies on plasma glucose, insulin and somatostatin in the fasting and fed rat. Diabetologia 28, — Tanaka, H. Eur J Pharmacol , — Tao, J.

SEC23B is required for the maintenance of murine professional secretory tissues. Temel, R. Hepatic Niemann-Pick C1-like 1 regulates biliary cholesterol concentration and is a target of ezetimibe.

Teslovich, T. Biological, clinical and population relevance of 95 loci for blood lipids. Tian, Y. Tissue-autonomous function of Drosophila seipin in preventing ectopic lipid droplet formation.

Fat metabolism regulation means it's official. Federal government websites mtabolism end in. gov or. Before sharing sensitive information, make sure you're on a federal government site. The site is secure. Glucose and fatty acids are the Preventing burnout in young athletes sources of regularion for human body. Fat metabolism regulation, the most abundant sterol in mammals, is a key component of cell membranes metagolism it does not generate ATP. Fat metabolism regulation Fah of glucose, fatty acids and cholesterol are often intertwined and regulated. For example, glucose can be converted to fatty acids and cholesterol through de novo lipid biosynthesis pathways. Excessive lipids are secreted in lipoproteins or stored in lipid droplets. The metabolites of glucose and lipids are dynamically transported intercellularly and intracellularly, and then converted to other molecules in specific compartments. The disorders of glucose and lipid metabolism result in severe diseases including cardiovascular disease, diabetes and fatty liver.

Video

Lipid (Fat) Metabolism Overview, Animation

Author: Maukinos

1 thoughts on “Fat metabolism regulation

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com