Category: Diet

Lipid metabolism and glucose utilization

Lipid metabolism and glucose utilization

CAS PubMed Google Scholar Arner, Mrtabolism. Myokines in insulin resistance and type 2 diabetes. Plasma triglycerides followed a time-response Metabolisk similar to glycemia profile in L line at 12 h post-glucose treatment. The regulation of glycogen synthase by protein phosphatase 1 in 3T3-L1 adipocytes. We apologize to the many investigators whose work we could not cite owing to a limit on the number of references.

Metzbolism you for visiting utiliztion. You are using gluclse browser version with limited support for CSS. To obtain the best experience, Lioid recommend you utiliztaion a more up to Foods that increase thermogenesis browser or Lipid metabolism and glucose utilization off compatibility mode in Internet Explorer.

Metabolisn the metaboliism, to ensure continued support, we are displaying the site without styles and JavaScript. The epidemic of type 2 diabetes and impaired glucose tolerance is Lipid metabolism and glucose utilization of the main causes of morbidity and mortality worldwide.

In both disorders, tissues such metaboliism muscle, glucosee and Gut health and nutrient absorption become less Glycose or metabbolism to insulin.

This state is also linked to other Lipid metabolism and glucose utilization health problems, such as obesity, polycystic ovarian disease, hyperlipidaemia, hypertension and atherosclerosis. Lioid pathophysiology of insulin resistance involves a complex network of signalling pathways, activated by the insulin receptor, which regulates Lipid metabolism and glucose utilization metabolism and its utiliation in cells.

But recent studies have shown that numerous utillization hormones and Lipid metabolism and glucose utilization Chromium browser shortcuts attenuate insulin action, mrtabolism are Happiness in type tuilization diabetes.

This is Lipid metabolism and glucose utilization preview of subscription content, access via your institution. Klip, A. Glucose transport and glucose transporters in muscle and their metabolic regulation.

Lipid metabolism and glucose utilization Care 13— High sugar carbohydrate foods CAS PubMed Google Scholar. Bruning, J.

et al. Metaholism muscle-specific insulin receptor knockout exhibits Lipid metabolism and glucose utilization of the metabolic syndrome metabolisj NIDDM without mftabolism glucose tolerance. Cell 2— Metabolim, E. Utilizaton targeting of the GLUT4 gene impairs insulin action in muscle and liver.

Nature— ADS CAS Utilizatiion Google Scholar. Gavrilova, O. Surgical implantation of adipose tissue reverses diabetes in metabolisn mice. CAS PubMed PubMed Central Google Scholar. Kulkarni, R. Tissue-specific knockout of the insulin receptor in pancreatic beta cells creates an insulin secretory defect similar to that in type 2 diabetes.

Cell 96— Patti, M. The insulin receptor—a critical link in glucose homeostasis and insulin action. Basic Clin. Butler, A. Tissue-specific versus generalized gene targeting of the igf1 and igf1r genes and their roles in insulin-like growth factor physiology.

Endocrinology— Skorokhod, A. Origin of insulin receptor-like tyrosine kinases in marine sponges. White, M. The IRS-signalling system: a network of docking proteins that mediate insulin action.

Pessin, J. Signaling pathways in insulin action: molecular targets of insulin resistance. Tamemoto, H. Insulin resistance and growth retardation in mice lacking insulin receptor substrate Araki, E. Alternative pathway of insulin signaling in mice with targeted disruption of the IRS-1 gene.

Kido, Y. Tissue-specific insulin resistance in mice with mutations in the insulin receptor, IRS-1, and IRS Withers, D. Disruption of IRS-2 causes type 2 diabetes in mice.

Fantin, V. Mice lacking insulin receptor substrate 4 exhibit mild defects in growth, reproduction, and glucose homeostasis. Fasshauer, M. Essential role of insulin receptor substrate 1 in differentiation of brown adipocytes.

Mendez, R. Jr, White, M. Stimulation of protein synthesis, eukaryotic translation initiation factor 4E phosphorylation, and PHAS-I phosphorylation by insulin requires insulin receptor substrate 1 and phosphatidylinositol 3-kinase.

Tsuruzoe, K. Insulin receptor substrate 3 IRS-3 and IRS-4 impair IRS and IRSmediated signaling. Hotamisligil, G. IRSmediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science— Craparo, A. Yuan, M. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkβ.

Kim, J. Prevention of fat-induced insulin resistance by salicylate. Elchebly, M. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Shepherd, P. Insulin stimulation of glycogen synthesis and glycogen synthase activity is blocked by wortmannin and rapamycin in 3T3-L1 adipocytes: evidence for the involvement of phosphoinositide 3-kinase and p70 ribosomal protein-S6 kinase.

Myers, M. Jr et al. Natl Acad. USA 89— ADS CAS PubMed PubMed Central Google Scholar. Pons, S. The structure and function of p55PIK reveal a new regulatory subunit for phosphatidylinositol 3-kinase. Antonetti, D. Insulin receptor substrate 1 binds two novel splice variants of the regulatory subunit of phosphatidylinositol 3-kinase in muscle and brain.

Fruman, D. Structural organization and alternative splicing of the murine phosphoinositide 3-kinase p85 alpha gene. Genomics 37— Kerouz, N. Hypoglycaemia, liver necrosis and perinatal death in mice lacking all isoforms of phosphoinositide 3-kinase p85α.

Nature Genet. Terauchi, Y. Increased insulin sensitivity and hypoglycaemia in mice lacking the p85α subunit of phosphoinositide 3-kinase. Lietzke, S. Structural basis of 3-phosphoinositide recognition by pleckstrin homology domains.

Cell 6— Kessler, A. Peterson, R. Kinase phosphorylation: keeping it all in the family. Mackay, D. Rho GTPases. Ziegler, S. Molecular cloning and characterization of a novel receptor protein tyrosine kinase from human placenta. Oncogene 8— Alessi, D. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha.

: Lipid metabolism and glucose utilization

Top bar navigation

A novel hepatokine, HFREP1, plays a crucial role in the development of insulin resistance and type 2 diabetes. Wu HT, Lu FH, Ou HY, Su YC, Hung HC, Wu JS, et al. The role of hepassocin in the development of non-alcoholic fatty liver disease.

Auberger P, Falquerho L, Contreres JO, Pages G, Le Cam G, Rossi B, et al. Characterization of a natural inhibitor of the insulin receptor tyrosine kinase: cDNA cloning, purification, and anti-mitogenic activity.

Stefan N, Fritsche A, Weikert C, Boeing H, Joost HG, Haring HU, et al. Plasma fetuin-A levels and the risk of type 2 diabetes.

Mukhopadhyay S, Bhattacharya S. Plasma fetuin-A triggers inflammatory changes in macrophages and adipocytes by acting as an adaptor protein between NEFA and TLR Pal D, Dasgupta S, Kundu R, Maitra S, Das G, Mukhopadhyay S, et al.

Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med. Haukeland JW, Dahl TB, Yndestad A, Gladhaug IP, Loberg EM, Haaland T, et al. Fetuin A in nonalcoholic fatty liver disease: in vivo and in vitro studies.

Mathews ST, Singh GP, Ranalletta M, Cintron VJ, Qiang X, Goustin AS, et al. Improved insulin sensitivity and resistance to weight gain in mice null for the Ahsg gene.

de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, et al. Omentin plasma levels and gene expression are decreased in obesity. Reinehr T, Roth CL.

Fetuin-A and its relation to metabolic syndrome and fatty liver disease in obese children before and after weight loss. Circulating fetuin-A and free fatty acids interact to predict insulin resistance in humans.

Olivier E, Soury E, Ruminy P, Husson A, Parmentier F, Daveau M, et al. Fetuin-B, a second member of the fetuin family in mammals. Biochem J. Walker GE, Follenzi A, Bruscaggin V, Manfredi M, Bellone S, Marengo E, et al.

Fetuin B links vitamin D deficiency and pediatric obesity: direct negative regulation by vitamin D. J Steroid Biochem Mol Biol. Motsenbocker MA, Tappel AL. A selenocysteine-containing selenium-transport protein in rat plasma.

Biochim Biophys Acta. Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start.

Nat Rev Drug Discov. Sethi JK, Hotamisligil GS. The role of TNF alpha in adipocyte metabolism. Semin Cell Dev Biol.

Xu H, Uysal KT, Becherer JD, Arner P, Hotamisligil GS. Altered tumor necrosis factor-alpha TNF-alpha processing in adipocytes and increased expression of transmembrane TNF-alpha in obesity. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance.

Hu FB, Meigs JB, Li TY, Rifai N, Manson JE. Inflammatory markers and risk of developing type 2 diabetes in women. Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes.

Trends Endocrinol Metab. Lang CH, Dobrescu C, Bagby GJ. Tumor necrosis factor impairs insulin action on peripheral glucose disposal and hepatic glucose output. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. Liu C, Feng X, Li Q, Wang Y, Li Q, Hua M.

Adiponectin, TNF-alpha and inflammatory cytokines and risk of type 2 diabetes: a systematic review and meta-analysis. Butcher MJ, Hallinger D, Garcia E, Machida Y, Chakrabarti S, Nadler J, et al. Association of proinflammatory cytokines and islet resident leucocytes with islet dysfunction in type 2 diabetes.

Olson NC, Callas PW, Hanley AJ, Festa A, Haffner SM, Wagenknecht LE, et al. Circulating levels of TNF-alpha are associated with impaired glucose tolerance, increased insulin resistance, and ethnicity: the Insulin Resistance Atherosclerosis Study.

Banerjee M, Saxena M. Interleukin-1 IL-1 family of cytokines: role in type 2 diabetes. Clin Chim Acta. Major CD, Wolf BA. Interleukin-1beta stimulation of c-Jun NH 2 -terminal kinase activity in insulin-secreting cells: evidence for cytoplasmic restriction. Ammendrup A, Maillard A, Nielsen K, Aabenhus Andersen N, Serup P, Dragsbaek Madsen O, et al.

The c-Jun amino-terminal kinase pathway is preferentially activated by interleukin-1 and controls apoptosis in differentiating pancreatic beta-cells.

Kaneto H, Xu G, Fujii N, Kim S, Bonner-Weir S, Weir GC. Involvement of c-Jun N-terminal kinase in oxidative stress-mediated suppression of insulin gene expression. Jager J, Gremeaux T, Cormont M, Le Marchand-Brustel Y, Tanti JF. Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression.

Miura K, Kodama Y, Inokuchi S, Schnabl B, Aoyama T, Ohnishi H, et al. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Tilg H, Moschen AR, Szabo G. Kirii H, Niwa T, Yamada Y, Wada H, Saito K, Iwakura Y, et al.

Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol.

Osborn O, Brownell SE, Sanchez-Alavez M, Salomon D, Gram H, Bartfai T. Treatment with an Interleukin 1 beta antibody improves glycemic control in diet-induced obesity. Salmenniemi U, Ruotsalainen E, Pihlajamaki J, Vauhkonen I, Kainulainen S, Punnonen K, et al. Multiple abnormalities in glucose and energy metabolism and coordinated changes in levels of adiponectin, cytokines, and adhesion molecules in subjects with metabolic syndrome.

Moschen AR, Molnar C, Enrich B, Geiger S, Ebenbichler CF, Tilg H. Adipose and liver expression of interleukin IL -1 family members in morbid obesity and effects of weight loss.

Mol Med. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukinreceptor antagonist in type 2 diabetes mellitus.

N Engl J Med. Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol. Rose-John S.

IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL Int J Biol Sci. Schaper F, Rose-John S. Interleukin Biology, signaling and strategies of blockade.

Cytokine Growth Factor Rev. Kraakman MJ, Kammoun HL, Allen TL, Deswaerte V, Henstridge DC, Estevez E, et al. Blocking IL-6 trans-signaling prevents high-fat diet-induced adipose tissue macrophage recruitment but does not improve insulin resistance.

Rotter V, Nagaev I, Smith U. Interleukin-6 IL-6 induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. Timper K, Denson JL, Steculorum SM, Heilinger C, Engstrom-Ruud L, Wunderlich CM, et al.

IL-6 improves energy and glucose homeostasis in obesity via enhanced central IL-6 trans-signaling. Cell Rep. Stouthard JM, Oude Elferink RP, Sauerwein HP. Interleukin-6 enhances glucose transport in 3T3-L1 adipocytes. Lagathu C, Bastard JP, Auclair M, Maachi M, Capeau J, Caron M.

Chronic interleukin-6 IL-6 treatment increased IL-6 secretion and induced insulin resistance in adipocyte: prevention by rosiglitazone.

Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. Park J, Ryu DR, Li JJ, Jung DS, Kwak SJ, Lee SH, et al.

Am J Physiol Renal Physiol. Sung FL, Zhu TY, Au-Yeung KK, Siow YL. Kidney Int B. Chen J, Guo Y, Zeng W, Huang L, Pang Q, Nie L, et al. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity.

Nio Y, Yamauchi T, Iwabu M, Okada-Iwabu M, Funata M, Yamaguchi M, et al. Monocyte chemoattractant protein-1 MCP-1 deficiency enhances alternatively activated M2 macrophages and ameliorates insulin resistance and fatty liver in lipoatrophic diabetic A-ZIP transgenic mice.

Ahmed SF, Shabayek MI, Abdel Ghany ME, El-Hefnawy MH, El-Mesallamy HO Role of CTRP3 CTRP9 and MCP-1 for the evaluation of T2DM associated coronary artery disease in Egyptian postmenopausal females.

PLoS ONE. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al.

A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Roca-Rivada A, Castelao C, Senin LL, Landrove MO, Baltar J, Belen Crujeiras A, et al.

Huh JY, Mougios V, Kabasakalis A, Fatouros I, Siopi A, Douroudos II, et al. Exercise-induced irisin secretion is independent of age or fitness level and increased irisin may directly modulate muscle metabolism through AMPK activation.

Xiong XQ, Geng Z, Zhou B, Zhang F, Han Y, Zhou YB, et al. FNDC5 attenuates adipose tissue inflammation and insulin resistance via AMPK-mediated macrophage polarization in obesity. Seale P, Conroe HM, Estall J, Kajimura S, Frontini A, Ishibashi J, et al.

Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice. Moreno-Navarrete JM, Ortega F, Serrano M, Guerra E, Pardo G, Tinahones F, et al.

Irisin is expressed and produced by human muscle and adipose tissue in association with obesity and insulin resistance. Rutti S, Howald C, Arous C, Dermitzakis E, Halban PA, Bouzakri K.

IL improves beta-cell survival and protects against IL-1beta-induced beta-cell death. Mol Metab. Stanya KJ, Jacobi D, Liu S, Bhargava P, Dai L, Gangl MR, et al. Direct control of hepatic glucose production by interleukin in mice. Jiang LQ, Franck N, Egan B, Sjogren RJ, Katayama M, Duque-Guimaraes D, et al.

Autocrine role of interleukin on skeletal muscle glucose metabolism in type 2 diabetic patients involves microRNA let Grabstein KH, Eisenman J, Shanebeck K, Rauch C, Srinivasan S, Fung V, et al.

Cloning of a T cell growth factor that interacts with the beta chain of the interleukin-2 receptor. Sun H, Liu D. Hydrodynamic delivery of interleukin 15 gene promotes resistance to high fat diet-induced obesity, fatty liver and improves glucose homeostasis.

Gene Ther. Barra NG, Reid S, MacKenzie R, Werstuck G, Trigatti BL, Richards C, et al. Interleukin contributes to the regulation of murine adipose tissue and human adipocytes. Nielsen AR, Hojman P, Erikstrup C, Fischer CP, Plomgaard P, Mounier R, et al. Association between interleukin and obesity: interleukin as a potential regulator of fat mass.

Matthews VB, Astrom MB, Chan MH, Bruce CR, Krabbe KS, Prelovsek O, et al. Brain-derived neurotrophic factor is produced by skeletal muscle cells in response to contraction and enhances fat oxidation via activation of AMP-activated protein kinase. Karczewska-Kupczewska M, Kowalska I, Nikolajuk A, Adamska A, Zielinska M, Kaminska N, et al.

Diabetes Care. Pedersen BK. The diseasome of physical inactivity—and the role of myokines in muscle—fat cross talk. J Physiol. Tsuchida A, Nakagawa T, Itakura Y, Ichihara J, Ogawa W, Kasuga M, et al.

The effects of brain-derived neurotrophic factor on insulin signal transduction in the liver of diabetic mice. Wei J, Shimazu J, Makinistoglu MP, Maurizi A, Kajimura D, Zong H, et al.

Glucose Uptake and Runx2 synergize to orchestrate osteoblast differentiation and bone formation. Lee NK, Sowa H, Hinoi E, Ferron M, Ahn JD, Confavreux C, et al. Endocrine regulation of energy metabolism by the skeleton.

Kim SP, Frey JL, Li Z, Kushwaha P, Zoch ML, Tomlinson RE, et al. Sclerostin influences body composition by regulating catabolic and anabolic metabolism in adipocytes. Chen L, Li Q, Yang Z, Ye Z, Huang Y, He M, et al. Osteocalcin, glucose metabolism, lipid profile and chronic low-grade inflammation in middle-aged and elderly Chinese.

Diabet Med. Mera P, Laue K, Ferron M, Confavreux C, Wei J, Galan-Diez M, et al. Osteocalcin signaling in myofibers is necessary and sufficient for optimum adaptation to exercise. Khrimian L, Obri A, Ramos-Brossier M, Rousseaud A, Moriceau S, Nicot AS, et al.

Gpr mediates osteocalcin's regulation of cognition. J Exp Med. Wei J, Hanna T, Suda N, Karsenty G, Ducy P. Osteocalcin promotes beta-cell proliferation during development and adulthood through Gprc6a. Otani T, Mizokami A, Hayashi Y, Gao J, Mori Y, Nakamura S, et al.

Signaling pathway for adiponectin expression in adipocytes by osteocalcin. Fulzele K, Riddle RC, DiGirolamo DJ, Cao X, Wan C, Chen D, et al. Insulin receptor signaling in osteoblasts regulates postnatal bone acquisition and body composition.

Shu H, Pei Y, Chen K, Lu J. Significant inverse association between serum osteocalcin and incident type 2 diabetes in a middle-aged cohort. Diabetes Metab Res Rev. Hwang YC, Jee JH, Jeong IK, Ahn KJ, Chung HY, Lee MK. Circulating osteocalcin level is not associated with incident type 2 diabetes in middle-aged male subjects: mean 8.

Higashibata Y, Sakuma T, Kawahata H, Fujihara S, Moriyama K, Okada A, et al. Identification of promoter regions involved in cell- and developmental stage-specific osteopontin expression in bone, kidney, placenta, and mammary gland: an analysis of transgenic mice.

J Bone Miner Res. Kiefer FW, Neschen S, Pfau B, Legerer B, Neuhofer A, Kahle M, et al. Osteopontin deficiency protects against obesity-induced hepatic steatosis and attenuates glucose production in mice.

Zeyda M, Gollinger K, Todoric J, Kiefer FW, Keck M, Aszmann O, et al. Osteopontin is an activator of human adipose tissue macrophages and directly affects adipocyte function.

Ono M, Itakura Y, Nonomura T, Nakagawa T, Nakayama C, Taiji M, et al. Intermittent administration of brain-derived neurotrophic factor ameliorates glucose metabolism in obese diabetic mice. Sodhi CP, Phadke SA, Batlle D, Sahai A. Hypoxia stimulates osteopontin expression and proliferation of cultured vascular smooth muscle cells: potentiation by high glucose.

Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron.

Yan QW, Yang Q, Mody N, Graham TE, Hsu CH, Xu Z, et al. The adipokine lipocalin 2 is regulated by obesity and promotes insulin resistance.

Paton CM, Rogowski MP, Kozimor AL, Stevenson JL, Chang H, Cooper JA. Lipocalin-2 increases fat oxidation in vitro and is correlated with energy expenditure in normal weight but not obese women. Lemma S, Sboarina M, Porporato PE, Zini N, Sonveaux P, Di Pompo G, et al.

Energy metabolism in osteoclast formation and activity. Int J Biochem Cell Biol. Rashad NM, El-Shal AS, Etewa RL, Wadea FM. Lipocalin-2 expression and serum levels as early predictors of type 2 diabetes mellitus in obese women.

IUBMB Life. Ye Z, Wang S, Yang Z, He M, Zhang S, Zhang W, et al. Serum lipocalin-2, cathepsin S and chemerin levels and nonalcoholic fatty liver disease. Mol Biol Rep. Wang Y, Lam KS, Kraegen EW, Sweeney G, Zhang J, Tso AW, et al.

Lipocalin-2 is an inflammatory marker closely associated with obesity, insulin resistance, and hyperglycemia in humans. Clin Chem. Garcia-Martin A, Rozas-Moreno P, Reyes-Garcia R, Morales-Santana S, Garcia-Fontana B, Garcia-Salcedo JA, et al. Circulating levels of sclerostin are increased in patients with type 2 diabetes mellitus.

Urano T, Shiraki M, Ouchi Y, Inoue S. Association of circulating sclerostin levels with fat mass and metabolic disease—related markers in Japanese postmenopausal women. Daniele G, Winnier D, Mari A, Bruder J, Fourcaudot M, Pengou Z, et al.

Sclerostin and insulin resistance in prediabetes: evidence of a cross talk between bone and glucose metabolism. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, et al. Klotho converts canonical FGF receptor into a specific receptor for FGF Mirza MA, Alsio J, Hammarstedt A, Erben RG, Michaelsson K, Tivesten A, et al.

Circulating fibroblast growth factor is associated with fat mass and dyslipidemia in two independent cohorts of elderly individuals. Silswal N, Touchberry CD, Daniel DR, McCarthy DL, Zhang S, Andresen J, et al.

FGF23 directly impairs endothelium-dependent vasorelaxation by increasing superoxide levels and reducing nitric oxide bioavailability.

Hesse M, Frohlich LF, Zeitz U, Lanske B, Erben RG. Ablation of vitamin D signaling rescues bone, mineral, and glucose homeostasis in Fgf deficient mice. Matrix Biol. Streicher C, Zeitz U, Andrukhova O, Rupprecht A, Pohl E, Larsson TE, et al.

Long-term Fgf23 deficiency does not influence aging, glucose homeostasis, or fat metabolism in mice with a nonfunctioning vitamin D receptor. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. Fasshauer M, Bluher M.

Adipokines in health and disease. Trends Pharmacol Sci. Perez-Perez A, Vilarino-Garcia T, Fernandez-Riejos P, Martin-Gonzalez J, Segura-Egea JJ, Sanchez-Margalet V. Role of leptin as a link between metabolism and the immune system. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM.

Positional cloning of the mouse obese gene and its human homologue. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals.

Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O'Kirwan F, et al. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults.

Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, et al. Leptin-replacement therapy for lipodystrophy. Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, et al. Wannamethee SG, Lowe GD, Rumley A, Cherry L, Whincup PH, Sattar N. Adipokines and risk of type 2 diabetes in older men.

Myers MG, Cowley MA, Munzberg H. Mechanisms of leptin action and leptin resistance. Annu Rev Physiol. Coppari R, Ichinose M, Lee CE, Pullen AE, Kenny CD, McGovern RA, et al. The hypothalamic arcuate nucleus: a key site for mediating leptin's effects on glucose homeostasis and locomotor activity.

Mittendorfer B, Horowitz JF, DePaoli AM, McCamish MA, Patterson BW, Klein S. Recombinant human leptin treatment does not improve insulin action in obese subjects with type 2 diabetes. Polyzos SA, Kountouras J, Zavos C, Tsiaousi E. The role of adiponectin in the pathogenesis and treatment of non-alcoholic fatty liver disease.

Jung TW, Lee YJ, Lee MW, Kim SM, Jung TW. Full-length adiponectin protects hepatocytes from palmitate-induced apoptosis via inhibition of c-Jun NH2 terminal kinase. Febs J. Polyzos SA, Kountouras J, Mantzoros CS. Adipokines in nonalcoholic fatty liver disease. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al.

Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Chen YL, Tao J, Zhao PJ, Tang W, Xu JP, Zhang KQ, et al. Adiponectin receptor PAQR-2 signaling senses low temperature to promote C.

elegans longevity by regulating autophagy. Nat Commun. Savage DB, Sewter CP, Klenk ES, Segal DG, Vidal-Puig A, Considine RV, et al. Costandi J, Melone M, Zhao A, Rashid S.

Human resistin stimulates hepatic overproduction of atherogenic ApoB-containing lipoprotein particles by enhancing ApoB stability and impairing intracellular insulin signaling.

Circ Res. Banerjee RR, Rangwala SM, Shapiro JS, Rich AS, Rhoades B, Qi Y, et al. Regulation of fasted blood glucose by resistin. Greenhill C. Liver: asprosin - new hormone involved in hepatic glucose release. Romere C, Duerrschmid C, Bournat J, Constable P, Jain M, Xia F, et al. Asprosin, a fasting-induced glucogenic protein hormone.

Duerrschmid C, He Y, Wang C, Li C, Bournat JC, Romere C, et al. Asprosin is a centrally acting orexigenic hormone. Zhang L, Chen C, Zhou N, Fu Y, Cheng X.

Circulating asprosin concentrations are increased in type 2 diabetes mellitus and independently associated with fasting glucose and triglyceride. Zabel BA, Allen SJ, Kulig P, Allen JA, Cichy J, Handel TM, et al. Chemerin activation by serine proteases of the coagulation, fibrinolytic, and inflammatory cascades.

Horn P, Metzing UB, Steidl R, Romeike B, Rauchfuss F, Sponholz C, et al. Chemerin in peritoneal sepsis and its associations with glucose metabolism and prognosis: a translational cross-sectional study. Crit Care. Fernandez-Trasancos A, Agra RM, Garcia-Acuna JM, Fernandez AL, Gonzalez-Juanatey JR, Eiras S.

Omentin treatment of epicardial fat improves its anti-inflammatory activity and paracrine benefit on smooth muscle cells. Pan HY, Guo L, Li Q. Changes of serum omentin-1 levels in normal subjects and in patients with impaired glucose regulation and with newly diagnosed and untreated type 2 diabetes.

Diabetes Res Clin Pract. Liang Q, Zhong L, Zhang J, Wang Y, Bornstein SR, Triggle CR, et al. FGF21 maintains glucose homeostasis by mediating the cross talk between liver and brain during prolonged fasting.

Wente W, Efanov AM, Brenner M, Kharitonenkov A, Koster A, Sandusky GE, et al. Mathews ST, Rakhade S, Zhou X, Parker GC, Coscina DV, Grunberger G. Fetuin-null mice are protected against obesity and insulin resistance associated with aging. Srinivas PR, Wagner AS, Reddy LV, Deutsch DD, Leon MA, Goustin AS, et al.

Serum alpha 2-HS-glycoprotein is an inhibitor of the human insulin receptor at the tyrosine kinase level. Mori K, Emoto M, Yokoyama H, Araki T, Teramura M, Koyama H, et al. Association of serum fetuin-A with insulin resistance in type 2 diabetic and nondiabetic subjects. Choi HY, Hwang SY, Lee CH, Hong HC, Yang SJ, Yoo HJ, et al.

Increased selenoprotein p levels in subjects with visceral obesity and nonalcoholic Fatty liver disease. Diabetes Metab J. Misu H, Takamura T, Takayama H, Hayashi H, Matsuzawa-Nagata N, Kurita S, et al.

A liver-derived secretory protein, selenoprotein P, causes insulin resistance. Misu H, Ishikura K, Kurita S, Takeshita Y, Ota T, Saito Y, et al. Inverse correlation between serum levels of selenoprotein P and adiponectin in patients with type 2 diabetes.

Yang SJ, Hwang SY, Choi HY, Yoo HJ, Seo JA, Kim SG, et al. Serum selenoprotein P levels in patients with type 2 diabetes and prediabetes: implications for insulin resistance, inflammation, and atherosclerosis.

Chawla A, Nguyen KD, Goh YPS. Macrophage-mediated inflammation in metabolic disease. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS.

Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nov O, Shapiro H, Ovadia H, Tarnovscki T, Dvir I, Shemesh E, et al. Interleukin-1beta regulates fat-liver crosstalk in obesity by auto-paracrine modulation of adipose tissue inflammation and expandability.

Mirea AM, Tack CJ, Chavakis T, Joosten LAB, Toonen EJM. IL-1 Family Cytokine Pathways Underlying NAFLD: towards new treatment strategies. Trends Mol Med. Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka HI, Spinas GA, et al.

Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. Rotter Sopasakis V, Larsson BM, Johansson A, Holmang A, Smith U.

Short-term infusion of interleukin-6 does not induce insulin resistance in vivo or impair insulin signalling in rats. Quan Y, Jiang CT, Xue B, Zhu SG, Wang X. High glucose stimulates TNFalpha and MCP-1 expression in rat microglia via ROS and NF-kappaB pathways. Acta Pharmacol Sin. Mo L, Shen J, Liu Q, Zhang Y, Kuang J, Pu S, et al.

Irisin is regulated by CAR in liver and is a mediator of hepatic glucose and lipid metabolism. Rana KS, Pararasa C, Afzal I, Nagel DA, Hill EJ, Bailey CJ, et al.

Plasma irisin is elevated in type 2 diabetes and is associated with increased E-selectin levels. Cardiovasc Diabetol. Almendro V, Fuster G, Busquets S, Ametller E, Figueras M, Argiles JM, et al.

Effects of IL on rat brown adipose tissue: uncoupling proteins and PPARs. Barra NG, Chew MV, Holloway AC, Ashkar AA. Interleukin treatment improves glucose homeostasis and insulin sensitivity in obese mice.

Duan Y, Li F, Wang W, Guo Q, Wen C, Li Y, et al. Interleukin in obesity and metabolic dysfunction: current understanding and future perspectives.

Obes Rev. Kernie SG, Liebl DJ, Parada LF. BDNF regulates eating behavior and locomotor activity in mice. Embo j. Krabbe KS, Nielsen AR, Krogh-Madsen R, Plomgaard P, Rasmussen P, Erikstrup C, et al.

Brain-derived neurotrophic factor BDNF and type 2 diabetes. Li B, Lang N, Cheng ZF. Serum levels of brain-derived neurotrophic factor are associated with diabetes risk, complications, and obesity: a cohort study from chinese patients with type 2 diabetes.

Mol Neurobiol. Mosialou I, Shikhel S, Liu JM, Maurizi A, Luo N, He Z, et al. MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Komarova SV, Ataullakhanov FI, Globus RK. Bioenergetics and mitochondrial transmembrane potential during differentiation of cultured osteoblasts.

Am J Physiol Cell Physiol. Zoch ML, Abou DS, Clemens TL, Thorek DL, Riddle RC. In vivo radiometric analysis of glucose uptake and distribution in mouse bone. Bone Res. Esen E, Chen J, Karner CM, Okunade AL, Patterson BW, Long F. WNT-LRP5 signaling induces Warburg effect through mTORC2 activation during osteoblast differentiation.

Ferron M, Wei J, Yoshizawa T, Del Fattore A, DePinho RA, Teti A, et al. Insulin signaling in osteoblasts integrates bone remodeling and energy metabolism.

Mizokami A, Yasutake Y, Higashi S, Kawakubo-Yasukochi T, Chishaki S, Takahashi I, et al. Oral administration of osteocalcin improves glucose utilization by stimulating glucagon-like peptide-1 secretion.

Kiefer FW, Zeyda M, Gollinger K, Pfau B, Neuhofer A, Weichhart T, et al. Neutralization of osteopontin inhibits obesity-induced inflammation and insulin resistance.

Glass O, Henao R, Patel K, Guy CD, Gruss HJ, Syn WK, et al. Serum Interleukin-8, Osteopontin, and Monocyte Chemoattractant Protein 1 Are Associated With Hepatic Fibrosis in Patients With Nonalcoholic Fatty Liver Disease. Hepatol Commun.

Takemoto M, Yokote K, Nishimura M, Shigematsu T, Hasegawa T, Kon S, et al. Enhanced expression of osteopontin in human diabetic artery and analysis of its functional role in accelerated atherogenesis.

Xu Y, Ma X, Pan X, He X, Xiao Y, Bao Y. Correlations between serum concentration of three bone-derived factors and obesity and visceral fat accumulation in a cohort of middle aged men and women. Ukita M, Yamaguchi T, Ohata N, Tamura M.

Sclerostin enhances adipocyte differentiation in 3T3-L1 cells. J Cell Biochem. Hu X, Ma X, Luo Y, Xu Y, Xiong Q, Pan X, et al. Associations of serum fibroblast growth factor 23 levels with obesity and visceral fat accumulation.

Clin Nutr. Gateva A, Assyov Y, Tsakova A, Kamenov Z. Prediabetes is characterized by higher FGF23 levels and higher prevalence of Vitamin D deficiency compared to normal glucose tolerance subjects. Horm Metab Res. Wahl P, Xie H, Scialla J, Anderson CA, Bellovich K, Brecklin C, et al.

Earlier onset and greater severity of disordered mineral metabolism in diabetic patients with chronic kidney disease. Reyes-Garcia R, Garcia-Martin A, Garcia-Fontana B, Morales-Santana S, Rozas-Moreno P, Munoz-Torres M.

FGF23 in type 2 diabetic patients: relationship with bone metabolism and vascular disease. Keywords: cytokine, glucose metabolism, lipid metabolism, insulin resistance, inflammation. Citation: Shi J, Fan J, Su Q and Yang Z Cytokines and Abnormal Glucose and Lipid Metabolism.

Received: 28 June ; Accepted: 30 September ; Published: 30 October Copyright © Shi, Fan, Su and Yang. This is an open-access article distributed under the terms of the Creative Commons Attribution License CC BY.

The use, distribution or reproduction in other forums is permitted, provided the original author s and the copyright owner s are credited and that the original publication in this journal is cited, in accordance with accepted academic practice.

No use, distribution or reproduction is permitted which does not comply with these terms. cn ; Zhen Yang, yangzhen xinhuamed. Importantly, as metabolic regulation by glucose occurs mainly at the transcriptional level [22] , we assessed the mRNA levels of target genes involved in glucose transport, glycolysis, gluconeogenesis, lipogenesis and fatty acid β-oxidation, in liver and muscle, at the three post-injection time intervals.

It was approved by the Direction Departementale des Services Veterinaires French veterinary services to carry out animal experiments INRA —36, April 14th, The INRA experimental station is certified for animal services under the permit number A The study was conducted with two lines of rainbow trout Oncorhynchus mykiss , Walbaum , designated as Lean line L and Fat line F , obtained after five generations of divergent selection for high or low muscle fat content using a nondestructive method Distell Fish Fatmeter as detailed by Quillet et al.

Muscle fat content was found more than 3 times higher in the F line than in the L in g-trout of this fifth generation [37]. Fish were kept unfed for 48 h before the time of treatment in order to obtain fish with basal levels of plasma metabolites and an empty digestive tract.

Fish were lightly anaesthetized with 0. Blood was quickly removed from the caudal vein using syringes rinsed with an anticoagulant 0.

All fish were sampled at the expected time ±5 min. Frozen samples of liver mg and muscle mg were homogenized in 2 ml of buffer containing mM NaCl, 10 mM Tris, 1 mM EGTA, 1 mM EDTA pH 7.

Tubes were kept in ice during the whole process to avoid protein denaturation. Homogenates were centrifuged at g for 15 min at 4°C and supernatants were again centrifuged at 20, g for 30 min. The concentration of protein in each sample was determined using Bio-Rad protein assay kit Bio-Rad Laboratories, Munich, Germany with bovine serum albumin as standard.

Liver and muscle protein lysates 10 µg of protein for Akt; 20 µg for AMPK were subjected to SDS-PAGE and Western blotting using appropriate antibodies. Anti-phospho Akt Ser , anti-carboxyl terminal Akt, anti-phospho AMPK Thr and anti-AMPK antibodies were used Cell signaling Technology, Saint Quentin Yvelings, France.

All these antibodies have been shown to cross-react successfully with rainbow trout proteins of interest [16] , [33]. After washing, membranes were incubated with an IRDye infrared secondary antibody LI-COR Biosciences and spots were quantified by Odyssey Infrared Imaging System software Version 3.

One microgram of the resulting total RNA was reverse transcribed into cDNA using the SuperScript III RNaseH-reverse transcriptase kit Invitrogen and random primers Promega, Charbonnières, France according to the instructions of each manufacturer, including no RNA and no reverse transcriptase control.

Target gene expression levels were determined by quantitative real-time RT-PCR, using specific primers [32] , [33] , [39] , [40]. Quantitative RT-PCR was carried out on a LightCycle II Roche Diagnostics, Neuilly-sur-Seine, France using SYBR Green I Master Roche Diagnostics GmbH, Mannheim, Germany.

The transcripts assessed were glucokinase GK , 6-phosphofructokinase 6PFK and pyruvate kinase PK for glycolysis; glucosephosphatase G6Pase1 and G6Pase2 , fructose 1,6-bisphosphatase FBPase and phosphoenolpyruvate carboxykinase PEPCK for gluconeogenesis; glucose facilitative transporter type 2 GLUT2 and 4 GLUT4 for glucose transport; sterol regulatory element binding protein 1c SREBP1c for transcription factor regulating lipogenesis; glucose 6-phosphate dehydrogenase G6PD , ATP citrate lyase ACLY , acetyl-CoA carboxylase ACC and fatty acid synthase FAS for de novo lipogenesis; carnitine palmitoyl transferase 1 CPT1a and CPT1b , 3-hydroxyacyl-CoA dehydrogenase HOAD and acyl-CoA oxidase ACO for fatty acid β-oxidation.

No template control was applied for each primer. Relative quantification of target gene expression was performed using the mathematical model described by Pfaffl [41].

Results are mostly expressed as means ± SD. When an interaction was significant, means were compared using the Student-Newman-Keuls multiple comparison test. Plasma concentration of specific metabolites at 3, 8 and 12 h after intraperitoneal injection of glucose or saline solution are presented in Table 1.

In both lines, glucose administration resulted in a pronounced and persistent hyperglycemia, as observed at 3 h post-injection. Thereafter at 8 h post-injection, glycemia had returned to basal values. However, L line exhibited hyperglycemia again at 12 h after glucose treatment, in contrast to the normal glycemia observed in F line.

Plasma triglycerides followed a time-response pattern similar to glycemia profile in L line at 12 h post-glucose treatment. In the saline injected fish, F line was found to have higher plasma triglyceride and free fatty acid levels than L line, but no such genotypic difference was observed in the glucose treated fish.

Regarding tissue glycogen content Fig. Glucose treatment was linked to liver glycogen depletion in L line, whereas in F line, it was associated with unaltered liver glycogen content in F line. With regard to glycogen content in muscle, a slight but not significant decrease was found in L line, whereas an inverse effect was observed in F line.

In case of interaction, mean values not sharing a common lowercase letter are significantly different from each other. Changes in the phosphorylation status of components of the insulin signaling pathway Akt-TOR and major cellular energy sensor AMPK were analyzed in the liver and muscle of the two trout lines sampled 3 h after treatment, using Western blot.

No significant difference was noted in the ratio of phosphorylated to total protein of Akt and AMPK between the two genotypes as well as treatment, in both liver and muscle Fig.

Western blots were performed on 6 individual samples per treatment and a representative blot is shown here. Relative fold difference in the mRNA levels of hepatic glycolytic enzymes at 3, 8 and 12 h post-injection is shown in Fig.

Compared to L line, the transcripts of 6PFK and PK were also more abundant in the F line at 8 h post-injection, irrespective of saline or glucose treatment. Hepatic PK expression however remained enhanced in F line at 12 h. GLUT2 mRNA level in the liver was not modified by glucose treatment and did not vary between the two genotypes.

Glucokinase GK , 6-phosphofructokinase 6PFK , pyruvate kinase PK and glucose transporter type 2 GLUT2 mRNA levels were measured using real-time quantitative RT-PCR. Expression values were normalized by 18S-ribosomal RNA transcripts.

Relative transcript levels of key gluconeogenic enzymes in the liver are presented in Fig. In both lines, glucose treatment was associated with a decrease in the expression of G6Pase1 at 8 h post-injection, but conversely it was found to enhance the expression of FBPase and PEPCK at 3 h post-injection.

Particularly, PEPCK transcripts were more abundant in F line than L line, irrespective of treatment. In addition, disordinal interaction between treatment and line was observed for G6Pase1, G6Pase2 and FBPase mRNA levels at 3 h post-injection, where glucose loading up-regulated the expression of these enzymes specifically in the L line.

Glucosephosphatase isoform 1 G6Pase1 and isoform 2 G6Pase2 , fructose 1,6-bisphosphatase FBPase and phosphoenolpyruvate carboxykinase PEPCK mRNA levels were measured using real-time quantitative RT-PCR. Concerning the expression of glycolytic markers in white muscle Fig. Apart from that, no other changes were found in the transcript levels of HK, 6PFK, PK and GLUT4 due to treatment or genotype.

Hexokinase HK , 6-phosphofructokinase 6PFK , pyruvate kinase PK and glucose transporter type 4 GLUT4 mRNA levels were measured using real-time quantitative RT-PCR.

Expression values were normalized by β-actin-transcripts. As represented in Fig. Similar induction was observed also for ACLY expression, but only in the L line, leading to a significant interaction.

Transcripts of SREBP1c, ACC and FAS were not significantly influenced by the glucose load. Except for an enhanced expression of ACC in the F line at 8 h post-injection, no genotypic difference was noted in the target lipogenic markers. Concerning fatty acid β-oxidation in the liver Fig.

Disordinal treatment × line interaction was observed in HOAD and ACO expression at 3 h post treatment, with a significant induction in the L line after a glucose load. Sterol regulatory element binding protein 1c SREBP1c , glucose 6-phosphate dehydrogenase G6PD , ATP citrate lyase ACLY , acetyl-CoA carboxylase ACC and fatty acid synthase FAS mRNA levels were measured using real-time quantitative RT-PCR.

Carnitine palmitoyl transferase isoform 1 a CPT1a and b CPT1b , 3-hydroxyacyl-CoA dehydrogenase HOAD and acyl-CoA oxidase ACO mRNA levels were measured using real-time quantitative RT-PCR.

Mean values not sharing a common lowercase letter are significantly different from each other. Changes in relative mRNA levels of key enzymes involved in muscle fatty acid oxidation are shown in Fig.

Glucose administration was found to down-regulate the expression of CPT1 isoforms, HOAD and ACO in both lines, at 3 or 12 h post-injection, irrespective of the treatment. F line exhibited consistently higher CPT1a expression than L line at all the post-injection time points.

Similar genotypic difference was observed also in the transcript levels of CPT1b and HOAD at 12 h post-injection. Finally, a significant treatment × line interaction was observed in ACO expression at 3 h post-injection, where glucose loading reduced ACO expression specifically in F line.

Previous nutritional studies in the two selected trout lines suggested that the F line displayed a higher ability to metabolize glucose and to synthesize lipids de novo than the L line [16] , [32] , [33].

We thus postulated that the F line has a better capability to maintain glucose homeostasis after a dietary glucose load than L line. To check this hypothesis, the two rainbow trout lines were intraperitoneally injected with glucose or saline solution to investigate the genotypic differences in the regulation of glucose and lipid metabolism after a high glucose flux.

This study is clearly different from our previous study based on the use of dietary carbohydrates [16] related to the route of the glucose supplementation intake of dietary carbohydrates [16] versus intraperitoneal glucose injection and the nutritional status of the fish fed fish [16] versus unfed fish.

Moreover, the present study describes the potential differential regulation of metabolism by glucose between two rainbow trout lines which is highly interesting for a better understanding of the glucose use in carnivorous animals.

The time-dependent regulation of metabolic gene expression by glucose is different for each gene: some are differentially expressed already 3 h after injection the hepatic gluconeogenic genes for example and others are differently expressed only 12 h after injection fatty acid oxidation genes in muscle for example suggesting that the genes are not regulated by glucose with similar molecular mechanisms.

As expected, a strong and persistent hyperglycemia was induced after glucose treatment in both rainbow trout lines, confirming the poor ability of this species to regulate glucose homeostasis [19] , [42].

The high glycemia had no effect on the mRNA levels of hepatic glucose transporter GLUT2, probably due to the fact that this gene is present constitutively in the plasma membrane [43].

Moreover, the expression of GLUT2 has been observed to be high in rainbow trout, irrespective of the nutritional status [44]. Concerning hepatic glycolysis, Borrebaek et al. reported that high starch content in the diet up-regulated GK activity in Atlantic salmon [45].

Similarly in rainbow trout, a strong induction of GK expression was observed following the intake of a carbohydrate rich diet [42]. With respect to gluconeogenesis, glucose was found to enhance the transcript levels of FBPase and PEPCK in the liver of both trout lines, reinforcing the fact that in carnivorous fish, glucose does not exert a mammalian like inhibitory effect on the gluconeogenic pathway [16] , [44] , [46] , [47].

In particular, the augmentation of the gluconeogenic potential that we observed after a glucose load in these two trout lines, have also been described in other carnivorous fish such as red sea bream and yellowtail [14]. Nevertheless, in concordance with the lowered glycemic values at 8 h after glucose treatment, hepatic G6Pase was found to be down-regulated in both lines.

In the white skeletal muscle of the glucose treated fish from both lines, we observed a paradoxical decrease in the expression of HK, which indicates an atypical regulation of muscle glucose metabolism in response to high influx of glucose, corroborating previous finding in salmonids [16] , [19].

This observation also serves as an evidence for the poor capacity of glucose utilization in the peripheral tissues of carnivorous fish [1] , irrespective of the genotype. Regarding the genotype effect, F line apparently had a more efficient and stable glucose clearance mechanism to deal with a glucose load than L line, in agreement with previous findings that reported significantly lower levels of glucose in the F line at 24 h after refeeding [33].

Moreover, the return of plasma glucose level to basal values at 8 h after glucose treatment in F line was relatively much faster than the 24 h glycemia recovery period previously reported in rainbow trout, after administering a similar dose of glucose [7].

These differences in glycemia profiles between the two lines after glucose challenge could be due to genetic difference as a side effect of the selection for the target trait, i.

the muscle fat content. Even if the existence of a genetic component that would determine the ability to utilize glucose is unknown in fish, it can be hypothesized, as suggested by the variation in glucose tolerance among three natural strains of Chinook salmon [30] and by selectively bred fast growing families of rainbow trout which have higher plasma insulin levels than their slow growing counterparts [48].

The higher efficiency of plasma glucose clearance in F line was basically associated with its enhanced hepatic glycolytic potential higher transcript levels of GK, 6PFK and PK , consistent with previous observations [16] , [32].

Overall, these data suggest that genetic selection could be a determinant of glucose homeostasis as in Chinook salmon [30]. However, it is important to note that, the transcript levels of gluconeogenic enzyme PEPCK were also higher in F line, suggesting a greater endogenous glucose synthesis potential than in L line.

Concomitant weaker control of gluconeogenesis and enhanced glycolytic potential was similarly observed in the 4th generation F line fish, irrespective of dietary carbohydrate levels [16]. In fish, liver is the major tissue that regulates glucose disposal [49] and generally, hepatic glycogen content is higher in fish fed a carbohydrate-rich diet than in those fed a carbohydrate-free diet due to excess glucose storage [4] , [50].

With respect to tissue glycogen deposition in both lines, we observed a significant interaction between glucose treatment and genotype in both liver and muscle. The hyperglycemia found in L line at 12 h after glucose treatment can be probably explained by an apparent lower glycolytic potential in the liver of this line compared to the F line concomitantly with a decreased potential of glycogen synthesis in liver at 8 h post-injection.

This in turn may be related to the lower muscle lipid content, i. On the other hand, normoglycemia was observed in F line at 12 h post-injection, in concordance with a slight but not statistically significant increase in muscle glycogen deposition.

These cumulative findings endorse our hypothesis that F line has a better ability to maintain glucose homeostasis than L line. Lipogenesis possibly plays a crucial role in the glucose homeostasis of rainbow trout, by converting excess glucose into lipids [4].

G6PD and ACLY are the key enzymes that provide NADPH for fatty acid biosynthesis and divert glycolytic carbon flux into lipid biosynthesis, respectively. We found that glucose administration enhanced the transcript levels of G6PD and ACLY, but not those of FAS and SREBP1c, similar to the observation made after long term feeding of a carbohydrate rich diet in the 4th generation fish [16].

The absence of induction of SREBP1c and FAS expression after glucose treatment in the present study might be partly related to the 48 h-fasting applied before injection.

In mammals, glucose uptake inhibits fatty acid oxidation, both in mitochondria and peroxisome due to insulin secretion and action [52] , [53]. This discrepancy may be caused by the experimental differences between the two studies: intake of dietary carbohydrates [16] may be able to induce insulin secretion related to increase of blood glucose plus other incretin factors glucose-dependent insulinotropic polypeptide GIP and glucagon-like peptide 1 GLP-1 for example [54] which are not present after glucose injection.

Furthermore, glucose is known to stimulate hepatic lipolysis in rainbow trout, both in vivo and in vitro , by enhancing the activity of lipolytic enzymes [55] , [56].

Somatostatin was found to mediate the stimulation of hepatic lipolysis in several species such as rainbow trout, coho salmon and lamprey [57] , [58]. Hence, the glucose enhanced HOAD mRNA levels that we observed, might be resulting from elevated plasma somatostatin concentration.

The effect of glucose treatment on muscle fatty acid oxidation was different from our observations in the liver, confirming tissue specific regulation [59]. Expression of the genes encoding the fatty acid oxidation enzymes, i. CPT1a, CPT1b, HOAD and ACO were significantly inhibited by glucose in the muscle, differing from earlier reports of inertness in rainbow trout fed a high carbohydrate diet [16] , [27] , [59].

intake of a high carbohydrate diet and the nutritional status of the fish fasted vs. Inhibition of fatty acid oxidation markers when coupled together with the decrease in the level of plasma triglycerides, suggests that the external glucose load might trigger a switch from the use of fatty acids as fuel in fasted fish to the use of glucose as fuel in glucose treated fish.

In contrast to the higher lipogenic potential demonstrated in F line of the previous generation [16] , [33] , we found no genotypic differences in the target genes related to de novo lipogenesis, except for an elevated expression of ACC in the F line. Thus, the higher muscle fat content in F line cannot be strictly attributed to its lipogenic ability, as previously noted [32].

Likewise, increased muscle fat content in F line is also not due to a decrease in fatty acid oxidation capacity [32]. Conversely, we found an enhanced fatty acid oxidation potential especially at the fatty acid transport level with CPT1 in the liver and muscle of F line, contrary to our previous observations [16] , [32] , [33].

Even though we cannot eliminate a putative difference linked to the generation of selection 5th vs. Indeed, unfed fish use fatty acids as the preferential energy source [60] in contrast to fed fish; we can hypothesize that fatty acid potential could be strongly changed in this context in the fish lines.

Previous studies demonstrated that there were very few line × diet interactions for the markers of lipid metabolism in both 3rd and 4th generations of selection [16] , [32]. In agreement with this, limited but significant interactions were observed between glucose treatment and genotype in the hepatic expression of ACLY, HOAD and ACO, where glucose specifically enhanced their expression in the L line.

In particular, we can say that glucose affects hepatic fatty acid oxidation markers in a genotype specific manner. The present study mainly reveals the differences in the transcriptional regulation of glucose and lipid metabolism after a GTT between two genotypes of rainbow trout.

In both lines, intraperitoneal administration of glucose led to a prolonged hyperglycemia, this relative inability to utilize high levels of glucose was associated with a lack of regulation of gluconeogenesis.

However, compared to L line, regulation of glycemia was more stable in F line, linked to higher muscle glycogen content and an enhanced expression of glycolytic gene markers in the liver, after glucose treatment. Some of these observations are coherent with the findings of previous nutritional challenge studies [16] , [32] , [33] and support our hypothesis that the F line has a higher capability to maintain glucose homeostasis after a glucose load than the L line.

Our data are also helpful for understanding glucose use in carnivorous fish. Therefore, genetic selection is apparently a possible way to modify the poor capability of glucose hemostasis in carnivorous fish, primarily by influencing the regulation of key enzymes involved in glucose and lipid metabolism.

We thank W. Dai and T. Cerezo for sampling assistance. We thank K. Dias, A. Surget and A. Herman for technical assistance in the laboratory.

We also acknowledge the technical staff of the INRA experimental fish farm at Lees-Athas P. Maunas and N. Turonnet for fish rearing. Conceived and designed the experiments: SP FM.

Performed the experiments: JJ PA VV. Analyzed the data: JJ SP FM. Contributed to the writing of the manuscript: JJ SP FM BSK. Browse Subject Areas?

Click through the PLOS taxonomy to find articles in your field. Article Authors Metrics Comments Media Coverage Reader Comments Figures.

Planas, Universitat de Barcelona, Spain Received: March 31, ; Accepted: July 23, ; Published: August 20, Copyright: © Jin et al. Introduction Compared to mammals that have the ability to deal rapidly with a glucose load or a diet rich in carbohydrates, carnivorous teleost are generally recognized as poor users of glucose [1] — [4].

Experimental design and sampling procedure The study was conducted with two lines of rainbow trout Oncorhynchus mykiss , Walbaum , designated as Lean line L and Fat line F , obtained after five generations of divergent selection for high or low muscle fat content using a nondestructive method Distell Fish Fatmeter as detailed by Quillet et al.

Statistical analysis Results are mostly expressed as means ± SD. Results Plasma metabolites levels Plasma concentration of specific metabolites at 3, 8 and 12 h after intraperitoneal injection of glucose or saline solution are presented in Table 1.

Download: PPT. Table 1. Tissue glycogen content Regarding tissue glycogen content Fig. Figure 1. Glycogen content in liver A and muscle B of rainbow trout from L line L; grey bars and F line F; black bars , 3 h after IP administration of glucose Glu or saline Sal solution.

Insulin signaling pathway and cellular energy sensor AMPK Changes in the phosphorylation status of components of the insulin signaling pathway Akt-TOR and major cellular energy sensor AMPK were analyzed in the liver and muscle of the two trout lines sampled 3 h after treatment, using Western blot.

Figure 2. Western blot analysis of AMPK and Akt phosphorylation in rainbow trout liver A and muscle B samples from L line L; grey bars and F line F; black bars , 3 h after IP administration of glucose or saline solution.

Messenger RNA levels of target genes Relative fold difference in the mRNA levels of hepatic glycolytic enzymes at 3, 8 and 12 h post-injection is shown in Fig.

Figure 3. Gene expression of selected glycolytic enzymes and glucose transporter in the liver of rainbow trout from L line L; grey bars and F line F; black bars at 3 h A , 8 h B and 12 h C after IP administration of glucose or saline solution. Figure 4. Gene expression of selected gluconeogenic enzymes in the liver of rainbow trout from L line L; grey bars and F line F; black bars at 3 h A , 8 h B and 12 h C after IP administration of glucose or saline solution.

Figure 5. Gene expression of selected glycolytic enzymes and glucose transporter in the muscle of rainbow trout from L line L; grey bars and F line F; black bars at 3 h A , 8 h B and 12 h C after IP administration of glucose or saline solution.

Figure 6. Gene expression of selected transcription factors and enzymes involved in NADPH generation and lipogenesis in the liver of rainbow trout from L line L; grey bars and F line F; black bars at 3 h A , 8 h B and 12 h C after IP administration of glucose or saline solution.

Signalling mechanisms linking hepatic glucose and lipid metabolism Article CAS Google Metxbolism. Hepatic acetyl CoA links adipose tissue Lipid metabolism and glucose utilization to hepatic insulin emtabolism and type 2 diabetes. Tilg H, Moschen AR, Roden M. Duan Y, Li F, Wang W, Guo Q, Wen C, Li Y, et al. Svoboda, M. CAS PubMed PubMed Central Google Scholar Fischer, J. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase.
Glucose Homeostasis and Insulin Resistance

Nat Rev Mol Cell Biol 7, — Haemmerle, G. Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase.

ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-α and PGC Nat Med 17, — Halestrap, A. The SLC16 gene family—Structure, role and regulation in health and disease. The monocarboxylate transporter family-Role and regulation. IUBMB Life 64, — Hall, R. Regulation of phosphoenolpyruvate carboxykinase and insulin-like growth factor-binding protein-1 gene expression by insulin.

Han, J. The CREB coactivator CRTC2 controls hepatic lipid metabolism by regulating SREBP1. Hardie, D. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol 13, — Hariri, H. Mdm1 maintains endoplasmic reticulum homeostasis by spatially regulating lipid droplet biogenesis.

Harms, M. Brown and beige fat: development, function and therapeutic potential. Nat Med 19, — Hatting, M. Insulin regulation of gluconeogenesis. Ann NY Acad Sci , 21— He, G. ARH is a modular adaptor protein that interacts with the LDL receptor, clathrin, and AP Hediger, M.

The ABCs ofmembrane transporters in health and disease SLC series : introduction. Mol Aspect Med 34, 95— Heid, I. Genome-wide association analysis of high-density lipoprotein cholesterol in the population-based KORA study sheds new light on intergenic regions.

Circ Cardiovasc Genet 1, 10— Hemmings, B. Cold Spring Harb Perspect Biol 4, a Henderson, R. The genetics and screening of familial hypercholesterolaemia. J Biomed Sci 23, Henne, W. The assembly of lipid droplets and their roles in challenged cells. Hers, H. Gluconeogenesis and related aspects of glycolysis.

Annu Rev Biochem 52, — Herzig, S. CREB regulates hepatic gluconeogenesis through the coactivator PGC Hobbs, H. Molecular genetics of the LDL receptor gene in familial hypercholesterolemia. Hum Mutat 1, — Hoffmann, T. A large electronic-health-record-based genome-wide study of serum lipids.

Nat Genet 50, — Hogan, M. Hepatic insulin resistance following chronic activation of the CREB coactivator CRTC2. Horton, J. Molecular biology of PCSK9: its role in LDL metabolism. Trends Biochem Sci 32, 71— PCSK9: a convertase that coordinates LDL catabolism.

J Lipid Res 50, S—S SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Hossain, P. Obesity and diabetes in the developing world—a growing challenge.

Hua, X. Sterol resistance in CHO cells traced to point mutation in SREBP cleavage-activating protein. Cell 87, — 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. Am J Physiol E1—E McGarry JD Banting lecture dysregulation of fatty acid metabolism in the etiology of type 2 diabetes.

Vaulont S, Vasseur-Cognet M, Kahn A Glucose regulation of gene transcription. Koo SH, Dutcher AK, Towle HC Glucose and insulin function through two distinct transcription factors to stimulate expression of lipogenic enzyme genes in liver.

Mol Cell — Najjar SM, Yang Y, Fernstrom MA et al Insulin acutely decreases hepatic fatty acid synthase activity. Wolfrum C, Asilmaz E, Luca E, Friedman JM, Stoffel M Foxa2 regulates lipid metabolism and ketogenesis in the liver during fasting and in diabetes. Nature — Kalaany NY, Gauthier KC, Zavacki AM et al LXRs regulate the balance between fat storage and oxidation.

Gerin I, Dolinsky VW, Shackman JG et al LXRbeta is required for adipocyte growth, glucose homeostasis, and beta cell function. Repa JJ, Mangelsdorf DJ The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol — Tobin KA, Ulven SM, Schuster GU et al Liver X receptors as insulin-mediating factors in fatty acid and cholesterol biosynthesis.

Cao G, Liang Y, Broderick CL et al Antidiabetic action of a liver x receptor agonist mediated by inhibition of hepatic gluconeogenesis. Stulnig TM, Oppermann U, Steffensen KR, Schuster GU, Gustafsson JA Liver X receptors downregulate 11beta-hydroxysteroid dehydrogenase type 1 expression and activity.

Stulnig TM, Steffensen KR, Gao H et al Novel roles of liver X receptors exposed by gene expression profiling in liver and adipose tissue.

Mol Pharmacol — Glass CK Differential recognition of target genes by nuclear receptor monomers, dimers, and heterodimers.

Endocr Rev — Chen G, Liang G, Ou J, Goldstein JL, Brown MS Central role for liver X receptor in insulin-mediated activation of Srebp-1c transcription and stimulation of fatty acid synthesis in liver. Yoshikawa T, Ide T, Shimano H et al Cross-talk between peroxisome proliferator-activated receptor PPAR alpha and liver X receptor LXR in nutritional regulation of fatty acid metabolism.

PPARs suppress sterol regulatory element binding protein-1c promoter through inhibition of LXR signaling. Mol Endocrinol — Ide T, Shimano H, Yoshikawa T et al Cross-talk between peroxisome proliferator-activated receptor PPAR alpha and liver X receptor LXR in nutritional regulation of fatty acid metabolism.

LXRs suppress lipid degradation gene promoters through inhibition of PPAR signaling. Miyata KS, McCaw SE, Patel HV, Rachubinski RA, Capone JP The orphan nuclear hormone receptor LXR alpha interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling.

Anderson SP, Dunn C, Laughter A, et al Overlapping transcriptional programs regulated by the nuclear receptors peroxisome proliferator-activated receptor alpha, retinoid X receptor, and liver X receptor in mouse liver. Grefhorst A, Elzinga BM, Voshol PJ et al Stimulation of lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles.

Am J Physiol Endocrinol Metab E—E Miyazaki M, Ntambi JM Role of stearoyl-coenzyme A desaturase in lipid metabolism. Prostaglandins Leukot Essent Fatty Acids — Vessby B, Unsitupa M, Hermansen K et al Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy men and women: The KANWU Study.

Diabetologia — Hu T, Foxworthy P, Siesky A et al Hepatic peroxisomal fatty acid beta oxidation is regulated by liver X receptor alpha. Endocrinology — Gibbons G Old fat, make way for new fat. Nat Med — Lemberger T, Braissant O, Juge-Aubry C et al PPAR tissue distribution and interactions with other hormone-signaling pathways.

Ann N Y Acad Sci — Browning JD, Horton JD Molecular mediators of hepatic steatosis and liver injury. Lee CH, Chawla A, Urbiztondo N et al Transcriptional repression of atherogenic inflammation: modulation by PPARdelta. Tanaka T, Yamamoto J, Iwasaki S et al Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome.

Reddy JK, Hashimoto T Peroxisomal beta-oxidation and peroxisome proliferator-activated receptor alpha: an adaptive metabolic system. Vidal-Puig AJ, Considine RV, Jimenez-Linan M et al Peroxisome proliferator-activated receptor gene expression in human tissues.

Effects of obesity, weight loss, and regulation by insulin and glucocorticoids. Bernal-Mizrachi C, Weng S, Feng C et al Dexamethasone induction of hypertension and diabetes is PPAR-alpha dependent in LDL receptor-null mice.

Koo SH, Satoh H, Herzig S et al PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB Kersten S, Seydoux J, Peters JM, et al Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting. Linton MF, Fazio S Re-emergence of fibrates in the management of dyslipidemia and cardiovascular risk.

Curr Atheroscler Rep — Bays H, Stein EA Pharmacotherapy for dyslipidaemia—current therapies and future agents. Expert Opin Pharmacother — Hermanowski-Vosatka A, Gerhold D, Mundt SS et al PPARalpha agonists reduce 11beta-hydroxysteroid dehydrogenase type 1 in the liver.

Biochem Biophys Res Commun — Guerre-Millo M, Gervois P, Raspe E et al Peroxisome proliferator-activated receptor alpha activators improve insulin sensitivity and reduce adiposity. Staels B, Fruchart JC Therapeutic roles of peroxisome proliferator-activated receptor agonists.

Rao MS, Reddy JK Hepatocarcinogenesis of peroxisome proliferators. Knight B, Yeap BB, Yeoh GC, Olynyk JK Inhibition of adult liver progenitor oval cell growth and viability by an agonist of the peroxisome proliferator activated receptor PPAR family member gamma, but not alpha or delta.

Carcinogenesis — Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. Horton JD, Goldstein JL, Brown MS SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver.

Yamamoto T, Shimano H, Nakagawa Y et al SREBP-1 interacts with hepatocyte nuclear factor-4 alpha and interferes with PGC-1 recruitment to suppress hepatic gluconeogenic genes.

Chakravarty K, Cassuto H, Reshef L, Hanson RW Factors that control the tissue-specific transcription of the gene for phosphoenolpyruvate carboxykinase-C.

Crit Rev Biochem Mol Biol — Shimano H, Horton JD, Shimomura I et al Isoform 1c of sterol regulatory element binding protein is less active than isoform 1a in livers of transgenic mice and in cultured cells. Foretz M, Guichard C, Ferre P, Foufelle F Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes.

Ribaux PG, Iynedjian PB Analysis of the role of protein kinase B cAKT in insulin-dependent induction of glucokinase and sterol regulatory element-binding protein 1 SREBP1 mRNAs in hepatocytes. Mol Cell Biol — Zhou G, Myers R, Li Y et al Role of AMP-activated protein kinase in mechanism of metformin action.

Jump DB, Botolin D, Wang Y et al Fatty Acid regulation of hepatic gene transcription. J Nutr — Puigserver P, Rhee J, Donovan J et al Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction.

Barthel A, Schmoll D Novel concepts in insulin regulation of hepatic gluconeogenesis. Ide T, Shimano H, Yahagi N et al SREBPs suppress IRSmediated insulin signalling in the liver.

Nat Cell Biol — Hasty AH, Shimano H, Yahagi N et al Sterol regulatory element-binding protein-1 is regulated by glucose at the transcriptional level. Matsuzaka T, Shimano H, Yahagi N et al Insulin-independent induction of sterol regulatory element-binding protein-1c expression in the livers of streptozotocin-treated mice.

Pawar A, Botolin D, Mangelsdorf DJ, Jump DB The role of liver X receptor-alpha in the fatty acid regulation of hepatic gene expression. Stoeckman AK, Ma L, Towle HC Mlx is the functional heteromeric partner of the carbohydrate response element-binding protein in glucose regulation of lipogenic enzyme genes.

He Z, Jiang T, Wang Z, Levi M, Li J Modulation of carbohydrate response element-binding protein gene expression in 3T3-L1 adipocytes and rat adipose tissue. Promrat K, Lutchman G, Uwaifo GI et al A pilot study of pioglitazone treatment for nonalcoholic steatohepatitis.

Hepatology — Uyeda K, Yamashita H, Kawaguchi T Carbohydrate responsive element-binding protein ChREBP : a key regulator of glucose metabolism and fat storage. Biochem Pharmacol — Dentin R, Benhamed F, Pegorier JP et al Polyunsaturated fatty acids suppress glycolytic and lipogenic genes through the inhibition of ChREBP nuclear protein translocation.

Tomlinson JW, Walker EA, Bujalska IJ et al 11beta-hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Lancet — Tomlinson JW, Stewart MP Mechanisms of disease: selective inhibition of 11β-hydroxysteroid dehydrogenase type 1 as a novel treatment for the metabolic syndrome.

CAS Google Scholar. Friedman JE, Yun JS, Patel YM, McGrane MM, Hanson RW Glucocorticoids regulate the induction of phosphoenolpyruvate carboxykinase GTP gene transcription during diabetes.

Letteron P, Brahimi-Bourouina N, Robin MA et al Glucocorticoids inhibit mitochondrial matrix acyl-CoA dehydrogenases and fatty acid beta-oxidation. Am J Physiol G—G Lamberts SW, Koper JW, de Jong FH The endocrine effects of long-term treatment with mifepristone RU Harris HJ, Kotelevtsev Y, Mullins JJ, Seckl JR, Holmes MC Intracellular regeneration of glucocorticoids by 11beta-hydroxysteroid dehydrogenase 11beta-HSD -1 plays a key role in regulation of the hypothalamic—pituitary—adrenal axis: analysis of 11beta-HSDdeficient mice.

Alberts P, Engblom L, Edling N et al Selective inhibition of 11beta-hydroxysteroid dehydrogenase type 1 decreases blood glucose concentrations in hyperglycaemic mice. Kahn BB, Alquier T, Carling D, Hardie DG AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism.

Shaw RJ, Lamia KA, Vasquez D et al The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Hardie DG, Scott JW, Pan DA, Hudson ER Management of cellular energy by the AMP-activated protein kinase system.

FEBS Lett — Lochhead PA, Salt IP, Walker KS, Hardie DG, Sutherland C 5-aminoimidazolecarboxamide riboside mimics the effects of insulin on the expression of the 2 key gluconeogenic genes PEPCK and glucosephosphatase.

Combs TP, Berg AH, Obici S, Scherer PE, Rossetti L Endogenous glucose production is inhibited by the adipose-derived protein Acrp Yamauchi T, Kamon J, Minokoshi Y et al Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase.

Spranger J, Kroke A, Mohlig M et al Adiponectin and protection against type 2 diabetes mellitus. Kola B, Hubina E, Tucci SA et al Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. Murata M, Okimura Y, Iida K et al Ghrelin modulates the downstream molecules of insulin signaling in hepatoma cells.

Ruderman NB, Saha AK, Kraegen EW Minireview: malonyl CoA, AMP-activated protein kinase, and adiposity. Saha AK, Avilucea PR, Ye JM et al Pioglitazone treatment activates AMP-activated protein kinase in rat liver and adipose tissue in vivo. Kersten S, Desvergne B, Wahli W Roles of PPARs in health and disease.

Klein J, Westphal S, Kraus D et al Metformin inhibits leptin secretion via a mitogen-activated protein kinase signalling pathway in brown adipocytes. J Endocrinol — Musi N, Goodyear LJ Targeting the AMP-activated protein kinase for the treatment of type 2 diabetes.

Curr Drug Targets Immune Endocr Metabol Disord — Minokoshi Y, Alquier T, Furukawa N et al AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Carling D AMP-activated protein kinase: balancing the scales.

Lin MH, Lu SC, Huang PC, Liu YC, Liu SY A high-cholesterol, n -3 polyunsaturated fatty acid diet causes different responses in rats and hamsters. Ann Nutr Metab — Download references. Due to limited space we only could discuss some of the key pathways.

We apologise to all the researchers whose work was omitted because of constraints on the number of references. We would like to thank C. Loracher for intensive discussion of the subject. Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee , , Nuthetal, Germany.

Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité-University-Medicine Berlin, Berlin, Germany.

You can also search for this author in PubMed Google Scholar. Correspondence to M. Reprints and permissions. Weickert, M. Signalling mechanisms linking hepatic glucose and lipid metabolism.

Diabetologia 49 , — Download citation. Received : 04 November Accepted : 03 February Published : 23 May Issue Date : August 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. Download PDF. Abstract Fatty liver and hepatic triglyceride accumulation are strongly associated with obesity, insulin resistance and type 2 diabetes, and are subject to nutritional influences.

Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease Article Open access 26 June The mechanisms of action of metformin Article Open access 03 August Tcf7l2 in hepatocytes regulates de novo lipogenesis in diet-induced non-alcoholic fatty liver disease in mice Article Open access 10 February Use our pre-submission checklist Avoid common mistakes on your manuscript.

Introduction The exact mechanisms that link obesity, impaired glucose metabolism, hepatic lipid accumulation and insulin resistance are unknown, but our knowledge is rapidly increasing. Role of the liver in glucose metabolism In the liver, insulin regulates fasting glucose concentrations by inhibiting hepatic glucose production and stimulating glycogen synthesis.

Role of the liver in lipid metabolism Consistent with its function as an anabolic hormone, insulin promotes the synthesis, and inhibits the degradation of lipids. Acute vs prolonged exposure to insulin Insulin is commonly viewed as a positive regulator of fatty acid synthesis, as it promotes the expression of FASN and ACAC.

Full size image. Concluding remarks The studies discussed in this review indicate that hepatic fat accumulation, insulin resistance and disturbed glucose metabolism are inter-related at a molecular level.

Biochem Soc Trans — Article PubMed 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.

Biochem J — Article PubMed CAS Google Scholar Dentin R, Girard J, Postic C Carbohydrate responsive element binding protein ChREBP and sterol regulatory element binding protein-1c SREBP-1c : two key regulators of glucose metabolism and lipid synthesis in liver.

Biochimie —86 Article PubMed CAS Google Scholar Ishii S, Iizuka K, Miller BC, Uyeda K Carbohydrate response element binding protein directly promotes lipogenic enzyme gene transcription. Proc Natl Acad Sci USA — Article PubMed CAS Google Scholar Steffensen KR, Gustafsson JA Putative metabolic effects of the liver X receptor LXR.

Diabetes 53 Suppl 1 :S36—S42 PubMed CAS Google Scholar Marchesini G, Brizi M, Bianchi G et al Nonalcoholic fatty liver disease: a feature of the metabolic syndrome. Diabetes — PubMed CAS Google Scholar Angulo P Nonalcoholic fatty liver disease.

N Engl J Med — Article PubMed CAS Google Scholar Seppala-Lindroos A, Vehkavaara S, Hakkinen AM et al Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J Clin Endocrinol Metab — Article PubMed CAS Google Scholar Sutinen J, Hakkinen AM, Westerbacka J et al Rosiglitazone in the treatment of HAART-associated lipodystrophy—a randomized double-blind placebo-controlled study.

Antivir Ther — PubMed CAS Google Scholar Gavrilova O, Marcus-Samuels B, Graham D et al Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice.

J Biol Chem — Article PubMed CAS Google Scholar Samuel VT, Liu ZX, Qu X et al Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J Biol Chem — Article PubMed CAS Google Scholar Petersen KF, Dufour S, Befroy D et al Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes.

Diabetes — PubMed CAS Google Scholar Gastaldelli A, Toschi E, Pettiti M et al Effect of physiological hyperinsulinemia on gluconeogenesis in nondiabetic subjects and in type 2 diabetic patients.

Diabetes — PubMed CAS Google Scholar Boden G Effects of free fatty acids on gluconeogenesis and glycogenolysis. Life Sci — Article PubMed CAS Google Scholar Roden M, Bernroider E Hepatic glucose metabolism in humans—its role in health and disease. Best Pract Res Clin Endocrinol Metab — Article PubMed CAS Google Scholar Nordlie RC, Foster JD, Lange AJ Regulation of glucose production by the liver.

Annu Rev Nutr — Article PubMed CAS Google Scholar Pilkis SJ, Granner DK Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis. Diabetes — PubMed CAS Google Scholar Lam TK, Gutierrez-Juarez R, Pocai A, Rossetti L Regulation of blood glucose by hypothalamic pyruvate metabolism.

Science — Article PubMed CAS Google Scholar Pocai A, Obici S, Schwartz GJ, Rossetti L A brain—liver circuit regulates glucose homeostasis. Cell Metab —61 Article PubMed CAS Google Scholar Horton JD, Shah NA, Warrington JA et al Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes.

Proc Natl Acad Sci USA — Article PubMed CAS Google Scholar Randle PJ Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes Metab Rev — Article PubMed CAS Google Scholar Ruderman NB, Saha AK, Vavvas D, Witters LA Malonyl-CoA, fuel sensing, and insulin resistance.

Am J Physiol E1—E18 PubMed CAS Google Scholar McGarry JD Banting lecture dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes —18 PubMed CAS Google Scholar Vaulont S, Vasseur-Cognet M, Kahn A Glucose regulation of gene transcription.

J Biol Chem — Article PubMed CAS Google Scholar Koo SH, Dutcher AK, Towle HC Glucose and insulin function through two distinct transcription factors to stimulate expression of lipogenic enzyme genes in liver. Mol Cell —86 Article PubMed CAS Google Scholar Najjar SM, Yang Y, Fernstrom MA et al Insulin acutely decreases hepatic fatty acid synthase activity.

Cell Metab —53 Article PubMed CAS Google Scholar Wolfrum C, Asilmaz E, Luca E, Friedman JM, Stoffel M Foxa2 regulates lipid metabolism and ketogenesis in the liver during fasting and in diabetes.

Nature — Article PubMed CAS Google Scholar Kalaany NY, Gauthier KC, Zavacki AM et al LXRs regulate the balance between fat storage and oxidation.

Cell Metab — Article PubMed CAS Google Scholar Gerin I, Dolinsky VW, Shackman JG et al LXRbeta is required for adipocyte growth, glucose homeostasis, and beta cell function.

J Biol Chem — Article PubMed CAS Google Scholar Repa JJ, Mangelsdorf DJ The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol — Article PubMed CAS Google Scholar Tobin KA, Ulven SM, Schuster GU et al Liver X receptors as insulin-mediating factors in fatty acid and cholesterol biosynthesis.

J Biol Chem — PubMed CAS Google Scholar Cao G, Liang Y, Broderick CL et al Antidiabetic action of a liver x receptor agonist mediated by inhibition of hepatic gluconeogenesis. J Biol Chem — Article PubMed CAS Google Scholar Stulnig TM, Oppermann U, Steffensen KR, Schuster GU, Gustafsson JA Liver X receptors downregulate 11beta-hydroxysteroid dehydrogenase type 1 expression and activity.

Diabetes — PubMed CAS Google Scholar Stulnig TM, Steffensen KR, Gao H et al Novel roles of liver X receptors exposed by gene expression profiling in liver and adipose tissue. Mol Pharmacol — Article PubMed CAS Google Scholar Glass CK Differential recognition of target genes by nuclear receptor monomers, dimers, and heterodimers.

Endocr Rev — Article PubMed CAS Google Scholar Chen G, Liang G, Ou J, Goldstein JL, Brown MS Central role for liver X receptor in insulin-mediated activation of Srebp-1c transcription and stimulation of fatty acid synthesis in liver. Proc Natl Acad Sci USA — Article PubMed CAS Google Scholar Yoshikawa T, Ide T, Shimano H et al Cross-talk between peroxisome proliferator-activated receptor PPAR alpha and liver X receptor LXR in nutritional regulation of fatty acid metabolism.

Mol Endocrinol — PubMed CAS Google Scholar Ide T, Shimano H, Yoshikawa T et al Cross-talk between peroxisome proliferator-activated receptor PPAR alpha and liver X receptor LXR in nutritional regulation of fatty acid metabolism.

Mol Endocrinol — PubMed CAS Google Scholar Miyata KS, McCaw SE, Patel HV, Rachubinski RA, Capone JP The orphan nuclear hormone receptor LXR alpha interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling.

J Biol Chem — Article PubMed CAS Google Scholar Anderson SP, Dunn C, Laughter A, et al Overlapping transcriptional programs regulated by the nuclear receptors peroxisome proliferator-activated receptor alpha, retinoid X receptor, and liver X receptor in mouse liver.

Mol Pharmacol — Article PubMed CAS Google Scholar Grefhorst A, Elzinga BM, Voshol PJ et al Stimulation of lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles. Am J Physiol Endocrinol Metab E—E Article PubMed CAS Google Scholar Miyazaki M, Ntambi JM Role of stearoyl-coenzyme A desaturase in lipid metabolism.

Article PubMed CAS Google Scholar Vessby B, Unsitupa M, Hermansen K et al Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy men and women: The KANWU Study.

Diabetologia — Article PubMed CAS Google Scholar Hu T, Foxworthy P, Siesky A et al Hepatic peroxisomal fatty acid beta oxidation is regulated by liver X receptor alpha. Cell Metab — Article PubMed CAS Google Scholar Gibbons G Old fat, make way for new fat.

Nat Med — Article PubMed CAS Google Scholar Lemberger T, Braissant O, Juge-Aubry C et al PPAR tissue distribution and interactions with other hormone-signaling pathways. Ann N Y Acad Sci — PubMed CAS Google Scholar Browning JD, Horton JD Molecular mediators of hepatic steatosis and liver injury.

J Clin Invest — Article PubMed CAS Google Scholar Lee CH, Chawla A, Urbiztondo N et al Transcriptional repression of atherogenic inflammation: modulation by PPARdelta. Science — Article PubMed CAS Google Scholar Tanaka T, Yamamoto J, Iwasaki S et al Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome.

Proc Natl Acad Sci USA — Article PubMed CAS Google Scholar Reddy JK, Hashimoto T Peroxisomal beta-oxidation and peroxisome proliferator-activated receptor alpha: an adaptive metabolic system. Annu Rev Nutr — Article PubMed CAS Google Scholar Vidal-Puig AJ, Considine RV, Jimenez-Linan M et al Peroxisome proliferator-activated receptor gene expression in human tissues.

J Clin Invest — PubMed CAS Google Scholar Bernal-Mizrachi C, Weng S, Feng C et al Dexamethasone induction of hypertension and diabetes is PPAR-alpha dependent in LDL receptor-null mice. Nat Med — Article PubMed CAS Google Scholar Koo SH, Satoh H, Herzig S et al PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB Nat Med — Article PubMed CAS Google Scholar Kersten S, Seydoux J, Peters JM, et al Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting.

J Clin Invest — PubMed CAS Google Scholar Linton MF, Fazio S Re-emergence of fibrates in the management of dyslipidemia and cardiovascular risk. Curr Atheroscler Rep —35 PubMed CAS Google Scholar Bays H, Stein EA Pharmacotherapy for dyslipidaemia—current therapies and future agents.

Expert Opin Pharmacother — Article PubMed CAS Google Scholar Hermanowski-Vosatka A, Gerhold D, Mundt SS et al PPARalpha agonists reduce 11beta-hydroxysteroid dehydrogenase type 1 in the liver. Biochem Biophys Res Commun — Article PubMed CAS Google Scholar Guerre-Millo M, Gervois P, Raspe E et al Peroxisome proliferator-activated receptor alpha activators improve insulin sensitivity and reduce adiposity.

J Biol Chem — Article PubMed CAS Google Scholar Staels B, Fruchart JC Therapeutic roles of peroxisome proliferator-activated receptor agonists. Diabetes — PubMed CAS Google Scholar Rao MS, Reddy JK Hepatocarcinogenesis of peroxisome proliferators. Ann N Y Acad Sci — PubMed CAS Google Scholar Knight B, Yeap BB, Yeoh GC, Olynyk JK Inhibition of adult liver progenitor oval cell growth and viability by an agonist of the peroxisome proliferator activated receptor PPAR family member gamma, but not alpha or delta.

Carcinogenesis — Article PubMed CAS Google Scholar Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. J Clin Invest — Article PubMed CAS Google Scholar Horton JD, Goldstein JL, Brown MS SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver.

J Clin Invest — Article PubMed CAS Google Scholar Yamamoto T, Shimano H, Nakagawa Y et al SREBP-1 interacts with hepatocyte nuclear factor-4 alpha and interferes with PGC-1 recruitment to suppress hepatic gluconeogenic genes.

J Biol Chem — Article PubMed CAS Google Scholar Chakravarty K, Cassuto H, Reshef L, Hanson RW Factors that control the tissue-specific transcription of the gene for phosphoenolpyruvate carboxykinase-C.

Crit Rev Biochem Mol Biol — Article PubMed CAS Google Scholar Shimano H, Horton JD, Shimomura I et al Isoform 1c of sterol regulatory element binding protein is less active than isoform 1a in livers of transgenic mice and in cultured cells.

J Clin Invest — PubMed CAS Google Scholar Foretz M, Guichard C, Ferre P, Foufelle F Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes.

Proc Natl Acad Sci USA — PubMed CAS Google Scholar Ribaux PG, Iynedjian PB Analysis of the role of protein kinase B cAKT in insulin-dependent induction of glucokinase and sterol regulatory element-binding protein 1 SREBP1 mRNAs in hepatocytes.

Mol Cell Biol — PubMed CAS Google Scholar Zhou G, Myers R, Li Y et al Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest — Article PubMed CAS Google Scholar Jump DB, Botolin D, Wang Y et al Fatty Acid regulation of hepatic gene transcription.

J Nutr — PubMed CAS Google Scholar Puigserver P, Rhee J, Donovan J et al Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. Nature — Article PubMed CAS Google Scholar Barthel A, Schmoll D Novel concepts in insulin regulation of hepatic gluconeogenesis.

Am J Physiol Endocrinol Metab E—E PubMed CAS Google Scholar Ide T, Shimano H, Yahagi N et al SREBPs suppress IRSmediated insulin signalling in the liver. Nat Cell Biol — Article PubMed CAS Google Scholar Hasty AH, Shimano H, Yahagi N et al Sterol regulatory element-binding protein-1 is regulated by glucose at the transcriptional level.

J Biol Chem — Article PubMed CAS Google Scholar Matsuzaka T, Shimano H, Yahagi N et al Insulin-independent induction of sterol regulatory element-binding protein-1c expression in the livers of streptozotocin-treated mice.

J Nutr — PubMed CAS Google Scholar Pawar A, Botolin D, Mangelsdorf DJ, Jump DB The role of liver X receptor-alpha in the fatty acid regulation of hepatic gene expression. J Biol Chem — Article PubMed CAS Google Scholar Stoeckman AK, Ma L, Towle HC Mlx is the functional heteromeric partner of the carbohydrate response element-binding protein in glucose regulation of lipogenic enzyme genes.

J Biol Chem — Article PubMed CAS Google Scholar He Z, Jiang T, Wang Z, Levi M, Li J Modulation of carbohydrate response element-binding protein gene expression in 3T3-L1 adipocytes and rat adipose tissue.

Am J Physiol Endocrinol Metab E—E Article PubMed CAS Google Scholar Promrat K, Lutchman G, Uwaifo GI et al A pilot study of pioglitazone treatment for nonalcoholic steatohepatitis.

Hepatology — Article PubMed CAS Google Scholar Uyeda K, Yamashita H, Kawaguchi T Carbohydrate responsive element-binding protein ChREBP : a key regulator of glucose metabolism and fat storage. Biochem Pharmacol — Article PubMed CAS Google Scholar Dentin R, Benhamed F, Pegorier JP et al Polyunsaturated fatty acids suppress glycolytic and lipogenic genes through the inhibition of ChREBP nuclear protein translocation.

J Clin Invest — Article PubMed CAS Google Scholar Tomlinson JW, Walker EA, Bujalska IJ et al 11beta-hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Lancet — Article PubMed CAS Google Scholar Tomlinson JW, Stewart MP Mechanisms of disease: selective inhibition of 11β-hydroxysteroid dehydrogenase type 1 as a novel treatment for the metabolic syndrome.

J Biol Chem — PubMed CAS Google Scholar Letteron P, Brahimi-Bourouina N, Robin MA et al Glucocorticoids inhibit mitochondrial matrix acyl-CoA dehydrogenases and fatty acid beta-oxidation. Mol Endocrinol — Article PubMed CAS Google Scholar Lamberts SW, Koper JW, de Jong FH The endocrine effects of long-term treatment with mifepristone RU J Clin Endocrinol Metab — Article PubMed CAS Google Scholar Harris HJ, Kotelevtsev Y, Mullins JJ, Seckl JR, Holmes MC Intracellular regeneration of glucocorticoids by 11beta-hydroxysteroid dehydrogenase 11beta-HSD -1 plays a key role in regulation of the hypothalamic—pituitary—adrenal axis: analysis of 11beta-HSDdeficient mice.

Endocrinology — Article PubMed CAS Google Scholar Alberts P, Engblom L, Edling N et al Selective inhibition of 11beta-hydroxysteroid dehydrogenase type 1 decreases blood glucose concentrations in hyperglycaemic mice.

Diabetologia — Article PubMed CAS Google Scholar Kahn BB, Alquier T, Carling D, Hardie DG AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism.

Cell Metab —25 Article PubMed CAS Google Scholar Shaw RJ, Lamia KA, Vasquez D et al The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin.

Science — Article PubMed CAS Google Scholar Hardie DG, Scott JW, Pan DA, Hudson ER Management of cellular energy by the AMP-activated protein kinase system.

J Biol Chem — Article PubMed CAS Google Scholar Lochhead PA, Salt IP, Walker KS, Hardie DG, Sutherland C 5-aminoimidazolecarboxamide riboside mimics the effects of insulin on the expression of the 2 key gluconeogenic genes PEPCK and glucosephosphatase.

Diabetes — PubMed CAS Google Scholar Combs TP, Berg AH, Obici S, Scherer PE, Rossetti L Endogenous glucose production is inhibited by the adipose-derived protein Acrp J Clin Invest — Article PubMed CAS Google Scholar Yamauchi T, Kamon J, Minokoshi Y et al Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase.

Nat Med — PubMed CAS Google Scholar Spranger J, Kroke A, Mohlig M et al Adiponectin and protection against type 2 diabetes mellitus. Lancet — Article PubMed CAS Google Scholar Kola B, Hubina E, Tucci SA et al Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase.

J Biol Chem — Article PubMed CAS Google Scholar Murata M, Okimura Y, Iida K et al Ghrelin modulates the downstream molecules of insulin signaling in hepatoma cells. J Biol Chem — Article PubMed CAS Google Scholar Ruderman NB, Saha AK, Kraegen EW Minireview: malonyl CoA, AMP-activated protein kinase, and adiposity.

Endocrinology — Article PubMed CAS Google Scholar Saha AK, Avilucea PR, Ye JM et al Pioglitazone treatment activates AMP-activated protein kinase in rat liver and adipose tissue in vivo. Biochem Biophys Res Commun — Article PubMed CAS Google Scholar Kersten S, Desvergne B, Wahli W Roles of PPARs in health and disease.

Nature — Article PubMed CAS Google Scholar Klein J, Westphal S, Kraus D et al Metformin inhibits leptin secretion via a mitogen-activated protein kinase signalling pathway in brown adipocytes. J Endocrinol — Article PubMed CAS Google Scholar Musi N, Goodyear LJ Targeting the AMP-activated protein kinase for the treatment of type 2 diabetes.

Curr Drug Targets Immune Endocr Metabol Disord — Article PubMed CAS Google Scholar Minokoshi Y, Alquier T, Furukawa N et al AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature — Article PubMed CAS Google Scholar Carling D AMP-activated protein kinase: balancing the scales.

Biochimie —91 Article PubMed CAS Google Scholar Lin MH, Lu SC, Huang PC, Liu YC, Liu SY A high-cholesterol, n -3 polyunsaturated fatty acid diet causes different responses in rats and hamsters.

Ann Nutr Metab — Article PubMed CAS Google Scholar Download references. Acknowledgements Due to limited space we only could discuss some of the key pathways. Duality of interest statement The authors are not aware of any duality of interest.

Author information Authors and Affiliations Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee , , Nuthetal, Germany M.

Pfeiffer Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité-University-Medicine Berlin, Berlin, Germany M.

Effect of lipid oxidation on glucose utilization in humans

As fat accumulates, there is an ongoing increase in the levels of plasma FFAs, which causes insulin resistance. In addition, the deficit of another product of adipose tissue e.

To counter insulin resistance and prevent hyperglycemia, insulin levels increase. In individuals with a genetic predisposition for diabetes, however, the pancreas cannot compensate for the increased secretory demands placed on it, resulting in type 2 diabetes.

The pivotal role of FFAs in the development of insulin resistance and type 2 diabetes suggests that the optimal therapeutic intervention should decrease plasma FFA levels. The PPAR family is intimately involved in lipid metabolism.

Two subtypes of these receptors are the site of action of synthetic PPAR agonists: PPAR-α and PPAR-γ. The former increases fatty acid oxidation, whereas the latter results in the redistribution of fat from visceral to subcutaneous body fat and an increase in adiponectin.

The outcome of activation of PPAR-γ is a lowering of plasma FFA concentrations and improved insulin sensitivity.

The effects of PPAR-α on lipid metabolism may also bring about improvements in insulin sensitivity. The currently available PPAR agonists selectively activate either PPAR-α i. Potential mechanism of FFA on insulin resistance and atherogenesis in human muscle. The key initiating event is an increase in plasma FFA followed by increased uptake of FFA into muscle.

This leads to intramyocellular accumulation of fatty acyl-CoA and DAG and activation of PKC the β II and δ isoforms. It is assumed that activation of PKC interrupts insulin signaling by serine phosphorylation of IRS-1, resulting in a decrease in tyrosine phosphorylation of IRS PI, phosphatidylinositol.

This work was supported by National Institutes of Health Grants RAG, RDK, RHL, and RDK and a Mentor-Based Training Grant from the American Diabetes Association all to G.

A table elsewhere in this issue shows conventional and Système International SI units and conversion factors for many substances. Sign In or Create an Account. Search Dropdown Menu. header search search input Search input auto suggest.

filter your search All Content All Journals Diabetes Care. Advanced Search. User Tools Dropdown. Sign In. Skip Nav Destination Close navigation menu Article navigation. Volume 27, Issue 9. Previous Article Next Article.

FFAs AND INSULIN SECRETION. Article Information. Article Navigation. Lipids and Glucose in Type 2 Diabetes : What is the cause and effect? Guenther Boden, MD ; Guenther Boden, MD.

This Site. Google Scholar. Markku Laakso, MD Markku Laakso, MD. Address correspondencereprint requests to G. Boden, MD, Temple University Hospital, N.

Broad St. E-mail: bodengh tuhs. Diabetes Care ;27 9 — Article history Received:. Get Permissions. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. Figure 1—. View large Download slide. Figure 2—. Potential contributions of PPAR-α and PPAR-γ to improvements in insulin sensitivity.

McGarry JD: What if Minkowski had been ageusic? An alternative angle on diabetes. Jeppesen J, Hein HO, Suadicani P, Gyntelberg F: Relation of high TG-low HDL cholesterol and LDL cholesterol to the incidence of ischemic heart disease: an 8-year follow-up in the Copenhagen Male Study.

Arterioscler Thromb Vasc Biol. Gardner CD, Fortmann SP, Krauss RM: Association of small low-density lipoprotein particles with the incidence of coronary artery disease in men and women. Lamarche B, Tchernof A, Moorjani S, Cantin B, Dagenais GR, Lupien PJ, Despres JP: Small, dense low-density lipoprotein particles as a predictor of the risk of ischemic heart disease in men: prospective results from the Quebec Cardiovascular Study.

Mykkanen L, Kuusisto J, Haffner SM, Laakso M, Austin MA: LDL size and risk of coronary heart disease in elderly men and women. Hales CN, Barker DJ: Type 2 non-insulin-dependent diabetes mellitus: the thrifty phenotype hypothesis.

Arner P: Free fatty acids: do they play a central role in type 2 diabetes? Diabetes Obes Metab. Boden G, Jadali F, White J, Liang Y, Mozzoli M, Chen X, Coleman E, Smith C: Effects of fat on insulin-stimulated carbohydrate metabolism in normal men. J Clin Invest. Randle PJ, Garland PB, Hales CN, Newsholme EA: The glucose fatty-acid cycle: its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus.

Rennie MJ, Holloszy JO: Inhibition of glucose uptake and glycogenolysis by availability of oleate in well-oxygenated perfused skeletal muscle. Biochem J. Boden G, Chen X, Ruiz J, White JV, Rossetti L: Mechanisms of fatty acid-induced inhibition of glucose uptake.

Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW, Shulman GI: Mechanism of free fatty acid-induced insulin resistance in humans. Boden G, Chen X: Effects of fat on glucose uptake and utilization in patients with non-insulin-dependent diabetes. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, Slezak LA, Andersen DK, Hundal RS, Rothman DL, Petersen KF, Shulman GI: Effects of free fatty acids on glucose transport and IRSassociated phosphatidylinositol 3-kinase activity.

Homko CJ, Cheung P, Boden G: Effects of free fatty acids on glucose uptake and utilization in healthy women. Boden G, Lebed B, Schatz M, Homko C, Lemieux S: Effects of acute changes of plasma free fatty acids on intramyocellular fat content and insulin resistance in healthy subjects.

Itani SI, Ruderman NB, Schmieder F, Boden G: Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IκB-α. Bronfman M, Morales MN, Orellana A: Diacylglycerol activation of protein kinase C is modulated by long-chain acyl-CoA.

Biochem Biophys Res Commun. Itani SI, Zhou Q, Pories WJ, MacDonald KG, Dohm GL: Involvement of protein kinase C in human skeletal muscle insulin resistance and obesity. Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, Bergeron R, Kim JK, Cushman SW, Cooney GJ, Atcheson B, White MF, Kraegen EW, Shulman GI: Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 IRS-1 -associated phosphatidylinositol 3-kinase activity in muscle.

J Biol Chem. De Fea K, Roth RA: Protein kinase C modulation of insulin receptor substrate-1 tyrosine phosphorylation requires serine Ravichandran LV, Esposito DL, Chen J, Quon MJ: Protein kinase C-zeta phosphorylates insulin receptor substrate-1 and impairs its ability to activate phosphatidylinositol 3-kinase in response to insulin.

Kim JK, Kim YJ, Fillmore JJ, Chen Y, Moore I, Lee J, Yuan M, Li ZW, Karin M, Perret P, Shoelson SE, Shulman GI: Prevention of fat-induced insulin resistance by salicylate. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE: Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta.

Ross R: Atherosclerosis: an inflammatory disease. N Engl J Med. Ceriello A: Oxidative stress and glycemic regulation. Griffin ME, Marcucci MJ, Cline GW, Bell K, Barucci N, Lee D, Goodyear LJ, Kraegen EW, White MF, Shulman GI: Free fatty acid-induced insulin resistance is associated with activation of protein kinase C θ and alterations in the insulin signaling cascade.

Ferrannini E, Barrett EJ, Bevilacqua S, DeFronzo RA: Effect of fatty acids on glucose production and utilization in man. Gastaldelli A, Toschi E, Pettiti M, Frascerra S, Quinones-Galvan A, Sironi AM, Natali A, Ferrannini E: Effect of physiological hyperinsulinemia on gluconeogenesis in nondiabetic subjects and in type 2 diabetic patients.

Boden G, Cheung P, Stein TP, Kresge K, Mozzoli M: FFA cause hepatic insulin resistance by inhibiting insulin suppression of glycogenolysis. Am J Physiol Endocrinol Metab. Peraldi P, Spiegelman B: TNF-alpha and insulin resistance: summary and future prospects.

Mol Cell Biochem. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM: C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA: The hormone resistin links obesity to diabetes.

Kahn BB, Flier JS: Obesity and insulin resistance. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA: Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia.

J Clin Endocrinol Metab. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T: The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity.

Nat Med. Hotta K, Funahashi T, Bodkin NL, Ortmeyer HK, Arita Y, Hansen BC, Matsuzawa Y: Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Goldfine AB, Kahn RC: Apidonectin: linking the fat cell to insulin sensitivity.

Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y: Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity.

Halleux CM, Takahashi M, Delporte ML, Detry R, Funahashi T, Matsuzawa Y, Brichard SM: Secretion of adiponectin and regulation of apM1 gene expression in human visceral adipose tissue. Benjamin SM, Valdez R, Geiss LS, Rolka DB, Narayan KM: Estimated number of adults with prediabetes in the US in opportunities for prevention.

Diabetes Care. Crespin SR, Greenough WB III, Steinberg D: Stimulation of insulin secretion by long-chain free fatty acids: a direct pancreatic effect.

Boden G, Chen X, Rosner J, Barton M: Effects of a h fat infusion on insulin secretion and glucose utilization. Jensen CB, Storgaard H, Holst JJ, Dela F, Madsbad S, Vaag AA: Insulin secretion and cellular glucose metabolism after prolonged low-grade intralipid infusion in young men.

Kashyap S, Belfort R, Gastaldelli A, Pratipanawatr T, Berria R, Pratipanawatr W, Bajaj M, Mandarino L, DeFronzo R, Cusi K: A sustained increase in plasma free fatty acids impairs insulin secretion in nondiabetic subjects genetically predisposed to develop type 2 diabetes.

Boden G, Chen X, Iqbal N: Acute lowering of plasma fatty acids lowers basal insulin secretion in diabetic and nondiabetic subjects. Storgaard H, Jensen CB, Vaag AA, Volund A, Madsbad S: Insulin secretion after short- and long-term low-grade free fatty acid infusion in men with increased risk of developing type 2 diabetes.

Stefan N, Stumvoll M, Bogardus C, Tataranni PA: Elevated plasma nonesterified fatty acids are associated with deterioration of acute insulin response in IGT but not NGT. Pimenta W, Korytkowski M, Mitrakou A, Jenssen T, Yki-Jarvinen H, Evron W, Dailey G, Gerich J: Pancreatic beta-cell dysfunction as the primary genetic lesion in NIDDM: evidence from studies in normal glucose-tolerant individuals with a first-degree NIDDM relative.

Hirose H, Lee YH, Inman LR, Nagasawa Y, Johnson JH, Unger RH: Defective fatty acid-mediated beta-cell compensation in Zucker diabetic fatty rats: pathogenic implications for obesity-dependent diabetes. Chen X, Iqbal N, Boden G: The effects of free fatty acids on gluconeogenesis and glycogenolysis in normal subjects.

Ghazzi MN, Perez JE, Antonucci TK, Driscoll JH, Huang SM, Faja BW, Whitcomb RW: Cardiac and glycemic benefits of troglitazone treatment in NIDDM: the Troglitazone Study Group. Maggs DG, Buchanan TA, Burant CF, Cline G, Gumbiner B, Hsueh WA, Inzucchi S, Kelley D, Nolan J, Olefsky JM, Polonsky KS, Silver D, Valiquett TR, Shulman GI: Metabolic effects of troglitazone monotherapy in type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled trial.

Ann Intern Med. Mayerson AB, Hundal RS, Dufour S, Lebon V, Befroy D, Cline GW, Enocksson S, Inzucchi SE, Shulman GI, Petersen KF: The effects of rosiglitazone on insulin sensitivity, lipolysis, and hepatic and skeletal muscle triglyceride content in patients with type 2 diabetes.

Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA: An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma PPAR gamma.

Willson TM, Cobb JE, Cowan DJ, Wiethe RW, Correa ID, Prakash SR, Beck KD, Moore LB, Kliewer SA, Lehmann JM: The structure-activity relationship between peroxisome proliferator-activated receptor gamma agonism and the antihyperglycemic activity of thiazolidinediones.

J Med Chem. Tontonoz P, Hu E, Graves RA, Budavari AI, Spiegelman BM: mPPAR gamma 2: tissue-specific regulator of an adipocyte enhancer. Genes Dev. Kruszynska YT, Mukherjee R, Jow L, Dana S, Paterniti JR, Olefsky JM: Skeletal muscle peroxisome proliferator-activated receptor-gamma expression in obesity and non-insulin-dependent diabetes mellitus.

Tontonoz P, Hu E, Spiegelman BM: Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. de Souza CJ, Eckhardt M, Gagen K, Dong M, Chen W, Laurent D, Burkey BF: Effects of pioglitazone on adipose tissue remodeling within the setting of obesity and insulin resistance.

Maeda N, Takahashi M, Funahashi T, Kihara S, Nishizawa H, Kishida K, Nagaretani H, Matsuda M, Komuro R, Ouchi N, Kuriyama H, Hotta K, Nakamura T, Shimomura I, Matsuzawa Y: PPAR-γ ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein.

Boden G, Cheung P, Mozzoli M, Fried SK: Effect of thiazolidinediones on glucose and fatty acid metabolism in patients with type 2 diabetes. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R: Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association: October 7, Raskin P, Rendell M, Riddle MC, Dole JF, Freed MI, Rosenstock J: A randomized trial of rosiglitazone therapy in patients with inadequately controlled insulin-treated type 2 diabetes.

Cheng AY, Fantus IG: Thiazolidinedione-induced congestive heart failure. Ann Pharmacother. Kermani A, Garg A: Thiazolidinedione-associated congestive heart failure and pulmonary edema.

Mayo Clin Proc. Rubin C, Egan J, Schneider R: Combination therapy with pioglitazone and insulin in patients with type 2 diabetes Abstract. Rosenstock J, Einhorn D, Hershon K, Glazer NB, Yu S: Efficacy and safety of pioglitazone in type 2 diabetes: a randomised, placebo-controlled study in patients receiving stable insulin therapy.

Int J Clin Pract. Aronoff S, Rosenblatt S, Braithwaite S, Egan JW, Mathisen AL, Schneider RL: Pioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: a 6-month randomized placebo-controlled dose-response study: the Pioglitazone Study Group.

Phillips LS, Grunberger G, Miller E, Patwardhan R, Rappaport EB, Salzman A: Once- and twice-daily dosing with rosiglitazone improves glycemic control in patients with type 2 diabetes. Gulick T, Cresci S, Caira T, Moore DD, Kelly DP: The peroxisome proliferator-activated receptor regulates mitochondrial fatty acid oxidative enzyme gene expression.

Proc Natl Acad Sci U S A. Fatty acid ω-oxidation occurs in the endoplasmic reticulum of some cells, especially in the liver and kidney This process involves the monooxygenase-catalyzed oxidation of the fatty acid ω-carbon the carbon furthest from the carboxyl group , followed by successive oxidations of the β-carbon until the fatty acid chain is shortened by two carbon atoms.

This mechanism exists to break down large, water-insoluble fatty acids that, in greater quantities, would be hazardous to cells The ω-oxidation of fatty acids accelerates the rate of fatty acid degradation as feedback to various stressors such as hypoxia, inflammation, and stimulation by exogenous substances.

These substances increase the demand for FAO and detoxification. Correspondingly, the omega-oxidation of fatty acids reduces the likelihood that these stressors will lead to senescence and age-related diseases.

Triglycerides, phospholipids, sphingolipids, and cholesterol lipids are all produced by a series of enzymatic reactions between fatty acids and different chemical groups. Therefore, the regulation of fatty acid synthesis affects the metabolic homeostasis of lipids to a certain extent.

Acetyl-CoA carboxylase ACC catalyzes the rate-limiting step of de novo fatty acid synthesis Among the two subtypes, ACC1 and ACC2, the former locates in the cytoplasm and is used for fatty acid synthesis by converting acetyl coenzyme A into malonyl coenzyme A.

ACC2 is located on the cytoplasmic surface of mitochondria, and the product is used to inhibit CPT1 from reducing FAO SiRNA silencing of SREBP1 significantly reduced the expression of its downstream target genes ACC , FAS , and ATP citrate lyase, and weakened H 2 O 2 -induced senescence ACC1-dependent lipogenesis is the fundamental metabolic pathway downstream of AMPK, which induces autophagy and maintains cell survival during yeast senescence This evidence suggests that ACC directly modulates the senescence phenotype, and several drugs targeting ACC to slow senescence already exist.

Citrate treatment can increase ACC1 expression, promote excessive lipid biosynthesis, and lead to tumor cell senescence and growth inhibition The AD candidate drugs CMS and J play anti-senescence neuroprotective roles by inhibiting ACC1 and increasing acetyl-CoA levels Resveratrol plays an anti-senescence role by increasing ACC phosphorylation and enhancing mitochondrial lipolysis ability AMPK, the upstream negative regulator of ACC, also shows some anti-senescence activity.

Lipid metabolism depends heavily on AMPK. AMPK enhances lipolysis by upregulating ATGL expression AMPK can also suppress lipid production by inhibiting ACC activation by phosphorylating ACC1 Ser 79 Ala and ACC2 Ser Ala 92 and downregulating SREBP1c expression 93 , AMPK affects organism senescence directly or indirectly in a variety of ways.

For example, dietary restrictions delay the senescence of many species, in which AMPK is widely involved. elegans , AMPK and dietary restriction increase FAO through mitochondrial-peroxisome coordination, thereby maintaining homeostasis and plasticity within the mitochondrial network to extend life Greer et al.

elegans was mediated through the AMPK-FoxO transcription factors pathway. AMPKα subunit AAK-2 is activated by either a mutation that reduces insulin-like signaling or an environmental stressor that increases the AMP:ATP ratio, which can extend lifespan in C.

elegans 97 , A similar situation exists in mice. For instance, the pharmacological stimulation of AMPK can imitate caloric restriction induced comparable gene expression patterns and provide extensive protection against age-related diseases Fibroblast growth factor 21 can exhibit anti-senescence effects by elevating the expression of AMPK, which regulates lipid and glucose metabolic balance , blocking the p53 signaling pathway in an AMPK-dependent manner As an AMPK agonist, metformin plays a dual role in cancer prevention and anti-senescence , Inhibiting mTOR signaling plays a crucial role in mammalian senescence by reducing the accumulation of protein toxicity and oxidative stress 99 , increasing autophagy to clear damaged proteins and organelles , and enhancing the self-renewal capacity of hematopoietic stem cells and intestinal stem cells Ketone bodies are endogenous metabolites produced by the liver from fatty acids and ketogenic amino acids when glucose availability is low, such as during fasting, caloric restriction, or prolonged exercise Ketone bodies can be used as an alternative fuel source by many tissues, especially the brain, heart, and skeletal muscle, when glucose is scarce or absent These bodies can also cross the blood—brain barrier and supply energy to the central nervous system.

Ketone bodies are scary to clinicians because of the high mortality rate of ketoacidosis; however, in reality, the ketone bodies have signaling functions that regulate inflammation, epigenetics, oxidative stress, and other cellular processes by binding to specific receptors and enzymes — The bodies may also modulate gene expression and protein synthesis.

Ketone bodies are linked to multiple mechanisms of senescence and resilience, such as glucose sparing, mitochondrial biogenesis, autophagy, and hormesis — ; thus, they have anti-cancer, anti-angiogenic, and anti-atherogenic effects. Ketone bodies also reduce neuroinflammation and β-amyloid and tau accumulation, and improve memory and healthy lifespan in a senescent mouse model The most well-known ketone body, 3-hydroxybutyrate 3-OHB , binds to specific hydroxycarboxylic acid receptors to inhibit histone deacetylases, free fatty acid receptors, and nucleotide oligomerization domain NOD -like receptor protein 3 inflammasomes.

This initially inhibits lipolysis, inflammation, oxidative stress, cancer growth, angiogenesis, and atherosclerosis, and may increase lifespan associated with exercise and caloric restriction Additionally, 3-OHB protects muscle proteins from damage caused by systemic inflammation and is a crucial part of the metabolic defense against insulin-induced hypoglycemia Phospholipids are major components of biological membranes and consist of a glycerol backbone, two fatty acid chains, and a polar head group.

Phospholipids are essential for membrane structure, fluidity, and function, as well as for intracellular signaling, trafficking, and metabolism Phospholipids are synthesized in different cellular compartments by various enzymes and transported by specific carriers or vesicles.

Phospholipids mainly include phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, and cardiolipin.

Phospholipids can also be degraded or modified by phospholipases, acyltransferases, and other enzymes that regulate their turnover and diversity Phospholipids play important roles in senescence and age-related diseases. Senescence is associated with changes in phospholipid composition, metabolism, and transport in different tissues and organs.

These changes may affect membrane integrity, fluidity, and function, as well as cellular signaling and homeostasis , For example, senescence leads to decreased unsaturated fatty acid levels and increased saturated fatty acid levels in phospholipids, which may impair membrane fluidity and increase oxidative stress Senescence also alters the levels of specific phospholipid species, such as phosphatidylcholine, phosphatidylethanolamine, and cardiolipin, which may affect mitochondrial function and biogenesis Phospholipid interventions may have beneficial effects on senescence and lifespan.

Dietary supplementation or genetic manipulation of phospholipids or their precursors can modulate membrane properties, cellular signaling, and mitochondrial function in various model organisms.

For instance, phosphatidylcholine or choline supplementation improved cognitive function and memory in aged rodents Overexpression of cardiolipin synthase or supplementation of cardiolipin precursors can enhance mitochondrial function and extend lifespan in yeast, worms, flies, and mice Sphingolipids are a diverse class of lipids that are involved in various cellular processes such as membrane structure, signal transduction, cell cycle regulation, apoptosis, senescence, and inflammation Sphingolipids are synthesized de novo in the endoplasmic reticulum from non-sphingolipid precursors, such as serine and palmitoyl-CoA, and then further modified in the Golgi apparatus and other organelles to generate a variety of complex sphingolipids with different polar head groups The major bioactive sphingolipids include ceramide Cer , sphingosine, sphingosinephosphate S1P , and ceramidephosphate, which can act as second messengers or ligands for specific receptors to modulate cellular responses Sphingolipids have been implicated in the regulation of senescence and age-related diseases, as they can affect several hallmarks of senescence such as genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication In general, Cer and its derivatives induce cellular senescence and promote senescence phenotypes, while S1P and its receptor signaling delay senescence and extend lifespan For example, sphingolipids are involved in sarcopenia, an age-related disorder of loss of skeletal muscle mass and function.

Park et al. In addition, SPT deletion or myriocin treatment enhances mitochondrial function, autophagy, and proteostasis in aged muscle cells, suggesting that sphingolipids may impair these processes by affecting endoplasmic reticulum stress and calcium homeostasis.

Sterols are essential components of eukaryotic cell membranes, where they regulate membrane fluidity, permeability, and microdomain formation. Sterols also have important roles in animal physiology, as they are precursors of steroid hormones such as glucocorticoids, mineralocorticoids, androgens, estrogens, and progestins, which modulate various metabolic, reproductive, and immune functions Sterols are synthesized from acetyl-CoA through a complex pathway that involves more than 20 enzymes and intermediates The first steps of sterol biosynthesis occur in the cytosol and endoplasmic reticulum, where acetyl-CoA is converted to mevalonate by three enzymes: acetoacetyl-CoA thiolase, HMG-CoA synthase, and HMG-CoA reductase.

Mevalonate is then converted to isopentenyl pyrophosphate IPP and dimethylallyl pyrophosphate DMAPP , which are the building blocks of isoprenoids.

IPP and DMAPP are used to synthesize geranyl pyrophosphate, farnesyl pyrophosphate, and squalene, which are the precursors of sterols and other isoprenoids such as ubiquinone, dolichol, and prenylated proteins The final steps of sterol biosynthesis involve the cyclization of squalene to form lanosterol, which is then converted to cholesterol by a series of modifications such as hydroxylation, demethylation, reduction, and isomerization.

Take cholesterol, the predominant sterol in animals, for example, which is integral and indispensable to neuronal physiology during both development and adulthood.

As a major component of cell membranes and a precursor to steroid hormones, it helps regulate ion permeability, cell shape, intercellular interactions, and transmembrane signaling. Inherited diseases with mutations in cholesterol-related genes lead to impaired brain function. In these cases, brain cholesterol defects may be secondary to pathogenic factors and lead to functional deficits through altered synaptic function.

The Warburg effect reveals that tumor cells prefer ATP production by glycolysis over oxidative phosphorylation even in the presence of abundant oxygen The Warburg effect plays an important role in the inhibition of cellular senescence and tumor promotion Glycolysis is upregulated in most senescence phenotypes.

Glycolytic gene overexpression promotes altered cancer metabolism in a variety of tumor cells. LDHA, involved in the conversion of pyruvate to lactate, was significantly increased in radiotherapy-induced senescent HCT and MDA-MB cells , Lactate liberation acidulates the extracellular environment involved in SRSPs Lactic acid-induced acidic intracellular pH activates Snail to promote epithelial-mesenchymal transition.

Increased Snail expression facilitated lung cancer cells to escape oncogene-induced senescence by directly suppressing p16 INK4a expression In this way, LDHA expression in senescent fibroblasts promotes PC3 cell invasion in the co-culture of senescent fibroblasts and PC3 cells Pharmacological inhibition of LDHA induces tumor cell senescence by suppressing heat shock response Beyond radiotherapy- and chemotherapy-induced senescent cancer cells, glycolysis is also activated in non-neoplastic senescent cells.

The levels of intermediate metabolites of glycolysis, including 3-phosphoglycerate, glucose 6-phosphate, fructose 6-phosphate, and phosphoenolpyruvate, are significantly increased in senescent fibroblasts, which highlights increased glycolytic flux to promote pyruvate and lactate production Senescent spermatogonial stem cells exhibit JNK phosphorylation and enhanced glycolytic capacity compared with young cells Senescence fibroblasts exhibit a significant increase in glucose consumption Increased glycolysis appears to be mediated by the overexpression of multiple glycolytic enzymes in distinct kinds of induced senescence.

First, GLUT proteins are encoded by SLC2. GLUTs 1—4 have widely established roles as glucose transporters GLUT1 is overexpressed in aged lungs and regulates fibrogenesis In this way, hyperglycemia-induced GLUT1 overexpression activates the mTOR pathway to increase P16 and P21 levels while promoting macrophage establishment of the SRSP response Second, HKs catalyze the phosphorylation of glucose as the first rate-limiting step in glycolysis HK1 and HK2 isozymes are highly expressed in replicative senescence human fibroblasts Third, PK, the rate-limiting enzyme in the final step of glycolysis, catalyzes pyruvate production.

The levels of this enzyme are upregulated in replicative senescence fibroblasts due to increased TCA activity and oxygen consumption In conclusion, glycolysis is increased in replicative senescent cells, but the regulatory mechanism of elevated enzyme expression and activity needs to be further characterized.

Tumor cells maintain cell proliferation by activating glycolysis to provide energy and precursors. Pyruvate, a glycolytic terminal product, enters the TCA cycle to produce metabolic intermediates that inhibit cellular senescence Citrate is the product catalyzed by citrate synthase in the initiation of the TCA cycle.

Recently, citrate has been reported to extend lifespan through mTOR and AMPK. However, citrate treatment results in cellular senescence and inhibits the proliferation of MCF-7 and HCT cancer cells Mechanistically, DNA damage pathways coupled with MAPK and mTOR pathways lead to citrate-induced cellular senescence αKG, a metabolite catalyzed by isocitrate dehydrogenase IDH , extends the lifespan of C.

elegans 23 , Drosophila , and mice 22 ; attenuates mouse age-related bone loss 24 ; and delays age-related fertility decline in mammals Sustaining this biological activity requires a continuous supply of ATP from mitochondria; however, the partial inhibition of the electron transport chain by αKG reportedly extends the lifespan of C.

elegans and mammalian cells Mechanistically, αKG inhibits intracellular senescence by regulating the expression of histone epigenetic modifications of BMP signaling proteins In addition, αKG prolongs Drosophila lifespan by activating AMPK and inhibiting mTOR The IDH1 RH mutant indirectly promotes senescence of malignant glioma cells by reducing αKG production Abnormal glucose metabolism in cancer cells activates arrested cellular senescence.

Cancer cells not only show active glycolysis for sufficient energy but also precursors from the pentose phosphate pathway PPP for intracellular biosynthesis G6PD-mediated oxidative PPP oxPPP provides NADPH to counteract oxidative stress, while non-oxPPP generates ribosephosphate R5P to provide precursors for nucleotide synthesis G6PD, a unique rate-limiting enzyme in the PPP, is involved in counteracting cellular senescence.

Mechanistically, G6PD knockdown increases the levels of P21, a classical marker of senescence, which promotes HCC and HCT cell senescence In addition, TAp73 enhances cell cycle inhibitory protein P21 by regulating metabolism TAp73 directly activates G6PD to increase PPP flow to inhibit cancer cell senescence Thus, G6PD may mediate, at least in part, the regulation of the senescence phenotype of cancer cells by TAp G6PD also regulates cellular senescence by affecting telomerase activity , G6PD-deficient fibroblasts exhibit delayed growth and accelerated senescence.

Ectopic expression of the human telomerase reverse transcriptase hTERT activated telomerase activity to prevent cellular senescence in G6PD-deficient fibroblasts Thus, the knockdown of hTERT significantly reduces G6PD expression and telomerase activity to promote cancer cell senescence Pharmacological inhibition of 6PGD induces cellular senescence and MCF-7 cell cycle arrest Non-oxPPP also regulates cellular senescence.

Supplementation of ribose 5-phosphate in drug-induced senescent human dermal fibroblasts significantly inhibited cell enlargement, a morphological alteration of cellular senescence In addition, hTERT knockdown inhibited the expression and activity of transketolase, a key enzyme of non-oxppp; increased cancer cell senescence; and reduced tumor burden in a mouse model of heterotypic xenograft SRSPs describe the diverse array of proinflammatory and profibrotic factors secreted by senescent cells SRSPs include cytokines, chemokines, growth factors, proteases, and extracellular matrix components that can affect the surrounding tissue microenvironment and modulate various biological processes, including inflammation, immunity, tissue remodeling, and tumorigenesis SRSPs are considered crucial drivers of chronic inflammation and senescence phenotypes, as they accumulate throughout normal senescence and in age-related diseases.

SRSPs can have both beneficial and detrimental effects on the organism, depending on the expression context and duration. While SRSPs can promote wound healing, tissue repair, and immune surveillance by stimulating cell proliferation, angiogenesis, and immune cell recruitment , they can also induce cellular senescence in neighboring cells, disrupt tissue homeostasis and function, and facilitate the development and progression of age-related diseases such as cancer, neurodegeneration, cardiovascular disease, diabetes, and osteoporosis , Growing evidence has shown that lipid metabolism and SRSPs are interconnected at multiple levels.

Conversely, some SRSP factors, such as IL-1β, IL-6, IL-8, and TNF-α, can affect lipid metabolism by altering lipogenic enzymes e. Disturbances in glucose metabolism can also induce cellular senescence and SRSPs in various tissues, such as pancreatic beta cells, endothelial cells, neurons, astrocytes, myocytes, and adipocytes — Conversely, SRSPs can modulate its metabolic activity by altering the expression and activity of key metabolic enzymes and transcription factors such as AMPK , PGC-1α, sirtuin 1 , FoxO , NF-κB , and nuclear factor erythroid 2-related factor 2 These factors can regulate various aspects of glucose metabolism such as glycolysis, gluconeogenesis, glycogen synthesis and breakdown, pentose phosphate pathway, and the hexosamine biosynthetic pathway.

SRSPs can also impair glucose uptake and utilization in these tissues by interfering with insulin signaling and inducing insulin resistance Apoptosis and autophagy are two major forms of programmed cell death that play important roles in senescence and age-related diseases.

Apoptosis is a regulated process of cell elimination that involves the activation of caspases, cleavage of cellular substrates, and formation of apoptotic bodies that are phagocytosed by macrophages or neighboring cells Autophagy is a catabolic process of self-digestion that involves the formation of double-membrane vesicles called autophagosomes, which engulf cytoplasmic components and deliver them to lysosomes for degradation Both apoptosis and autophagy are essential for maintaining cellular homeostasis, tissue integrity, and organismal health, as they remove damaged or redundant cells and organelles, recycle nutrients, and regulate inflammation and immunity However, both processes also decline with senescence, leading to the accumulation of dysfunctional cells and organelles, oxidative stress, chronic inflammation, and impaired tissue repair and regeneration The relationship between apoptosis and autophagy in senescence is complex and context-dependent.

On the one hand, apoptosis and autophagy can cooperate or compensate for each other to maintain cellular quality control and prevent senescence or tumorigenesis.

For example, autophagy can remove damaged mitochondria that produce ROS and trigger apoptosis Autophagy can also degrade pro-apoptotic factors or inhibit apoptotic signaling pathways, thus protecting cells from excessive or inappropriate cell death.

On the other hand, apoptosis and autophagy can antagonize or compete to modulate cellular fate and function. For instance, apoptosis can inhibit autophagy by cleaving autophagy-related proteins or blocking autophagosome-lysosome fusion. Apoptosis can also induce autophagy as a survival mechanism or a secondary mode of cell death when caspase activation is impaired or overwhelmed The balance between apoptosis and autophagy in senescence is influenced by various factors, including genetic background, environmental stimuli, hormonal status, metabolic state, and disease conditions.

Lipid and glucose metabolism are the two main pathways that provide energy and substrates for cellular function and survival. Dysregulation of lipid or glucose metabolism can affect the balance between apoptosis and autophagy. Lipid metabolism is regulated by various factors, including hormones, nutrients, and oxygen levels.

Lipid metabolism can modulate apoptosis and autophagy by affecting the production of ROS, activation of signaling pathways, and formation of lipid droplets or membrane structures For example, excessive lipid accumulation can induce oxidative stress and ER stress, which can trigger apoptosis or autophagy by activating the JNK, p53, or PKR-like endoplasmic reticulum kinase PERK pathways — Conversely, lipid depletion can also induce apoptosis or autophagy by impairing mitochondrial function, reducing ATP levels, or activating the AMPK pathway , Moreover, lipid metabolism can influence autophagic membrane initiation and elongation by providing phospholipids or fatty acids as precursors or modulators Lipid metabolism can be regulated by autophagy through the degradation of lipid droplets or lipogenic enzymes in a process called lipophagy Similarly, glucose metabolism can regulate apoptosis and autophagy by affecting the production of ROS, activation of signaling pathways, and maintenance of energy homeostasis.

For example, high glucose levels can induce oxidative stress and ER stress, which can trigger apoptosis or autophagy by activating the p38 MAPK, NF-κB, or CHOP pathways — Conversely, low glucose levels can also induce apoptosis or autophagy by impairing mitochondrial function, reducing ATP levels, or activating the AMPK pathway Moreover, glucose metabolism can influence autophagy induction and progression by providing hexosamines or acetyl-CoA as regulators , Furthermore, glucose metabolism can be regulated by autophagy through the degradation of glycogen granules or glycolytic enzymes in a process called glycophagy The relationship between insulin signaling and glucose and lipid metabolism is essential for maintaining energy homeostasis and preventing metabolic diseases.

Insulin also stimulates lipid synthesis and storage by activating the same pathway and downstream targets such as SREBPs, ACC, and FAS Insulin also inhibits glucose production by suppressing the expression and activity of enzymes involved in gluconeogenesis, such as phosphoenolpyruvate carboxykinase PEPCK and glucosephosphatase G6Pase , or lipolysis, such as HSL and ATGL However, insulin signaling and glucose and lipid metabolism can be disrupted by various factors such as obesity, inflammation, oxidative stress, endoplasmic reticulum stress, and senescence.

These factors can impair insulin action and induce insulin resistance by interfering with insulin receptor function or downstream signaling components. For instance, oxidative stress can reduce insulin receptor tyrosine phosphorylation and Akt activation by increasing the activity of protein tyrosine phosphatase 1B or protein kinase C PKC Mitochondrial dysfunction can impair lipid oxidation and increase lipid accumulation in non-adipose tissues such as skeletal muscle or liver, which can cause lipotoxicity and impair insulin action by activating PKC, JNK, or IKKβ Endoplasmic reticulum stress can also induce insulin resistance by activating the unfolded protein response UPR , which can suppress insulin receptor expression or activate JNK or PERK, which can phosphorylate IRS-1 on serine residues and inhibit its tyrosine phosphorylation Conversely, senescence can also affect insulin signaling and glucose and lipid metabolism by reducing the expression or activity of insulin receptors or their substrates, increasing the levels of inflammatory cytokines or oxidative stress markers, impairing mitochondrial function or autophagy, or altering the composition or function of gut microbiota.

Several transcription factors play pivotal roles in modulating insulin action and lipid homeostasis in response to senescence. FoxO factors can induce the expression of genes involved in gluconeogenesis such as PEPCK and G6Pase , or genes involved in FAO such as CPT1 or ACOX , thereby antagonizing the effects of insulin on glucose and lipid metabolism FoxO factors can also induce the expression of genes involved in antioxidant defense such as superoxide dismutase or catalase, or genes involved in anti-inflammatory responses, such as IL or suppressor of cytokine signaling 3, thereby protecting against oxidative stress and inflammation induced by senescence FoxO factors can also induce the expression of genes involved in autophagy, such as LC3 or Atg12 , thereby promoting the clearance of damaged organelles or proteins accumulated during senescence Glucose is one of the main sources of cellular energy.

Excessive glucose intake and abnormally elevated circulating blood glucose levels are associated with chronic diseases such as obesity and diabetes. On the one hand, the metabolic changes caused by hyperglycemia and diabetes promote cellular senescence, leading to tissue dysfunction and various complications such as diabetic retinopathy.

Pancreatic β-cells sense elevated blood glucose levels and secrete insulin correspondingly to maintain blood glucose levels within a narrow range. Glycolysis is increased in diabetic β-cells, which inhibits β-cell function A diabetic environment and SRSPs are considered drivers of diabetic retinopathy senescence in vitro and in vivo.

High glucose exposure accelerates cellular senescence However, glycolysis was significantly reduced rather than increased in an in vitro model of senescence. The presence of a negative feedback regulatory mechanism in an in vitro model may reduce levels of the glucose transport proteins GLUT1 and GLUT3 and the glycolytic enzyme PFKFB3, affecting the rate of glucose transport On the other hand, cellular senescence may also affect insulin secretion and sensitivity, thereby increasing the risk of developing diabetes.

Adipose precursor cells affect neighboring non-senescent cells through the secretion of SRSP factors such as activin A, IL-6, and TNFα, leading to impaired adipogenesis and reduced insulin sensitivity, which may also be involved in T2D progression Abnormal lipid accumulation during senescence is mainly caused by increased fatty acid uptake, de novo lipogenesis, and decreased FAO processes.

Mitochondrial integrity and autophagy induction are also diminished during senescence, leading to decreased lipolysis. These changes further impart lipotoxicity to the cell, depleting energy in the tissues and altering cellular signaling, thereby accelerating senescence and the early onset of age-related diseases Multiple adipogenesis enzymes are upregulated in senescent hepatocytes, including FAS, ACC, and stearoyl-CoA desaturase SCD In addition to lipid synthesis, excessive lipid intake increases FAO in the mitochondria and the ROS produced, leading to mitochondrial DNA damage and mitochondrial dysfunction Excessive lipid intake inhibits autophagy activation signaling and reduces autophagy levels.

Reduced autophagy levels lead to the accumulation of large amounts of waste products and toxins in the cell, interfering with cellular function and homeostasis and accelerating cellular senescence Excess saturated fatty acids activate the NF-κB and JNK signaling pathways by binding to Toll-like receptor 4 on the cell surface, thereby inducing the expression and release of inflammatory factors , Stress can impair glucose and lipid metabolism by altering the levels of insulin, leptin, cortisol, and other hormones that modulate appetite, energy expenditure, and glucose uptake Stress can also induce lipotoxicity, which is the accumulation of excess lipids and their metabolites in non-adipose tissues, such as the liver, muscle, heart, and brain Lipotoxicity can cause cellular damage, inflammation, insulin resistance, and apoptosis, leading to metabolic disorders and age-related diseases Moreover, stress can affect the mitochondrial function and autophagy of cells, which are involved in glucose and lipid metabolism Mitochondria are organelles that produce energy from glucose and fatty acids, while autophagy is the process that degrades damaged or excess cellular components, such as lipids.

Stress can induce mitochondrial stress, which is the imbalance between mitochondrial biogenesis and degradation, resulting in mitochondrial dysfunction, oxidative stress, and impaired energy metabolism Stress can also modulate autophagy, which can have both beneficial and detrimental effects on senescence depending on the stress type and duration.

Autophagy can enhance stress resistance and protein homeostasis by removing damaged mitochondria and lipids, but it can also promote cell death and inflammation by activating ferroptosis, a form of iron-dependent lipid peroxidation. Environmental pollution, especially air pollution, can expose the body to various harmful substances, such as particulate matter, ozone, nitrogen dioxide, and polycyclic aromatic hydrocarbons.

These substances can induce oxidative stress, inflammation, and endothelial dysfunction, which can impair glucose and lipid metabolism — Oxidative stress can damage DNA, proteins, and lipids, leading to cellular senescence and apoptosis Inflammation can alter the levels of cytokines, such as IL-6 and TNF-α, which can affect the secretion and action of insulin, a hormone that regulates glucose uptake Glucose and lipid metabolism are regulated by various hormones, enzymes, and signaling pathways that respond to temperature changes, such as thermogenesis, cold exposure, or heat stress , , Temperature can modulate glucose and lipid metabolism by altering the levels of adipokines, thyroid hormones, catecholamines, and other hormones that modulate energy balance, thermoregulation, and glucose uptake.

Temperature can also affect lipid lipolysis and oxidation, which are the processes that break down and utilize lipids for energy. Lipolysis is the hydrolysis of triglycerides into fatty acids and glycerol, while oxidation is the conversion of fatty acids into acetyl-CoA, which is used by the Krebs cycle.

Temperature can induce lipolysis and oxidation by activating HSL, ATGL, and CPT, which are enzymes that catalyze lipid breakdown and transport. Temperature can also affect the glycerolipid metabolism and signaling of cells, which are involved in glucose and lipid metabolism Therefore, temperature can influence senescence through multiple aspects of glucose and lipid metabolism that affect cellular health and function Figure 3.

Figure 3. Environmental factors influence senescence through glucose and lipid metabolism. Environmental factors such as excessive glucose intake, lipid overaccumulation, stress, environmental pollution, and temperature can promote cellular senescence by increasing glycolysis and inhibiting fatty acid oxidation.

POI is associated with metabolic disturbances, including glucose and lipid metabolism, which may increase the risk of long-term complications, such as cardiovascular diseases and osteoporosis Glucose metabolism is regulated by insulin. Estrogen, the main ovarian hormone, modulates insulin sensitivity and glucose homeostasis by influencing insulin secretion, signaling, and action in various tissues.

Estrogen deficiency in POI may impair glucose metabolism and lead to insulin resistance, hyperglycemia, or diabetes mellitus Estrogen also affects lipid metabolism by regulating the expression and activity of enzymes and receptors involved in lipid synthesis, transport, and degradation Zhou et al.

Moreover, they identified several metabolites related to glucose metabolism and lipid metabolism that were altered in patients with POI, including lactate, pyruvate, citrate, carnitine, acylcarnitine, glycerol, and glycerophospholipids.

In conclusion, POI affects not only ovarian function but also metabolic health. POI patients may have impaired glucose and lipid metabolism due to estrogen deficiency, which may increase their risks of developing metabolic disorders or cardiovascular diseases.

Therefore, it is important to monitor and manage the metabolic status of POI patients and provide appropriate hormonal replacement therapy or lifestyle interventions to prevent or reduce the adverse outcomes associated with POI.

Platelets are cell fragments without nuclei that participate in hemostasis and thrombosis. The levels of lipid peroxides in platelets increase during senescence. Lipid peroxidation is important for cell function. It causes extensive damage to cell membranes and subcellular granules, leading to enzyme inactivation and destruction, and inhibition of metabolic pathways and cell division.

The accumulation of lipid peroxidation products in aged platelets may cause cumulative damage to the membrane structure of platelets and their subcellular granules such as alpha granules, mitochondria, and the endoplasmic reticulum.

Lipid peroxidation may directly affect metabolic pathways by reducing the activity of lipid-dependent enzymes such as NADH- or succinate-cytochrome c reductase or indirectly by releasing lysosomal enzymes from alpha granules Platelets have a rich glycocalyx on their surface, and platelet glycosylation plays an important role in physiological hemostasis mechanisms, regulating platelet-receptor protein interactions, and dynamically remodeling surface glycosylation through their own glucose metabolism system.

Platelet glycosylation also participates in platelet senescence and clearance to regulate platelet count; meanwhile, platelet glycosylation abnormalities are closely related to primary immune thrombocytopenia, coronary heart disease, and related diseases, and are potential targets for anti-platelet therapy Down syndrome DS is a genetic disorder caused by an extra copy of chromosome 21, which results in cognitive impairment, physical abnormalities, and increased risks of age-related comorbidities.

Individuals with DS typically exhibit premature senescence phenomena, including skin senescence, graying hair, cataracts, osteoporosis, atherosclerosis, immune decline, and memory loss Likewise, the glucose and lipid metabolism of patients with DS also changes, mainly manifesting as reduced serum cholesterol levels, increased triglyceride levels, increased insulin resistance, abnormal glucose tolerance, etc.

These changes may be related to genetic abnormalities, oxidative stress, mitochondrial dysfunction, and other factors in these patients.

Moreover, many changes occur in the brain lipids of patients with DS, including a significant reduction in esterified polyunsaturated fatty acids, especially fatty acids esterified into phosphatidylinositol and phosphatidylserine.

Lipid and glucose metabolism are two essential metabolic pathways that provide energy and building blocks for cancer cells. Cancer cells reprogram their lipid and glucose metabolism to support their rapid growth, survival, invasion, and resistance to therapy.

Cancer cells also enhance their lipid uptake by expressing more lipoprotein receptors, such as low-density lipoprotein receptor LDLR and scavenger receptor class B type I SR-BI , and lipid transporters, such as fatty acid transport proteins and FABPs , Moreover, cancer cells can mobilize lipids from adipose tissue or surrounding stromal cells through lipolysis or lipophagy , Lipids can be stored in lipid droplets or oxidized through mitochondrial β-oxidation to generate ATP and acetyl-CoA, which can fuel the TCA cycle and oxidative phosphorylation.

In response to this feature, ongoing efforts aim to adapt lipid metabolism as an anti-cancer drug. Some approaches are used in preclinical models in vitro and in vivo , and some drugs have entered clinical trials. Inhibiting lipid biosynthesis enzymes, such as FAS TVB , ACC Soraphen A , and SCD A , can induce endoplasmic reticulum stress and activate the unfolded protein response, leading to cell cycle arrest, apoptosis, or senescence.

By blocking lipid uptake receptors like LDLR GW or SR-BI ML , less cholesterol and other lipids are available for membrane production and signaling.

This blocking can also impair mitochondrial beta-oxidation and increase the accumulation of toxic lipids, such as ceramide and ROS, by inhibiting the lipid oxidase CPT1 etomoxir Inhibition of COX-2 or LPA receptors, lipid signaling molecules, modulates various pathways involved in cancer cell proliferation, survival, migration, angiogenesis, and inflammation , Finally, cancer progression can also be inhibited by altering the biophysical properties fluidity, curvature, and permeability of the membrane bilayer through the localization and activity of membrane-associated proteins and receptors, as typified by statins that reduce cholesterol concentrations in the plasma membrane and affecting lipid raft clustering and displacement of receptors in non-raft domains The main pathway of glucose metabolism in cancer cells is aerobic glycolysis, also known as the Warburg effect This is a phenomenon in which cancer cells preferentially convert glucose into lactate, even in the presence of oxygen and functional mitochondria.

Aerobic glycolysis allows cancer cells to rapidly consume glucose and produce ATP while avoiding the production of ROS that can damage DNA and proteins. Moreover, aerobic glycolysis provides cancer cells with various metabolic intermediates that can be used for the synthesis of nucleotides, amino acids, and lipids To support aerobic glycolysis, cancer cells increase their glucose uptake by overexpressing GLUTs, especially GLUT1 Cancer cells also upregulate key glycolytic enzymes, such as HK2, phosphofructokinase 1, pyruvate kinase M2, and LDHA, which are often regulated by oncogenic signals, such as KRAS, MYC, and HIF1α However, aerobic glycolysis is not the only mode of glucose metabolism in cancer cells.

Some cancer cells can also use oxidative phosphorylation OXPHOS to generate ATP from glucose-derived pyruvate in the mitochondria. OXPHOS can be activated in cancer cells under conditions, such as hypoxia, nutrient deprivation, or drug resistance OXPHOS is more efficient than glycolysis in terms of ATP production per glucose molecule but also generates more ROS.

OXPHOS can also provide cancer cells with acetyl-CoA and NADH, which can modulate various signaling pathways, such as histone acetylation and sirtuin activity. To support OXPHOS, cancer cells can modulate their pyruvate metabolism by regulating the expression or activity of pyruvate dehydrogenase, pyruvate dehydrogenase kinase, or pyruvate carboxylase.

Cancer cells can also use alternative substrates for OXPHOS, such as glutamine, fatty acids, or ketone bodies Cellular senescence can occur in both normal and cancer cells and has complex dependence effects on cancer development and treatment. On the one hand, cellular senescence is a cancer suppressor mechanism that prevents the proliferation of damaged or malignant cells.

SRSPs secreted by senescent cells recruit and activate immune cells to remove senescent cells Some immune cells, such as T helper 1 cells, can also trigger cancer cell senescence by secreting inflammatory cytokines. In addition, cellular senescence enhances the expression of cancer-associated antigens and immunogenic molecules on cancer cells, making them more susceptible to recognition and clearance by T cells.

Thus, T cells are a key component of cancer immunotherapy On the other hand, cellular senescence can also adversely affect T cell immunity and cancer therapy. First, senescent cells accumulate in cancers and normal tissues, releasing SRSPs with pro-tumorigenic effects such as promoting angiogenesis, invasion, metastasis, and drug resistance.

Second, T cell senescence decreases their proliferation, cytokine production, and cytotoxicity and increases the expression of inhibitory receptors and pro-apoptotic molecules , Therefore, cellular senescence is a double-edged sword that can influence T cell immunity and cancer therapy in different ways.

Understanding the molecular mechanisms and interactions between how sugar and lipid metabolism regulate T cell senescence could provide new insights and strategies for improving anti-cancer therapy. T cell metabolism is tightly linked to T cell activation, differentiation, and function.

T cells undergo metabolic reprogramming upon antigen stimulation, switching from a quiescent state that relies on oxidative phosphorylation to a highly glycolytic state that supports rapid proliferation and effector functions However, chronic or excessive stimulation can lead to T cell exhaustion or senescence, which are associated with metabolic dysregulation, such as impaired glucose uptake, reduced glycolytic capacity, and altered mitochondrial function In addition to glucose metabolism, lipid metabolism plays a crucial role in regulating T cell immunity.

Senescent T cells have reduced FAO capacity and increased dependence on FAS. Targeting FAS with pharmacological inhibitors or gene knockdown induces apoptosis in senescent non-functional T cells and improves anti-cancer immunity.

Conversely, enhancing FAO with pharmacological activators or gene overexpression can prevent or reverse T cell senescence and exhaustion by restoring mitochondrial function and reducing oxidative stress The relationships between different types of lipid components, glucose metabolites, and senescence have been reported , However, an in-depth study of the regulatory relationships of the key enzymes is necessary to screen anti-senescence drugs.

New anti-cancer drugs may also be developed by targeting tumor cell senescence. Various aspects of glucose and lipid metabolism are closely related to senescence. In lipid metabolism, decreased fatty acid catabolism is directly related to senescence onset, including mitochondrial β-oxidation 34 , 36 , 37 , 57 , , peroxisomal β-oxidation 54 , 55 , α-oxidation 82 , 84 , and ω-oxidation The most prominent key enzymes include CPT1 and ACOX1.

Drugs such as fenofibrate can exert anti-senescence effects by targeting PPARα, the gene upstream of CPT1 and ACOX1 Figure 4. In addition to fatty acid catabolism and synthesis, acetyl coenzyme A undergoes a variety of pathways to ketone bodies, phospholipids, sphingolipids, and sterols, which perform a wide range of biological functions and exert pro- or anti-senescence effects Figure 5.

The effects of these lipids on senescence often depend on the amount, subtype, and state of the cellular microenvironment. Cer induces cellular senescence, whereas S1P, another sphingolipid component, delays senescence and extends lifespans , Figure 4.

Targeting critical enzymes of fatty acid synthesis and catabolism to delay senescence. Activation of CPT1 and ACOX1 directly or indirectly through the peroxisome proliferator-activated receptor α PPARα pathway promotes fatty acid oxidation, both of which may have a senescence-delaying effect.

AMPK also inhibits senescence by inhibiting the mTOR and NF-κB pathways. The gray arrows represent normal metabolic pathways. The green pointed and blunt red arrows represent activating and inhibiting effects, respectively.

Figure 5. Various metabolic products of fatty acids have rich biological functions and affect senescence. Multiple glucose metabolism pathways are involved in the regulation of intracellular senescence.

Several rate-limiting enzymes in glycolysis, including HK2 and PK, are highly expressed in senescent cells , Although the pharmacological activation of Nrf2 increases HK2 expression 18 , other mechanisms require further investigation.

Several key enzymes of the PPP, a branch of the glycolytic pathway, are also involved in the regulation of intracellular senescence. The key enzymes promote cellular senescence by upregulating P21, P16, and telomerase activity Notably, the dietary TCA-cycling metabolites citrate and αKG delay cellular senescence 23 — Therefore, supplementation of these metabolites may provide an effective intervention for the treatment of senescence-related dysfunction Figure 6.

Figure 6. Glucose metabolism is upregulated in senescent cells. Multiple enzymes of glycolysis are upregulated. For instance, lactate release mediated by monocarboxylate transporters MCTs is involved in senescence-related secretory phenotypes SRSPs.

Several metabolites in the tricarboxylic acid TCA cycle, including α-ketoglutarate αKG and citrate, delay senescence. Environmental factors can affect senescence through glucose and lipid metabolism, which are essential for energy production and cellular function.

Alterations in the external environment such as nutrition, stress, environment, pollution, and temperature accelerate cellular senescence by regulating glucose and lipid metabolism through a variety of hormones, enzymes, and signaling pathways.

Thus, targeting aberrant metabolism may delay normal cellular senescence. In summary, fatty acid and glucose metabolism are essential in the maintenance of normal cellular function, both of which are disturbed by senescence.

Conversely, disturbances in fatty acid and glucose metabolism can further lead to senescence. Two pathway-related enzymes and metabolites are involved in the regulation of cellular senescence. While many advances have been made in recent years, several important issues must still be addressed, including: a What are the initiating factors of senescence and metabolic disorders?

b While reports suggest a correlation between metabolic disorders and senescence, the true causal relationship remains undetermined. c Anti-senescence therapies proven to work in humans are scarce.

Understanding the role of enzymes and metabolites in fatty acid and glucose metabolism may provide answers for a more comprehensive understanding of senescence. BL and QM investigated and wrote the first draft of the manuscript.

XG and HS wrote sections of the manuscript. ZX and YW contributed to conception the study and acquire funds. HZ contributed to design of the study and acquire funds. BL, QM, XG, HS, ZX, YW, and HZ were involved in drafting, revising the manuscript, and agree to be accountable for the content of the work.

All authors contributed to the article and approved the submitted version. This work was supported by the National Natural Science Foundation of China Nos. We sincerely thank the reviewers and editors of Frontiers in Nutrition for providing valuable suggestions on this paper.

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers.

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Burton, DG, and Krizhanovsky, V. Physiological and pathological consequences of cellular senescence. Cell Mol Life Sci. doi: PubMed Abstract CrossRef Full Text Google Scholar. López-Otín, C, Blasco, MA, Partridge, L, Serrano, M, and Kroemer, G.

The hallmarks of aging. Ma, R, Kutchy, NA, Chen, L, Meigs, DD, and Hu, G. Primary cilia and ciliary signaling pathways in aging and age-related brain disorders. Neurobiol Dis.

Kubben, N, and Misteli, T. Shared molecular and cellular mechanisms of premature ageing and ageing-associated diseases. Nat Rev Mol Cell Biol. Galluzzi, L, Vitale, I, Aaronson, SA, Abrams, JM, Adam, D, Agostinis, P, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death Cell Death Differ.

Gorgoulis, V, Adams, PD, Alimonti, A, Bennett, DC, Bischof, O, Bishop, C, et al. Cellular senescence: defining a path forward. Barzilai, N, Huffman, DM, Muzumdar, RH, and Bartke, A. The critical role of metabolic pathways in aging.

Riera, CE, and Dillin, A. Tipping the metabolic scales towards increased longevity in mammals. Nat Cell Biol. Dall'armi, C, Devereaux, KA, and Di Paolo, G. The role of lipids in the control of autophagy. Curr Biol.

Grunt, TW. Interacting cancer machineries: cell signaling, lipid metabolism, and epigenetics. Trends Endocrinol Metab.

Sunshine, H, and Iruela-Arispe, ML. Membrane lipids and cell signaling. Curr Opin Lipidol. Toth, MJ, and Tchernof, A. Lipid metabolism in the elderly.

Eur J Clin Nutr. Sênos Demarco, R, Clémot, M, and Jones, DL. The impact of ageing on lipid-mediated regulation of adult stem cell behavior and tissue homeostasis.

Mech Ageing Dev. Kuhla, A, Blei, T, Jaster, R, and Vollmar, B. Aging is associated with a shift of fatty metabolism toward lipogenesis. J Gerontol A Biol Sci Med Sci. Kim, YM, Shin, HT, Seo, YH, Byun, HO, Yoon, SH, Lee, IK, et al. Sterol regulatory element-binding protein Srebp mediated lipogenesis is involved in cell senescence.

J Biol Chem. Wang, Q, Nie, L, Zhao, P, Zhou, X, Ding, Y, Chen, Q, et al. Diabetes fuels periodontal lesions via Glut1-driven macrophage inflammaging.

Int J Oral Sci. Yao, GD, Yang, J, Li, XX, Song, XY, Hayashi, T, Tashiro, SI, et al. Blocking the utilization of glucose induces the switch from senescence to apoptosis in pseudolaric acid B-treated human lung cancer cells in vitro.

Acta Pharmacol Sin. Hariton, F, Xue, M, Rabbani, N, Fowler, M, and Thornalley, PJ. Sulforaphane delays fibroblast senescence by curbing cellular glucose uptake, increased glycolysis, and oxidative damage. Oxidative Med Cell Longev. Stolzing, A, Coleman, N, and Scutt, A. Glucose-induced replicative senescence in mesenchymal stem cells.

Rejuvenation Res. Wiley, CD, and Campisi, J. The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab. Li, X, Zhang, Z, Zhang, Y, Cao, Y, Wei, H, and Wu, Z. Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16 INK4a inactivation.

J Exp Clin Cancer Res. Asadi Shahmirzadi, A, Edgar, D, Liao, CY, Hsu, YM, Lucanic, M, Asadi Shahmirzadi, A, et al. Alpha-ketoglutarate, an endogenous metabolite, extends lifespan and compresses morbidity in aging mice. Cell Metab. Chin, RM, Fu, X, Pai, MY, Vergnes, L, Hwang, H, Deng, G, et al.

The metabolite alpha-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Wang, Y, Deng, P, Liu, Y, Wu, Y, Chen, Y, Guo, Y, et al. Alpha-ketoglutarate ameliorates age-related osteoporosis via regulating histone methylations.

Nat Commun. Fan, SZ, Lin, CS, Wei, YW, Yeh, SR, Tsai, YH, Lee, AC, et al. Dietary citrate supplementation enhances longevity, metabolic health, and memory performance through promoting Ketogenesis.

Aging Cell. CrossRef Full Text Google Scholar. Zhao, Y, Liu, X, Si, F, Huang, L, Gao, A, Lin, W, et al. Citrate promotes excessive lipid biosynthesis and senescence in tumor cells for tumor therapy.

Adv Sci Weinh. Adeva-Andany, MM, Carneiro-Freire, N, Seco-Filgueira, M, Fernández-Fernández, C, and Mouriño-Bayolo, D. Mitochondrial Β-oxidation of saturated fatty acids in humans.

Ferdinandusse, S, Rusch, H, Van Lint, AE, Dacremont, G, Wanders, RJ, and Vreken, P. Stereochemistry of the peroxisomal branched-chain fatty acid alpha- and beta-oxidation systems in patients suffering from different peroxisomal disorders. J Lipid Res. Miura, Y. The biological significance of Ω-oxidation of fatty acids.

Proc Jpn Acad Ser B Phys Biol Sci. Nakamura, MT, Yudell, BE, and Loor, JJ. Regulation of energy metabolism by long-chain fatty acids.

Prog Lipid Res. Puchalska, P, and Crawford, PA. Metabolic and signaling roles of ketone bodies in health and disease. Annu Rev Nutr. Carracedo, A, Cantley, LC, and Pandolfi, PP. Cancer metabolism: fatty acid oxidation in the limelight.

Nat Rev Cancer. Peterson, JM, Aja, S, Wei, Z, and Wong, GW. Ctrp1 protein enhances fatty acid oxidation via amp-activated protein kinase AMPK activation and acetyl-Coa carboxylase ACC inhibition. Braun, F, Rinschen, MM, Bartels, V, Frommolt, P, Habermann, B, Hoeijmakers, JH, et al.

Altered lipid metabolism in the aging kidney identified by three layered Omic analysis. Aging Albany NY. Murea, M, Freedman, BI, Parks, JS, Antinozzi, PA, Elbein, SC, and Ma, L. Lipotoxicity in diabetic nephropathy: the potential role of fatty acid oxidation.

Clin J Am Soc Nephrol. Houtkooper, RH, Argmann, C, Houten, SM, Cantó, C, Jeninga, EH, Andreux, PA, et al. The metabolic footprint of aging in mice. Sci Rep.

Wang, Y, Chen, Y, Guan, L, Zhang, H, Huang, Y, Johnson, CH, et al. Carnitine Palmitoyltransferase 1C regulates cancer cell senescence through mitochondria-associated metabolic reprograming. Vaiserman, A, and Krasnienkov, D. Telomere length as a marker of biological age: state-of-the-art, open issues, and future perspectives.

Front Genet. Guo, L, Chen, Y, Li, H, Yin, F, Ge, M, Hu, L, et al. Telomere length is maternally inherited and associated with lipid metabolism in Chinese population. Belal, SA, Sivakumar, AS, Kang, DR, Cho, S, Choe, HS, and Shim, KS.

Modulatory effect of linoleic and oleic acid on cell proliferation and lipid metabolism gene expressions in primary bovine satellite cells. Anim Cells Syst Seoul. Ma, Y, Zha, J, Yang, X, Li, Q, Zhang, Q, Yin, A, et al. Long-chain fatty acyl-CoA synthetase 1 promotes prostate cancer progression by elevation of lipogenesis and fatty acid beta-oxidation.

Rayess, H, Wang, MB, and Srivatsan, ES. Cellular senescence and tumor suppressor gene P Int J Cancer. Rufini, A, Tucci, P, Celardo, I, and Melino, G. Senescence and aging: the critical roles of P Guan, L, Chen, Y, Wang, Y, Zhang, H, Fan, S, Gao, Y, et al.

Effects of carnitine palmitoyltransferases on cancer cellular senescence. J Cell Physiol. Zhuang, W, Lian, G, Huang, B, Du, A, Gong, J, Xiao, G, et al.

CPT1 regulates the proliferation of pulmonary artery smooth muscle cells through the AMPK-PP21 pathway in pulmonary arterial hypertension. Mol Cell Biochem. Shao, H, Mohamed, EM, Xu, GG, Waters, M, Jing, K, Ma, Y, et al.

Carnitine palmitoyltransferase 1A functions to repress FOXO transcription factors to allow cell cycle progression in ovarian cancer. Mihaylova, MM, Cheng, CW, Cao, AQ, Tripathi, S, Mana, MD, Bauer-Rowe, KE, et al. Fasting activates fatty acid oxidation to enhance intestinal stem cell function during homeostasis and aging.

Cell Stem Cell. Fadó, R, Rodríguez-Rodríguez, R, and Casals, N. The return of malonyl-CoA to the brain: cognition and other stories. Knobloch, M, Pilz, GA, Ghesquière, B, Kovacs, WJ, Wegleiter, T, Moore, DL, et al. A fatty acid oxidation-dependent metabolic shift regulates adult neural stem cell activity.

Cell Rep. Franceschi, C, Bonafè, M, Valensin, S, Olivieri, F, De Luca, M, Ottaviani, E, et al. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. Malandrino, MI, Fucho, R, Weber, M, Calderon-Dominguez, M, Mir, JF, Valcarcel, L, et al. Enhanced fatty acid oxidation in adipocytes and macrophages reduces lipid-induced triglyceride accumulation and inflammation.

Am J Physiol Endocrinol Metab. Namgaladze, D, Lips, S, Leiker, TJ, Murphy, RC, Ekroos, K, Ferreiros, N, et al. Inhibition of macrophage fatty acid Β-oxidation exacerbates palmitate-induced inflammatory and endoplasmic reticulum stress responses. He, A, Chen, X, Tan, M, Chen, Y, Lu, D, Zhang, X, et al.

Acetyl-Coa derived from hepatic peroxisomal Β-oxidation inhibits autophagy and promotes steatosis via Mtorc1 activation. Mol Cell. Yang, L, Zhang, Y, Wang, S, Zhang, W, and Shi, R. Decreased liver peroxisomal beta-oxidation accompanied by changes in brain fatty acid composition in aged rats.

Neurol Sci. Périchon, R, Bourre, JM, Kelly, JF, and Roth, GS. The role of peroxisomes in aging. Fanelli, F, Sepe, S, D'amelio, M, Bernardi, C, Cristiano, L, Cimini, A, et al.

Age-dependent roles of peroxisomes in the hippocampus of a transgenic mouse model of Alzheimer's disease. Mol Neurodegener. Wang, ZW, Pan, WT, Lee, Y, Kakuma, T, Zhou, YT, and Unger, RH. The role of leptin resistance in the lipid abnormalities of aging.

FASEB J. El Hajj, HI, Vluggens, A, Andreoletti, P, Ragot, K, Mandard, S, Kersten, S, et al. The inflammatory response in acyl-CoA oxidase 1 deficiency pseudoneonatal adrenoleukodystrophy.

Moreno-Fernandez, ME, Giles, DA, Stankiewicz, TE, Sheridan, R, Karns, R, Cappelletti, M, et al.

Insulin signalling and the regulation of glucose and lipid metabolism Pol J Pathol. Combs TP, Berg AH, Obici S, Scherer PE, Rossetti L Endogenous glucose production is inhibited by the adipose-derived protein Acrp Carracedo, A, Cantley, LC, and Pandolfi, PP. In general, Cer and its derivatives induce cellular senescence and promote senescence phenotypes, while S1P and its receptor signaling delay senescence and extend lifespan However, chronic or excessive stimulation can lead to T cell exhaustion or senescence, which are associated with metabolic dysregulation, such as impaired glucose uptake, reduced glycolytic capacity, and altered mitochondrial function CAS PubMed PubMed Central Google Scholar Ryaboshapkina, M.

Lipid metabolism and glucose utilization -

Glucose Homeostasis and Insulin Resistance. Download PDF Flyer Back DOI: Glucose and Lipid Metabolism Author s : Leszek Szablewski Pp: 21 DOI: Cite as. About this chapter ×. Cite this chapter as: Leszek Szablewski ; Glucose and Lipid Metabolism, Glucose Homeostasis and Insulin Resistance 1: 1.

Close About this chapter. Related Journals Current Metabolomics. Current Cell Science. Glucolipid Metabolic Disorders. Current Indian Science. Current Molecular Medicine. Current Topics in Chemistry.

Current Chemical Biology. Because this is a bond-creating anabolic process, ATP is consumed. However, the creation of triglycerides and lipids is an efficient way of storing the energy available in carbohydrates. Triglycerides and lipids, high-energy molecules, are stored in adipose tissue until they are needed.

Although lipogenesis occurs in the cytoplasm, the necessary acetyl CoA is created in the mitochondria and cannot be transported across the mitochondrial membrane. To solve this problem, pyruvate is converted into both oxaloacetate and acetyl CoA. Two different enzymes are required for these conversions.

Oxaloacetate forms via the action of pyruvate carboxylase, whereas the action of pyruvate dehydrogenase creates acetyl CoA. Oxaloacetate and acetyl CoA combine to form citrate, which can cross the mitochondrial membrane and enter the cytoplasm.

In the cytoplasm, citrate is converted back into oxaloacetate and acetyl CoA. Oxaloacetate is converted into malate and then into pyruvate. Pyruvate crosses back across the mitochondrial membrane to wait for the next cycle of lipogenesis.

The acetyl CoA is converted into malonyl CoA that is used to synthesize fatty acids. Figure 6 summarizes the pathways of lipid metabolism. Figure 6. Lipids may follow one of several pathways during metabolism.

Glycerol and fatty acids follow different pathways. Lipids are available to the body from three sources. They can be ingested in the diet, stored in the adipose tissue of the body, or synthesized in the liver. Fats ingested in the diet are digested in the small intestine. The triglycerides are broken down into monoglycerides and free fatty acids, then imported across the intestinal mucosa.

Once across, the triglycerides are resynthesized and transported to the liver or adipose tissue. Fatty acids are oxidized through fatty acid or β-oxidation into two-carbon acetyl CoA molecules, which can then enter the Krebs cycle to generate ATP.

If excess acetyl CoA is created and overloads the capacity of the Krebs cycle, the acetyl CoA can be used to synthesize ketone bodies.

When glucose is limited, ketone bodies can be oxidized and used for fuel. Excess acetyl CoA generated from excess glucose or carbohydrate ingestion can be used for fatty acid synthesis or lipogenesis.

Acetyl CoA is used to create lipids, triglycerides, steroid hormones, cholesterol, and bile salts. Lipolysis is the breakdown of triglycerides into glycerol and fatty acids, making them easier for the body to process.

bile salts: salts that are released from the liver in response to lipid ingestion and surround the insoluble triglycerides to aid in their conversion to monoglycerides and free fatty acids.

cholecystokinin CCK : hormone that stimulates the release of pancreatic lipase and the contraction of the gallbladder to release bile salts.

chylomicrons: vesicles containing cholesterol and triglycerides that transport lipids out of the intestinal cells and into the lymphatic and circulatory systems.

fatty acid oxidation: breakdown of fatty acids into smaller chain fatty acids and acetyl CoA. hydroxymethylglutaryl CoA HMG CoA : molecule created in the first step of the creation of ketone bodies from acetyl CoA. ketone bodies: alternative source of energy when glucose is limited, created when too much acetyl CoA is created during fatty acid oxidation.

monoglyceride molecules: lipid consisting of a single fatty acid chain attached to a glycerol backbone. pancreatic lipases: enzymes released from the pancreas that digest lipids in the diet. triglycerides: lipids, or fats, consisting of three fatty acid chains attached to a glycerol backbone.

Skip to main content. Module 8: Metabolism and Nutrition. PubMed Google Scholar Crossref. Dugani SB, Akinkuolie AO, Paynter N, Glynn RJ, Ridker PM, Mora S. Association of lipoproteins, insulin resistance, and rosuvastatin with incident type 2 diabetes mellitus: secondary analysis of a randomized clinical trial [published online April 13, ].

doi: Google Scholar. Ridker PM, Danielson E, Fonseca FA, et al; JUPITER Study Group. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. Ridker PM, Pradhan A, MacFadyen JG, Libby P, Glynn RJ.

Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: an analysis from the JUPITER trial. Shalaurova I, Connelly MA, Garvey WT, Otvos JD. Lipoprotein insulin resistance index: a lipoprotein particle-derived measure of insulin resistance.

Metab Syndr Relat Disord. Mackey RH, Mora S, Bertoni AG, et al. Lipoprotein particles and incident type 2 diabetes in the Multi-Ethnic Study of Atherosclerosis.

Diabetes Care. PubMed Google Scholar. De Graaf JCP, Sniderman A. ApoB in Clinical Care. Houten, the Netherlands: Springer Media; Taskinen MR, Borén J.

New insights into the pathophysiology of dyslipidemia in type 2 diabetes. See More About Cardiology Dyslipidemia Cardiovascular Risk Factors Endocrinology Diabetes. Download PDF Cite This Citation Pagidipati NJ , Pencina M , Sniderman AD.

Access your subscriptions. Access through your institution. Add or change institution. Free access to newly published articles.

Purchase access. Rent article Rent this article from DeepDyve. Sign in to access free PDF. Save your search. Customize your interests.

Glucose and fatty acids Lipid metabolism and glucose utilization the major Natural weight loss inspiration of energy for human body. Cholesterol, Lipid metabolism and glucose utilization most abundant sterol in mammals, is a key component of cell membranes although it metaolism not glucosse ATP. The metabolisms of glucose, metaboliwm 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. In this issue of JAMA CardiologyDugani et al 2 provide further evidence as to how Liid glucose and lipid metabolism ane. Ridker et gluclse 3 first called attention to utilizatjon association LLipid statin therapy and Best exercises for fat burning risk for new-onset T2DM in the Justification for Lipid metabolism and glucose utilization Use of Utiliztion in Prevention: uti,ization Intervention Trial Evaluating Rosuvastatin JUPITER study. Subsequently, Ridker et al 4 reported that ktilization of those patients affected Lipid metabolism and glucose utilization utilizxtion on metsbolism metabolic trajectory toward T2DM Self-awareness and reflection Lipid metabolism and glucose utilization statins likely only accelerated this transition. In the present study, Dugani et al examine the relationship of the lipoprotein insulin resistance LPIR score, 5 a weighted composite of 6 measures of lipoprotein particles generated by nuclear magnetic resonance, with the incidence of T2DM in patients randomized to rosuvastatin therapy or placebo in the JUPITER study. The LPIR score is an advanced version of the ratio of triglycerides to high-density lipoprotein HDL cholesterol, which with triglycerides and apolipoprotein B ApoB have previously been shown to be significant predictors of the risk for incident T2DM. The authors find that the baseline hazard ratio for T2DM in the highest tertile of the LPIR score was substantially higher than that of the lowest tertile of the LPIR score in the placebo 4. This relationship remained after adjustment for systolic blood pressure, body mass index, and levels of high-sensitivity C-reactive protein, hemoglobin A 1cHDL cholesterol, low-density lipoprotein LDL cholesterol, triglycerides, and ApoB.

Lipid metabolism and glucose utilization -

Selenoprotein P, encoded by the Sepp 1 gene, is a secretory protein produced and secreted primarily by the liver. It is responsible for transporting selenium from the liver to extrahepatic tissues A recent study reported that selenoprotein P regulates insulin action and systemic energy metabolism in rodents and humans Cell-based studies revealed that administration of purified selenoprotein P resulted in impaired insulin signaling through reduced insulin-stimulated phosphorylation of the insulin receptor and AKT in mouse primary hepatocytes and immortalized myocytes in vitro , and increased phosphorylation of insulin receptor substrate 1 IRS1 at Ser In mice, the administration of selenoprotein P induced hepatic and peripheral insulin resistance, whereas both genetic deletion and RNA interference-mediated knockdown of selenoprotein P ameliorated insulin signaling and improved glucose tolerance Additionally, high circulating levels of adiponectin were observed in Sepp 1 knockout mice, revealing the occurrence of crosstalk between the hepatokine selenoprotein P and the adipokine adiponectin In humans, selenoprotein P is increased in patients with T2D and NAFLD , and is positively correlated with triglycerides, glucose, and insulin resistance However, most of current data comes from small sample clinical studies, thus further prospective large-scale studies are warranted.

The immune system is closely linked to metabolic changes, and components are changed in obesity and T2D The detection of immune cells in metabolic tissues and organs, such as macrophages, has highlighted a dynamic, ongoing crosstalk that exists between immune and metabolism regulatory systems Some pro-inflammatory cytokines, such TNF-α and IL-1β, take part in disrupting the insulin and lipid signaling pathways, thereby influencing insulin sensitivity and lipid metabolism Promisingly, some treatments targeting pro-inflammatory cytokines displayed improved glucose metabolism and insulin secretion and sensitivity in T2D TNF-α, a member of TNF family, is a potent pro-inflammatory cytokine and immuno-modulator produced mainly by macrophages and monocytes TNF-α is involved with multiple functions and plays a variety of roles in metabolic disorders TNF-α is a mediator of insulin resistance through its ability to block the action of insulin The TNF-α-mediated insulin resistance is partially through the down-regulation of key genes i.

In mice, administration of exogenous TNF-α could lead to insulin resistance, conversely neutralization of TNF-α improved insulin sensitivity Furthermore, TNF-α deficiency has been shown to significantly improve insulin sensitivity, and lower circulating levels of free fatty acids In humans, the circulating concentration of TNF-α are elevated in T2D, and this alteration is strongly associated with impaired glucose tolerance and enhanced insulin resistance, islet dysfunction, and increased T2D risk — Some studies have shown that a statistically significant reduction in the risk of developing T2D in treatment with TNF inhibition, and the improvement in insulin sensitivity in during prolonged treatment with the anti-TNF-α antibody infliximab in insulin resistant subjects However, most of these studies were not prospective and it is unclear whether these improvements are direct or indirect influences.

Further study is necessary to determine whether TNF-α inhibition can help in the management of human metabolic disease. IL-1β, a member of the IL-1 family, plays an important role in endocrinology and the regulation of responses correlated with inflammatory stress IL-1β is considered to mediate inflammation, steatosis, and fibrosis in liver In β-cells, IL-1β activates the JNK pathway 73 , which is involved in cytokine-mediated apoptosis 74 and mediates oxidative stress-induced suppression of insulin gene transcription Moreover, IL-1β decreases expression of the insulin receptor substrate IRS-1, inhibits glucose transporter GLUT 4 translocation to the plasma membrane, and reduces insulin-stimulated glucose uptake and lipogenesis In mice, IL-1β was found to promote hepatic steatosis by stimulating triglycerides, cholesterol accumulation, and lipid droplet formation and to regulate inflammation, hepatic insulin resistance, and fibrosis In contrast, inhibition of IL-1β was found to attenuate steatosis and liver injury 78 , improve atherosclerosis 79 , and lower glycemia Furthermore, the IL-1β deficiency mice exhibited less hepatic steatosis and intact insulin sensitivity In humans, serum levels of IL-1β are increased in obesity, NAFLD , T2D , and MetS 81 , but decrease significantly after extensive weight loss Emerging clinical studies showed that IL-1 receptor antagonist anakinra and IL-1β-specific antibody gevokizumab, canakinumab, LY treatment improved glucose metabolism and insulin secretion in patients with T2D 61 , Moreover, IL-1 antagonism was well-tolerated with no evidence of drug-related adverse events, apart from reactions at the anakinra injection site IL-6 is a multifunctional cytokine with complex roles and is expressed in numerous cells, including immune cells, skeletal muscle cells, and islet β-cells It has a dual role in modulating insulin sensitivity, acting as both an enhancer and inhibitor of insulin action To exert its biological effects, IL-6 utilizes two pathways: classic and trans-signaling.

While it is generally believed that the classic signaling is participated in the anti-inflammatory and regenerative activities of IL-6, the trans-signaling is involved in the pro-inflammatory responses induced by this cytokine 85 , Moreover, IL-6 is responsible for macrophage recruitment to adipose tissue in obesity, leading to the development of inflammation, insulin resistance, and T2D In addition, IL-6 has been shown to exert long-term inhibitory effects on the gene transcription of IRS-1, GLUT4, and PPAR, along with a marked reduction in IRS-1, and on insulin-stimulated tyrosine phosphorylation and insulin-stimulated glucose transport, which result in impaired insulin signaling and action It is worth noting that the central application of IL-6 suppresses feeding and improves glucose tolerance via enhanced trans-signaling in the CNS of obese mice, even under conditions of leptin resistance Studies have also shown that short- and long-term administration of adipocytes with IL-6 produces different influences on insulin signaling.

Short-term treatment with IL-6 has been found not to impair the effect of insulin in the adipose tissue of rats , but increased glucose uptake in adipocytes In contrast, chronic administration of IL-6 has been found to induce insulin resistance, suppress glucose transport, and reduce insulin-induced lipogenesis 88 , In humans, circulating levels of IL-6 are elevated in T2D, and this cytokine is an independent predictor of T2D MCP-1, a member of the chemokine chemotactic cytokine family, is a powerful monocyte agonist that plays a crucial role in the recruitment of macrophages MCP-1 is mediated by NF-kB activation and oxidative stress 94 , and up-regulated by oxidized lipids, endoplasmic reticulum stress 95 , and high glucose concentrations Additionally, MCP-1 links obesity to insulin resistance and hepatic steatosis Mice that were engineered to express the MCP-1 transgene showed macrophage infiltration into adipose tissue, elevated hepatic triglyceride content, and insulin resistance MCP-1 induces hepatic steatosis and insulin resistance via up-regulating the expression of SREBP- 1 c , a transcription factor that regulates the expression of genes important in lipid synthesis, and glucosephosphatase G6Pase , an enzyme involved in hepatic glucose production In contrast, MCP-1 knockout mice and inhibition of MCP-1 activity exhibited improvements in insulin resistance and hepatic steatosis 96 , Consistent with the findings in mice, humans show increased plasma levels of MCP-1 in T2D Skeletal muscle is considered to be the largest organ in the body of non-obese subjects and is now recognized as an active endocrine organ due to its function in releasing numerous myokines 3.

Myokines are part of a complex communication network within the body which connects skeletal muscle with other organs, such as adipose tissue, liver, and pancreas 3. Recent studies have indicated that myokines, such as irisin, interleukin IL , and IL, are deeply involved in glucose and lipid metabolism via autocrine, paracrine and endocrine activities It is speculated that the contractile activity influences skeletal muscle secretory functions, which may link physical activity to the health-promoting effects of exercise [5].

Irisin, encoded by the Fndc 5 gene, is the cleaved and secreted product of the fibronectin type III domain-containing protein 5 FNDC5. This myokine is a transmembrane protein expressed principally in skeletal muscle , but is also expressed in WAT to a lower extent Irisin participates in energy expenditure via stimulation of the browning of WAT , direct induction of glucose and fatty acid uptake, and regulation of gene expression of metabolic enzymes in human muscle via AMPK activation Cell-based studies revealed that the treatment of recombinant irisin greatly increased uptake of glucose and fatty acids, as well as expression of genes involved in glucose transport and lipid metabolism; however, the expression of genes involved in glycogenolysis or gluconeogenesis was suppressed Furthermore, exogenously administered irisin, adenovirus-over-expressed irisin and irisin transgenic mice exhibited improved glucose homeostasis, ameliorated hepatic steatosis, improved insulin resistance, reduced inflammatory cytokine production, and weight loss , , , FNDC 5 gene deficiency aggravated fat accumulation, obesity, insulin resistance, and inflammation accompanied with enhanced AMPK inhibition, macrophages recruitment, and M1 polarization In humans, irisin levels, and FNDC 5 gene expression are decreased in obese and T2D individuals A previous study reported that irisin increases immediately after exercise and is positively correlated with exercise intensity The positive role of irisin in metabolism supports the idea that moderate exercise is good for health.

However, there are some conflicting findings in the literature To date, the role of irisin in T2D is still not entirely clear. Further research is required to determine the links between irisin and T2D.

IL is well-known as an anti-inflammatory cytokine by inhibiting the secretion of some inflammatory cytokines derived from macrophages and monocytes Recently, however, IL was determined to also serve as a novel myokine that is synthesized and released by human myotubes under the conditions of accelerated glucose uptake and metabolism through autocrine pathway It has been demonstrated that IL is a master regulator of glucose metabolism, working via suppression of hepatic glucose production and directly inhibiting the transcription of hepatic genes that encode key gluconeogenic enzymes, such as PEPCK and G 6 P IL exposure increases skeletal muscle glucose uptake, oxidation, and glycogen synthesis via an Akt-dependent mechanism On the contrary, genetic deletion of IL in mice resulted in hyperglycemia, which progressed to hepatic insulin resistance and systemic metabolic disturbances And basal glycogen synthesis was found to be reduced in cultured myotubes upon exposure to an ILneutralizing antibody However, the molecular mechanisms underlying the regulation of IL expression and release by exercise are, as yet, unclear.

Further research should be undertaken to explore how exercise affects IL expression and secretion. IL, a member of IL-2 superfamily, is a myokine that is highly expressed in skeletal muscle and released by myotubes; it is also produced by a wide variety of other cells and tissues The IL secreted from skeletal muscle communicates with the adipose tissue to stimulate a reduction in fat mass and adipogenesis, and to decrease triglycerides and VLDL in blood , primarily through the UCPs and PPAR-δ signaling pathways It has been demonstrated in animal models and human that IL improves lipid and glucose metabolism, and insulin sensitivity; enhances mitochondrial activity; reduces WAT inflammation; and alleviates endoplasmic reticulum stress Genetic research reported that IL transgenic mice exhibited lean body condition, whereas IL gene knockout mice showed significant increase in weight gain without changes in appetite In humans, plasma IL is significantly decreased in obesity and negatively associated with fat mass Therefore, IL may be a feasible therapeutic target for prevention and treatment in obesity and T2D.

BDNF, a member of the neurotrophic factor family, is a protein produced in skeletal muscle cells that is increased by contraction BDNF regulates neuronal differentiation and synaptic plasticity, and its reduced levels are involved in the pathogenesis of Alzheimer's disease and other disorders It is reported that BDNF increases phosphorylation of AMPK and acetyl CoA carboxylase, enhances fat oxidation , regulates glucose metabolism, and ameliorates insulin sensitivity In obese diabetic mice, BDNF reduces food intake and lowers blood glucose levels Conversely, BDNF-deficient mice displayed hyperphagia, obesity, hyperleptinemia, and hyperinsulinemia In humans, plasma BDNF is decreased in individuals with both obesity and T2D, and is inversely correlated with serum levels of free fatty acids and insulin resistance Moreover, lower BDNF levels are involved with obesity and diabetic complications Bone is not only metabolically active, with glucose as the major energy source, but also actively takes part in systemic energy metabolism 4.

First, osteoblasts can uptake glucose a process primarily mediated by GLUT1 for utilization through aerobic glycolysis Interestingly, the total uptake of glucose by bone exceeds that of traditional glucose-utilizing organs, including muscle, WAT, and the liver Second, numerous key enzymes associated with the glycolytic pathway in carbohydrate metabolism are also present in osteoblasts and osteoclasts Moreover, osteoblasts express both the insulin receptor and GLUT , which provides the basis for bone-mediated regulation of glucose metabolism.

In addition, several osteokines bone-derived cytokines , such as osteocalcin , lipocalin 2 and sclerostin link bone and glucose metabolism, with involvement in modulation of glucose homeostasis, appetite, and browning of adipose tissue.

Osteocalcin, also known as bone γ-carboxyglutamic acid protein, is one of the osteoblast-specific proteins that is an established biomarker of bone turnover, and it is reported to be associated with glucose and fat metabolism , Circulating levels of undercarboxylated and bioactive osteocalcin double during aerobic exercise at the time levels of insulin decrease The endocrine functions of osteocalcin are fulfilled by its undercarboxylated form, termed undercarboxylated osteocalcin ucOC 4.

Osteocalcin receptors are present in both central nervous system and peripheral tissues, for instance, in the pancreas , adipocytes , and muscle , thereby facilitating its physiological functions. There is a growing body of experimental evidence suggesting that ucOC promotes pancreatic β-cell proliferation and insulin expression and secretion , , induces up-regelation of adiponectin in adipocytes to ameliorate insulin resistance , promotes release of glucagon-like peptide-1 to indirectly stimulate insulin secretion , and favors glucose and fatty acid uptake and utilization in muscle during exercise Mice lacking osteocalcin manifested decreased β-cell proliferation, glucose intolerance, and insulin resistance , In contrast, the metabolic abnormalities in these mice were improved by infusion of exogenous ucOC In humans, serum osteocalcin levels are significantly lower in subjects with T2D and MetS However, the results regarding incident T2D are controversial.

In several longitudinal studies, the serum osteocalcin level was found to not associate with the development of T2D Osteopontin, a member of small integrin-binding ligand N-linked glycoproteins SIBLINGs family, is a major non-collagenous bone matrix protein which participates in normal and pathological calcification This glycoprotein is expressed in a variety of cells, including osteoblasts, osteoclasts, macrophages, as well as T-lymphocytes Osteopontin acts as a mediator of obesity-related hepatic alterations including steatosis, inflammation, insulin resistance, and excess gluconeogenesis Cell-based experiments have shown that osteopontin impaired differentiation and insulin sensitivity of primary adipocytes as determined by inhibition PPAR-γ, adiponectin gene expression and insulin-stimulated glucose uptake Mice deficient in osteopontin have improved glucose tolerance and lower fasting plasma glucose, insulin, triglycerides, and proinflammatory cytokines after high fat diet regime compared to wild-type mice and antibody-mediated neutralization of osteopontin action reduces obesity-induced inflammation and insulin resistance In humans, serum osteopontin may reflect up-regulated gene expression during liver fibrosis in NAFLD and may serve as a test for advanced hepatic fibrosis in NAFLD Moreover, osteopontin is involved in the development of diabetic vascular complications , LCN2, a small secreted transport protein, was initially recognized for its role in innate immunity and was then identified as an adipokine capable of inducing insulin resistance More recently, a new insight on LCN2 was gained with the discovery of Lcn2 expression in mice being at least fold higher in bone than that in WAT Mechanistically, osteoblast-derived LCN2 has been shown to cross the blood-brain barrier and suppress appetite after binding to the melanocortin 4 receptor or MC4R in the hypothalamus by activating an MC4R-dependent anorexigenic appetite-suppressing pathway, thereby decreasing body weight and fat mass and improving insulin sensitivity Mice lacking Lcn 2 specifically in osteoblasts, rather than in adipocytes, showed increased food intake, fat mass, and body weight, along with decrease in glucose tolerance, insulin sensitivity, and serum insulin levels after glucose or arginine load Meanwhile, islet number and size, β-cell mass and proliferation, and insulin secretion were also decreased in the LCN lacking mice Conversely, chronic administration of exogenous LCN2 to lean and obese mice produced similar effects, with food intake, fat mass and body weight gain becoming reduced and glucose metabolism and energy expenditure becoming heightened In humans, postprandial serum levels of LCN2 become significantly increased in normal-weight individuals after high-fat meals, and this effect is accompanied by enhanced total energy expenditure; the effect is opposite decreased LCN2 in obese subjects Intriguingly, LCN2 expression and serum levels are higher in obesity , T2D , and NAFLD In addition, LCN2 levels were positively correlated with adiposity, hypertriglyceridemia, hyperglycemia, and insulin resistance index but negatively correlated with HDL cholesterol As such, there may be a compensatory mechanism at the early stage of this disease.

Sclerostin is a secreted protein predominantly expressed in osteocytes and is inhibited by osteoblast differentiation and bone formation Serum sclerostin levels were increased in mice models of disturbed metabolism, while sclerostin-deficient mice and those treated with a sclerostin-neutralizing antibody exhibited a reduction in the accumulation of WAT, along with corresponding enhancements in glucose and fatty acid metabolism, and increased insulin sensitivity In contrast, recombinant sclerostin treatment was found to enhance de novo lipid synthesis and reduce both fatty acid oxidation and the expression of genes associated with fatty acid catabolism In humans, circulating levels of sclerostin are increased in T2D and positively associated with BMI and fat mass Moreover, the serum sclerostin levels exhibit a positive correlation with fasting glucose and result in insulin resistance; but negatively correlated with whole-body glucose disposal and insulin clearance rate FGF23, a unique member of the FGF family, is derived from bone that acts as a hormone and regulates renal phosphate and vitamin D metabolism A growing body of epidemiological and experimental evidence suggests that FGF23 may regulate lipid and glucose metabolism as well as insulin action , but the underlying mechanisms are unclear.

Furthermore, FGF23 involvement has been implicated in the onset and progression of atherosclerosis via its effects on endothelial cell function FGF23 knockout mice presented with reduced fat mass, developed hypoglycemia and increased peripheral insulin sensitivity, and showed improved subcutaneous glucose tolerance , suggesting a link between FGF23 and insulin resistance.

However, another study in FGF23 lacking mice demonstrated no influence on aging, glucose homeostasis, or lipid metabolism with a non-functioning vitamin D receptor , suggesting that FGF23 may exert its effects depend on functioning vitamin D receptor.

In humans, serum FGF23 levels are elevated in individuals with obesity , MetS , prediabetes , and T2D Moreover, FGF23 is associated positively with triglycerides, BMI, waist circumference, and fat mass, and negatively correlated with HDL and apolipoprotein A1 However, another cross-sectional study of small sample did not show differences in circulating FGF23 levels between diabetic and non-diabetic patients, but reported that circulating FGF23 is associated with bone mineral density and preclinical vascular disease in T2D patients Further experimental studies are needed to shed more light on the underlying mechanisms between FGF23 and glucose and lipid metabolism, and prospective studies of large scale are needed to determine the association between FGF23 and metabolic disease, such as T2D.

Overnutrition and physical activity alter cytokines secretion, thereby influencing metabolic and immune regulatory pathways that caused or promoted metabolic disorders. These cytokines are part of a complex network that mediates communication between multiple organs and tissues e.

The emerging data support the contributions of certain cytokines to metabolic disorders. Given the disease-related changes in levels of relevant cytokines for instance, leptin, adiponectin, reisitin, FGF21, Fetuin A, TNF-α, IL-6, MCP-1 , these factors may serve as biomarkers for the early detection of metabolic disorders.

Moreover, based on preclinical studies, certain cytokines FGF21, leptin, adiponectin, irisin that can induce improvements in glucose and lipid metabolism and may emerge as novel targets of broader and more efficacious treatments and prevention of metabolic disease.

ZY, QS, and JF contributed conception of the paper. JS wrote the manuscript. ZY and QS revised the manuscript. This work was funded by the grants from the National Natural Science Foundation of China , and Shanghai Health System Outstanding Young Talents Training Program XYQ The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

T2D, type 2 diabetes; NAFLD, non-alcoholic fatty liver disease; MetS, metabolic syndrome; CNS, central nervous system; FFA, free fatty acids; BMI, body mass index; HDL, high-density lipoprotein; VLDL, very low-density lipoprotein; HFD, high-fat diet; WAT, white adipose tissue; FGF21, fibroblast growth factor 21; IRS1, insulin receptor substrate 1; TNF-α, tumor necrosis factor α; IL, Interleukin; MCP-1, monocyte chemotactic protein 1; NASH, non-alcoholic steatohepatitis; SREBPC, sterol regulatory element-binding protein C; ApoB, apolipoprotein B; BDNF, brain-derived neurotrophic factor; ucOC, undercarboxylated osteocalcin; LCN2, Lipocalin 2; PPAR, peroxisome proliferators-activated receptors; PKA, protein kinase A; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; GLUT, glucose transporter.

Meex RCR, Watt MJ. Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance. Nat Rev Endocrinol. doi: PubMed Abstract CrossRef Full Text Google Scholar. Cao H. Adipocytokines in obesity and metabolic disease.

J Endocrinol. Eckardt K, Gorgens SW, Raschke S, Eckel J. Myokines in insulin resistance and type 2 diabetes. Liu DM, Mosialou I, Liu JM. Bone: another potential target to treat, prevent and predict diabetes. Diabetes Obes Metab. Minokoshi Y, Kim YB, Peroni OD, Fryer LG, Muller C, Carling D, et al.

Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Morton GJ, Gelling RW, Niswender KD, Morrison CD, Rhodes CJ, Schwartz MW. Leptin regulates insulin sensitivity via phosphatidylinositolOH kinase signaling in mediobasal hypothalamic neurons.

Cell Metab. Unger RH, Zhou YT, Orci L. Regulation of fatty acid homeostasis in cells: novel role of leptin. Proc Natl Acad Sci USA.

Cummings BP, Bettaieb A, Graham JL, Stanhope KL, Dill R, Morton GJ, et al. Subcutaneous administration of leptin normalizes fasting plasma glucose in obese type 2 diabetic UCD-T2DM rats. Moon HS, Matarese G, Brennan AM, Chamberland JP, Liu X, Fiorenza CG, et al.

Efficacy of metreleptin in obese patients with type 2 diabetes: cellular and molecular pathways underlying leptin tolerance. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome.

J Clin Invest. Yoda-Murakami M, Taniguchi M, Takahashi K, Kawamata S, Saito K, Choi-Miura NH, et al. Biochem Biophys Res Commun. Heiker JT, Kosel D, Beck-Sickinger AG. Molecular mechanisms of signal transduction via adiponectin and adiponectin receptors. Biol Chem.

Yamauchi T, Kadowaki T. Adiponectin receptor as a key player in healthy longevity and obesity-related diseases. Spranger J, Kroke A, Mohlig M, Bergmann MM, Ristow M, Boeing H, et al.

Adiponectin and protection against type 2 diabetes mellitus. Tsuchida A, Yamauchi T, Ito Y, Hada Y, Maki T, Takekawa S, et al. J Biol Chem. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, et al.

The hormone resistin links obesity to diabetes. Qi Y, Nie Z, Lee YS, Singhal NS, Scherer PE, Lazar MA, et al. Loss of resistin improves glucose homeostasis in leptin deficiency. Singhal NS, Patel RT, Qi Y, Lee YS, Ahima RS. Loss of resistin ameliorates hyperlipidemia and hepatic steatosis in leptin-deficient mice.

Am J Physiol Endocrinol Metab. Gierej P, Gierej B, Kalinowski P, Wroblewski T, Paluszkiewicz R, Kobryn K, et al. Expression of resistin in the liver of patients with non-alcoholic fatty liver disease.

Pol J Pathol. McTernan CL, McTernan PG, Harte AL, Levick PL, Barnett AH, Kumar S. Resistin, central obesity, and type 2 diabetes. Ferland DJ, Watts SW. Chemerin: a comprehensive review elucidating the need for cardiovascular research.

Pharmacol Res. Bozaoglu K, Bolton K, McMillan J, Zimmet P, Jowett J, Collier G, et al. Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Krautbauer S, Wanninger J, Eisinger K, Hader Y, Beck M, Kopp A, et al.

Chemerin is highly expressed in hepatocytes and is induced in non-alcoholic steatohepatitis liver. Exp Mol Pathol. Ernst MC, Issa M, Goralski KB, Sinal CJ. Chemerin exacerbates glucose intolerance in mouse models of obesity and diabetes. Roh SG, Song SH, Choi KC, Katoh K, Wittamer V, Parmentier M, et al.

Chemerin—a new adipokine that modulates adipogenesis via its own receptor. Ernst MC, Haidl ID, Zuniga LA, Dranse HJ, Rourke JL, Zabel BA, et al. Disruption of the chemokine-like receptor-1 CMKLR1 gene is associated with reduced adiposity and glucose intolerance.

Neves KB, Nguyen Dinh Cat A, Alves-Lopes R, Harvey KY, Costa RMD, Lobato NS, et al. Chemerin receptor blockade improves vascular function in diabetic obese mice via redox-sensitive and Akt-dependent pathways. Am J Physiol Heart Circ Physiol. Takahashi M, Okimura Y, Iguchi G, Nishizawa H, Yamamoto M, Suda K, et al.

Chemerin regulates beta-cell function in mice. Sci Rep. Kukla M, Zwirska-Korczala K, Hartleb M, Waluga M, Chwist A, Kajor M, et al. Serum chemerin and vaspin in non-alcoholic fatty liver disease. Scand J Gastroenterol. Weigert J, Neumeier M, Wanninger J, Filarsky M, Bauer S, Wiest R, et al.

Systemic chemerin is related to inflammation rather than obesity in type 2 diabetes. Clin Endocrinol. Lehrke M, Becker A, Greif M, Stark R, Laubender RP, von Ziegler F, et al. Chemerin is associated with markers of inflammation and components of the metabolic syndrome but does not predict coronary atherosclerosis.

Eur J Endocrinol. CrossRef Full Text Google Scholar. Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, et al. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action.

Watanabe T, Watanabe-Kominato K, Takahashi Y, Kojima M, Watanabe R. Adipose Tissue-Derived Omentin-1 function and regulation.

Compr Physiol. Jialal I, Devaraj S, Kaur H, Adams-Huet B, Bremer AA. Increased chemerin and decreased omentin-1 in both adipose tissue and plasma in nascent metabolic syndrome. J Clin Endocrinol Metab. Tilg H, Moschen AR, Roden M. NAFLD and diabetes mellitus. Nat Rev Gastroenterol Hepatol.

Meex RC, Hoy AJ, Morris A, Brown RD, Lo JC, Burke M, et al. Fetuin B is a secreted hepatocyte factor linking steatosis to impaired glucose metabolism. Stefan N, Haring HU. The role of hepatokines in metabolism.

Fon Tacer K, Bookout AL, Ding X, Kurosu H, John GB, Wang L, et al. Research resource: Comprehensive expression atlas of the fibroblast growth factor system in adult mouse. Mol Endocrinol. Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, et al.

FGF as a novel metabolic regulator. Arner P, Pettersson A, Mitchell PJ, Dunbar JD, Kharitonenkov A, Ryden M. FGF21 attenuates lipolysis in human adipocytes - a possible link to improved insulin sensitivity.

FEBS Lett. Zhang X, Yeung DC, Karpisek M, Stejskal D, Zhou ZG, Liu F, et al. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Mutanen A, Heikkila P, Lohi J, Raivio T, Jalanko H, Pakarinen MP.

Serum FGF21 increases with hepatic fat accumulation in pediatric onset intestinal failure. J Hepatol. Lin Z, Tian H, Lam KS, Lin S, Hoo RC, Konishi M, et al.

Adiponectin mediates the metabolic effects of FGF21 on glucose homeostasis and insulin sensitivity in mice. Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, et al.

The effects of LY, an FGF21 analog, in obese human subjects with type 2 diabetes. Gao M, Zhan YQ, Yu M, Ge CH, Li CY, Zhang JH, et al.

Cell Signal. Wu HT, Ou HY, Hung HC, Su YC, Lu FH, Wu JS, et al. A novel hepatokine, HFREP1, plays a crucial role in the development of insulin resistance and type 2 diabetes.

Wu HT, Lu FH, Ou HY, Su YC, Hung HC, Wu JS, et al. The role of hepassocin in the development of non-alcoholic fatty liver disease. Auberger P, Falquerho L, Contreres JO, Pages G, Le Cam G, Rossi B, et al.

Characterization of a natural inhibitor of the insulin receptor tyrosine kinase: cDNA cloning, purification, and anti-mitogenic activity. Stefan N, Fritsche A, Weikert C, Boeing H, Joost HG, Haring HU, et al. Plasma fetuin-A levels and the risk of type 2 diabetes. Mukhopadhyay S, Bhattacharya S.

Plasma fetuin-A triggers inflammatory changes in macrophages and adipocytes by acting as an adaptor protein between NEFA and TLR Pal D, Dasgupta S, Kundu R, Maitra S, Das G, Mukhopadhyay S, et al.

Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med. Haukeland JW, Dahl TB, Yndestad A, Gladhaug IP, Loberg EM, Haaland T, et al.

Fetuin A in nonalcoholic fatty liver disease: in vivo and in vitro studies. Mathews ST, Singh GP, Ranalletta M, Cintron VJ, Qiang X, Goustin AS, et al. Improved insulin sensitivity and resistance to weight gain in mice null for the Ahsg gene.

de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, et al. Omentin plasma levels and gene expression are decreased in obesity. Reinehr T, Roth CL. Fetuin-A and its relation to metabolic syndrome and fatty liver disease in obese children before and after weight loss.

Circulating fetuin-A and free fatty acids interact to predict insulin resistance in humans. Olivier E, Soury E, Ruminy P, Husson A, Parmentier F, Daveau M, et al. Fetuin-B, a second member of the fetuin family in mammals. Biochem J. Walker GE, Follenzi A, Bruscaggin V, Manfredi M, Bellone S, Marengo E, et al.

Fetuin B links vitamin D deficiency and pediatric obesity: direct negative regulation by vitamin D. J Steroid Biochem Mol Biol. Motsenbocker MA, Tappel AL. A selenocysteine-containing selenium-transport protein in rat plasma. Biochim Biophys Acta. Dinarello CA.

Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start.

Nat Rev Drug Discov. Sethi JK, Hotamisligil GS. The role of TNF alpha in adipocyte metabolism. Semin Cell Dev Biol. Xu H, Uysal KT, Becherer JD, Arner P, Hotamisligil GS. Altered tumor necrosis factor-alpha TNF-alpha processing in adipocytes and increased expression of transmembrane TNF-alpha in obesity.

Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Hu FB, Meigs JB, Li TY, Rifai N, Manson JE. Inflammatory markers and risk of developing type 2 diabetes in women. Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes.

Trends Endocrinol Metab. Lang CH, Dobrescu C, Bagby GJ. Tumor necrosis factor impairs insulin action on peripheral glucose disposal and hepatic glucose output. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol.

Liu C, Feng X, Li Q, Wang Y, Li Q, Hua M. Adiponectin, TNF-alpha and inflammatory cytokines and risk of type 2 diabetes: a systematic review and meta-analysis. Butcher MJ, Hallinger D, Garcia E, Machida Y, Chakrabarti S, Nadler J, et al.

Association of proinflammatory cytokines and islet resident leucocytes with islet dysfunction in type 2 diabetes. Olson NC, Callas PW, Hanley AJ, Festa A, Haffner SM, Wagenknecht LE, et al. Circulating levels of TNF-alpha are associated with impaired glucose tolerance, increased insulin resistance, and ethnicity: the Insulin Resistance Atherosclerosis Study.

Banerjee M, Saxena M. Interleukin-1 IL-1 family of cytokines: role in type 2 diabetes. Clin Chim Acta.

Major CD, Wolf BA. Interleukin-1beta stimulation of c-Jun NH 2 -terminal kinase activity in insulin-secreting cells: evidence for cytoplasmic restriction.

Ammendrup A, Maillard A, Nielsen K, Aabenhus Andersen N, Serup P, Dragsbaek Madsen O, et al. The c-Jun amino-terminal kinase pathway is preferentially activated by interleukin-1 and controls apoptosis in differentiating pancreatic beta-cells. Kaneto H, Xu G, Fujii N, Kim S, Bonner-Weir S, Weir GC.

Involvement of c-Jun N-terminal kinase in oxidative stress-mediated suppression of insulin gene expression. Jager J, Gremeaux T, Cormont M, Le Marchand-Brustel Y, Tanti JF. Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression.

Miura K, Kodama Y, Inokuchi S, Schnabl B, Aoyama T, Ohnishi H, et al. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Tilg H, Moschen AR, Szabo G. Kirii H, Niwa T, Yamada Y, Wada H, Saito K, Iwakura Y, et al.

Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. Osborn O, Brownell SE, Sanchez-Alavez M, Salomon D, Gram H, Bartfai T. Treatment with an Interleukin 1 beta antibody improves glycemic control in diet-induced obesity.

Salmenniemi U, Ruotsalainen E, Pihlajamaki J, Vauhkonen I, Kainulainen S, Punnonen K, et al. Multiple abnormalities in glucose and energy metabolism and coordinated changes in levels of adiponectin, cytokines, and adhesion molecules in subjects with metabolic syndrome.

Moschen AR, Molnar C, Enrich B, Geiger S, Ebenbichler CF, Tilg H. Adipose and liver expression of interleukin IL -1 family members in morbid obesity and effects of weight loss.

Mol Med. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukinreceptor antagonist in type 2 diabetes mellitus.

N Engl J Med. Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol. Rose-John S. IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL Int J Biol Sci.

Schaper F, Rose-John S. Interleukin Biology, signaling and strategies of blockade. Cytokine Growth Factor Rev. Kraakman MJ, Kammoun HL, Allen TL, Deswaerte V, Henstridge DC, Estevez E, et al.

Blocking IL-6 trans-signaling prevents high-fat diet-induced adipose tissue macrophage recruitment but does not improve insulin resistance. Rotter V, Nagaev I, Smith U.

Interleukin-6 IL-6 induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. Timper K, Denson JL, Steculorum SM, Heilinger C, Engstrom-Ruud L, Wunderlich CM, et al. IL-6 improves energy and glucose homeostasis in obesity via enhanced central IL-6 trans-signaling.

Cell Rep. Stouthard JM, Oude Elferink RP, Sauerwein HP. Interleukin-6 enhances glucose transport in 3T3-L1 adipocytes. Lagathu C, Bastard JP, Auclair M, Maachi M, Capeau J, Caron M.

Chronic interleukin-6 IL-6 treatment increased IL-6 secretion and induced insulin resistance in adipocyte: prevention by rosiglitazone. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM.

C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. Park J, Ryu DR, Li JJ, Jung DS, Kwak SJ, Lee SH, et al. Am J Physiol Renal Physiol. Sung FL, Zhu TY, Au-Yeung KK, Siow YL.

Kidney Int B. Chen J, Guo Y, Zeng W, Huang L, Pang Q, Nie L, et al. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity.

Nio Y, Yamauchi T, Iwabu M, Okada-Iwabu M, Funata M, Yamaguchi M, et al. Monocyte chemoattractant protein-1 MCP-1 deficiency enhances alternatively activated M2 macrophages and ameliorates insulin resistance and fatty liver in lipoatrophic diabetic A-ZIP transgenic mice.

Ahmed SF, Shabayek MI, Abdel Ghany ME, El-Hefnawy MH, El-Mesallamy HO Role of CTRP3 CTRP9 and MCP-1 for the evaluation of T2DM associated coronary artery disease in Egyptian postmenopausal females. PLoS ONE. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ.

Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis.

Roca-Rivada A, Castelao C, Senin LL, Landrove MO, Baltar J, Belen Crujeiras A, et al. Huh JY, Mougios V, Kabasakalis A, Fatouros I, Siopi A, Douroudos II, et al.

Exercise-induced irisin secretion is independent of age or fitness level and increased irisin may directly modulate muscle metabolism through AMPK activation. Xiong XQ, Geng Z, Zhou B, Zhang F, Han Y, Zhou YB, et al. FNDC5 attenuates adipose tissue inflammation and insulin resistance via AMPK-mediated macrophage polarization in obesity.

Seale P, Conroe HM, Estall J, Kajimura S, Frontini A, Ishibashi J, et al. Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice. This results in decreased fatty acid oxidation. Elevated glucose, in turn, activates ChREBP and SREBP-1c, and causes increased pancreatic insulin secretion.

SREBP-1c blocks hepatic IRS2 signalling, further promoting hepatic glucose production, and probably counteracting the suppressive effect of SREBP-1c on gluconeogenic genes.

Insulin, ChREBP and SREBP-1c induce FASN and ACAC, leading to increased production of fatty acids. Therefore hepatic triglycerides accumulate as a result of both increased fatty acid production and reduced fatty acid oxidation in insulin-resistant states.

The red arrows indicate the direction of changes in insulin-resistant states. A network of transcription factors, co-repressors and co-activators act as sensors of hormonal and nutritional status in order to co-ordinate enzyme activities and metabolic pathways in the liver.

The factors discussed in the following section are directly or indirectly linked at the molecular level and have recently been shown to play key roles in both hepatic glucose and lipid metabolism Fig.

Model of interactions between key factors involved in hepatic glucose and lipid metabolism. LXRα and PPARα interact or compete with each other by forming obligate heterodimers with RXRα, thereby reciprocally reducing other pairings [ 40 , 41 ].

Activated LXRα induces SREBP-1c and blocks 11β-HSD1, thereby interfering with the inhibitory effect of cortisol on β-oxidation, and increasing synthesis of fatty acids via SREBP-1c. LXRα, PPARα, and AMPK activate β-oxidation of fatty acids. Cortisol and insulin inhibit fatty acid oxidation.

Induction of hepatic glucose production is indicated by red dots , suppression of hepatic glucose production by green dots. Liver X receptors LXRs are members of the nuclear receptor family, and are now recognised as important regulators of cholesterol metabolism, lipid biosynthesis and glucose homeostasis [ 5 ].

LXRs are also involved in regulating the storage and oxidation of dietary fat [ 31 ]. Two isoforms have been described, LXRα and LXRβ. The beta isoform is ubiquitously expressed and has recently been implicated in adipocyte growth, glucose homeostasis and beta cell function [ 32 ].

LXRα is highly expressed in the liver, adipose tissue, macrophages and the small intestine [ 33 ]. In hepatocytes, LXRα expression is induced by oxysterols and insulin [ 34 ], resulting in increased levels of lipogenic enzymes and the suppression of key gluconeogenic enzymes, including PEPCK and GPase [ 35 ].

LXRα also plays an important role in the regulation of glucocorticoid action, through inhibition of 11β-hydroxysteroid dehydrogenase type 1 11β- HSD1 [ 36 , 37 ].

LXRα forms obligate heterodimers with retinoid X receptors RXRs , which bind to LXR response elements in promoter regions of genes, resulting in the modulation of transcriptional processes, through interactions with co-activator or co-repressor complexes [ 38 ].

LXRα has been shown to strongly induce transcription of the gene for SREBP-1c via this mechanism [ 39 ], leading to the activation of lipogenic genes. It is thought that the transcription factor PPARα prevents the heterodimerisation of LXRα with RXR [ 40 ], and it follows that LXRα can antagonise the functions of PPARα [ 41 ].

A direct interaction between LXRα and PPARα has been proposed [ 42 ], which would be a logical way of preventing the simultaneous activation of the opposing pathways [ 43 ] Fig. LXR agonists have been proposed to act as glucose-lowering drugs. However, in mice, pharmacological LXR activation not only improves glucose metabolism, but also activates SREBP-1c and FASN, thereby causing severe hepatic steatosis [ 44 ], which, in turn, may have unfavourable effects on insulin sensitivity.

Despite this, steatosis associated with LXR activation does not necessarily affect insulin-mediated suppression of hepatic glucose production. Indeed, glucose metabolism may even improve, as a result of increased peripheral glucose uptake and metabolism [ 45 ], maybe in response to potential counteracting glucose-lowering effects of LXR agonists or changes in hepatic fatty acid profile upon LXR activation.

Changes in LXR-induced hepatic fatty acid profiles might be explained by enhanced transcription of the gene encoding stearoyl-CoA desaturase-1, which is involved in the conversion of saturated fatty acid into monounsaturated fatty acids [ 46 ]. Furthermore, an increased intake of dietary monounsaturated fatty acids improves insulin sensitivity in healthy humans [ 47 ].

Thus, hepatic accumulation of triglycerides, which are relatively rich in monounsaturated fatty acids, might be less deleterious than liver fat containing predominantly saturated fatty acids. Moreover, treatment with an LXRα agonist induces peroxisomal β-oxidation in PPARα-null mice, indicating an alternative PPARα-independent pathway that might serve as a counter-regulatory mechanism against LXR agonist-induced liver steatosis [ 48 ].

However, compared with PPARα-induced β-oxidation, this effect is modest. These mice are incapable of de novo fatty acid synthesis in the liver, leading to the development of hypoglycaemia and fatty liver in response to a zero-fat diet, which is reversed with dietary fat or treatment with a PPARα agonist.

The results showed that dietary fat or fatty acid pools newly synthesised by hepatic FASN new fat activated PPARα, leading to increased fatty acid oxidation, whereas pre-existing stored fat released from adipose tissue into the circulation old fat did not. Based on these findings, compartmentalisation of fatty acid metabolism in hepatocytes has been suggested [ 50 ].

PPARs are ligand-activated transcription factors that play an important role in adipocyte differentiation and fatty acid catabolism. Three subtypes α, δ [also known as β], γ have been identified and show tissue-specific expression [ 51 ].

PPARγ is only expressed at very low levels in the healthy liver, but levels are markedly increased in rodents with fatty liver and insulin resistance [ 52 ]. To date, it is unknown whether this phenomenon also exists in humans. PPARδ is ubiquitously expressed and is currently the least well understood of the PPAR subtypes.

It has been shown to modulate the inflammatory status of foam cells in atherosclerotic lesions and to be involved in muscle lipid metabolism in mice [ 53 , 54 ]. PPARα, the predominantly expressed form in the liver [ 55 , 56 ], is involved in promoting gluconeogenesis [ 57 , 58 ] and stimulates the transcription of genes that are critical for peroxisomal and mitochondrial oxidation of fatty acids [ 55 ].

By modulating gene expression, PPARα stimulates hepatic fatty acid oxidation to supply substrates that can be metabolised by other tissues. Supporting evidence is provided by the finding that PPARα-null mice exposed to prolonged fasting develop fatty liver and hypoglycaemia [ 59 ].

The hepatic expression of PPARα is nutritionally regulated: fasting activates PPARα, and fasted PPARα-null mice develop hypoglycaemia, hypoketonaemia, hyperlipidaemia and hepatic steatosis [ 59 ].

As mentioned above, PPARα forms heterodimers with RXRα, which enhances its binding to peroxisome proliferator response elements in target genes [ 41 ]. Thus, PPARα activation can suppress the LXRα-SREBP-1c pathway by interfering with the formation of SREBP-1c-activating LXR-RXR heterodimers [ 40 ] Fig.

The fibrate class of peroxisome proliferators act as PPARα agonists, and decrease plasma triglycerides and increase HDL levels, presumably through induction of genes involved in fatty acid β-oxidation [ 60 ]. In addition, PPARα agonists cause favourable changes in the particle size and subclass distribution of lipoproteins [ 61 ].

PPARα agonists reduce Hsd11b1 mRNA in the liver after chronic treatment, likely by an indirect mechanism, whereas PPARγ agonists do not probably because of the very low expression of PPARγ in the liver [ 62 ].

The existence of a negative feedback loop between PPARα and the glucocorticoid receptor has been proposed. Furthermore, PPARα agonists have been shown to have an insulin-sensitising activity [ 63 ] and to offer protection from cardiovascular disease reviewed in [ 64 ].

In the rodent liver, PPARα agonists have been shown to promote hepatocarcinogenesis [ 65 ]. However, PPARα activation has not been found to have tumorigenic effects in human hepatocytes [ 66 ]. SREBPs are membrane-bound transcription factors; they have been identified in three forms in humans and rodents.

SREBP-1c and SREBP-2 are the predominant subtypes in the rodent and human liver [ 67 ]. The main role of SREBP-2 is cholesterol synthesis, whereas SREBP-1c activates a complete programme of hepatic fatty acid synthesis [ 68 ] and reciprocally inhibits the expression of the gene for PEPCK when carbohydrates are abundant [ 69 , 70 ].

Overexpression of the gene for SREBP-1c leads to fatty liver in mice [ 71 ]. The effect of insulin on expression of the SREBP-1c gene is opposed by glucagon [ 74 ]. Changes in the expression of integral membrane proteins have been shown to inhibit the proteolytic activation of SREBPs.

SREBP-1c is inhibited by activation of AMP-activated protein kinase AMPK , a major cellular regulator of lipid and glucose metabolism see below [ 75 ]. The promoter of the SREBP-1c gene contains a regulatory element for LXRα [ 76 ], which strongly induces its transcription [ 39 ].

In turn, activated SREBP-1c stimulates the transcription of genes involved in de novo lipogenesis, such as ACAC and FASN , and interacts with regulatory elements in the promoters of various insulin-regulated genes. This involves competitive inhibition of PPARγ coactivator 1α PGC-1α , a co-regulator that activates PEPCK promotor activity and gluconeogenesis [ 77 ].

This effect of SREBP-1c seems to work in concert with insulin to suppress PGC-1α [ 78 ]. However, elevated levels of SREBP-1c have also been shown to induce insulin resistance by inhibiting hepatic IRS2 signalling.

A reduction in IRS2 expression restricts FOXO1 to the nucleus, leading to sustained gluconeogenesis [ 79 ]. This, in turn, may activate SREBP-1c, ChREBP and lipogenic enzymes, leading to triglyceride accumulation in the liver.

In addition to insulin, glucose has been shown to stimulate expression of the gene for SREBP-1c in a mouse hepatocyte cell line [ 80 ]. Exposure of insulin-depleted mice, generated by streptozotocin STZ administration, to fasting-refeeding protocols with glucose, fructose or sucrose showed markedly increased SREBP-1c on all carbohydrate diets.

Thus, nutritional regulation of SREBP-1c and lipogenic genes might even be independent of insulin, provided sufficient carbohydrates are available. Nutrients other than dietary carbohydrates have been shown to influence SREBP-1c. Polyunsaturated fatty acids PUFAs suppress the proteolytic release of SREBP-1 [ 82 ], independently of LXRα [ 83 ].

SREBP-1c plays a major role in the long-term control of glucose and lipid metabolism [ 3 ]. This indicates that other SREBP isoforms are able to partially compensate for the absence of SREBP-1c [ 84 ].

An alternative explanation may involve the transcription factor ChREBP, discussed in the next section, which has recently been shown to be regulated by factors similar to those involved in modulation of SREBP-1c e.

glucose, insulin, PUFA. The transcription factor ChREBP is translocated to the nucleus and activated in response to high glucose concentrations in the liver, independently of insulin.

As the name suggests, it was first identified by its ability to bind the carbohydrate-response element of the gene encoding liver pyruvate kinase L-PK. L-PK catalyses the conversion of phosphoenolpyruvate to pyruvate, which enters the Krebs cycle to generate citrate, the principal source of acetyl-CoA used for fatty acid synthesis [ 52 ].

Insulin indirectly regulates ChREBP through activation of glucokinase, which allows phosphorylated glucose to enter the pentose phosphate cycle, generating xylulose 5-phosphate and activating ChREBP via protein phosphatase 2A [ 3 ].

ChREBP has recently been shown to play a pivotal role in activating lipogenic genes [ 4 ]. ChREBP binds to its functional heterodimeric partner, Max-like protein X, and induces the transcription of lipogenic and glycolytic genes containing a carbohydrate response element, such as those encoding ACAC, FASN and L-PK [ 3 , 85 ].

Current knowledge on the molecular actions of ChREBP is limited. In adipocytes, the thiazolidinedione TZD troglitazone has been shown to induce ChREBP gene expression in a dose-dependent manner [ 86 ], which may contribute to the glucose-lowering effects of TZDs. TZDs are thought to improve insulin sensitivity in adipose tissue mainly by acting on PPARγ, which is not known to have a relevant function in the human liver, and prolonged treatment with TZDs has been shown to reduce liver fat content and liver volume [ 87 ].

Moreover, it is unknown whether TZDs induce hepatic ChREBP gene expression in humans. Potential favourable effects of ChREBP activation on glucose metabolism may, however, have the disadvantage of increased hepatic fat accumulation.

Polyunsaturated fatty acids PUFAs compromise the transcriptional activities of ChREBP by interfering with its translocation from the cytosol to the nucleus in response to glucose [ 88 ], independent of AMPK activation [ 89 ].

Thus, ChREBP is likely to be a major determinant of the inhibitory effect of PUFAs on both glycolytic and lipogenic genes [ 89 ]. The highest levels of expression have been found in the liver, gonads, adipose tissue and the brain [ 90 ].

In vivo, 11β-HSD1 converts inactive cortisone to active cortisol in humans or inactive dehydrocorticosterone to active corticosterone in rodents and 11β-HSD2 catalyses the reverse reaction. Glucocorticoids, PPAR-γ agonists and proinflammatory cytokines increase 11β-HSD1 activity.

Insulin has been shown to suppress expression of the gene for 11β-HSD1 Hsd11b1 in rat hepatocytes and hepatoma cells, while oestrogens, growth hormone and insulin reduce Hsd11b1 expression in the rodent liver [ 90 ]. In uncomplicated obesity, 11β-HSD1 activity has been proposed to be downregulated, probably as a compensatory mechanism to prevent insulin resistance.

This downregulation may be disturbed in type 2 diabetic patients, leading to insulin resistance and increased fat deposition in various organs, including the liver [ 92 ]. Glucocorticoids are essential factors involved in energy homeostasis, with cortisol being the principal active glucocorticoid in humans.

Glucocorticoids stimulate the transcription of glucogenic genes including those for PEPCK and GPase [ 70 , 93 ] , inhibit mitochondrial matrix acyl-CoA dehydrogenases and fatty acid β-oxidation, and may produce fatty liver in humans [ 94 ]. Antagonism of the receptor prevents obesity in rodents.

However, as already discussed, LXR agonists also activate SREBP-1c and thereby cause hepatic steatosis in mice, which may limit the therapeutic use of these agents. Another potential option to inhibit unfavourable glucocorticoid actions would be to block the glucocorticoid receptor [ 96 ].

However, long-term systemic therapy with glucocorticoid receptor antagonists may induce counter-regulatory mechanisms by activating the hypothalamic-pituitary-adrenal axis [ 97 ].

This phenomenon has also been suspected to appear in 11β-HSD1-null mice [ 98 ]. To circumvent these potential problems, highly selective inhibitors of 11β-HSD1 have recently been developed. Treatment of rodents with a non-steroidal selective inhibitor of 11β-HSD1 for 7 days significantly decreased both hepatic G-6P-ase and PEPCK mRNA as well as blood glucose and serum insulin concentrations [ 99 ].

AMP-activated protein kinase AMPK belongs to a family of highly conserved serine-threonine kinases and is present in various organs, including the liver [ ].

AMPK has a key role in the regulation of energy control as a metabolic sensor and regulator kinase. When activated, AMPK initiates a series of responses that are aimed at protecting the cell against ATP depletion, by stimulating fatty acid oxidation or glycolysis and inhibiting ATP-consuming anabolic pathways such as gluconeogenesis, protein and fatty acid synthesis [ ].

AMPK is phosphorylated and thereby activated by the protein—threonine kinase LKB1, which seems to be the major upstream AMPK-activating factor [ ]. Activation of AMPK results in inhibition of lipogenic factors such as SREBP-1c [ 75 ], FASN, ACAC [ ] and ChREBP [ ].

Induction of AMPK in hepatoma cells also decreases PEPCK and GPase transcription, likely in an insulin-independent manner [ ]. AMPK is not known to be activated by insulin, raising the possibility that insulin and AMPK regulate PEPCK by different and, perhaps, converging pathways [ 70 ].

AMPK may prevent insulin resistance in part by inhibiting factors that antagonise insulin signalling [ ]. Deletion of liver LKB1 in mice results in a near complete loss of AMPK activity, leading to lipogenic gene expression.

Increased gluconeogenesis in these mice could be explained by the observed nuclear translocation of TORC2, which transcriptionally coactivates cAMP-response-element-binding protein CREB , leading to increased expression of PGC-1α, thereby driving the expression of glucogenic genes [ ].

A number of hormonal and nutritionally regulated factors have been proposed to be involved in the regulation of hepatic AMPK activity. The adipocyte-derived hormone adiponectin has been shown to activate AMPK both in liver and skeletal muscle and to reduce hepatic glucose production and the expression of hepatic gluconeogenic genes, while increasing β-oxidation of fatty acids in the liver [ , ].

This may, at least in part, explain the positive associations between adiponectin and diabetes risk in epidemiological studies [ ].

Conversely, the orexigenic hormone ghrelin inhibits AMPK in the rat liver and in adipose tissue, while stimulating AMPK activity in the heart and hypothalamus [ ].

Ghrelin also decreases the effect of insulin on PEPCK in human hepatoma cells [ ], and may therefore contribute to the development of hepatic insulin resistance and lipid accumulation.

Other factors, such as adipocyte-secreted leptin or resistin which is mainly expressed in monocytes and macrophages in humans may be involved in the regulation of liver AMPK. Hypoglycaemic agents such as metformin and rosiglitazone have been proposed to indirectly activate AMPK, probably by interference with the respiratory chain [ ].

AMPK has been shown to be activated by metformin in skeletal muscle in vitro and in vivo reviewed in [ ] and by pioglitazone in cultured cells and in rat liver and adipose tissue in vivo [ ]. Interestingly, metformin action was turned off in the absence of LKB1, pointing to AMPK activation as an essential part of metformin action, at least in the mouse liver [ ].

However, thiazolidinediones [ ] and metformin [ ] exert various other molecular actions, which are not well understood at present. Moreover, chronic pharmacological activation of AMPK may lead to inhibition of insulin release, and may have unfavourable effects on cell proliferation and glycogen storage in cardiomyocytes reviewed in [ ].

Fasting has been shown to increase hypothalamic AMPK activity, while refeeding inhibits it, with reduced hypothalamic AMPK activity exerting anorexigenic effects [ , ]. Thus, non-selective AMPK activation may not be favourable in the context of body weight regulation.

The studies discussed in this review indicate that hepatic fat accumulation, insulin resistance and disturbed glucose metabolism are inter-related at a molecular level. In insulin-resistant hyperglycaemic states the suppressive effects of insulin and glucose on hepatic glucose production is reduced, whereas undamped hepatic lipogenesis and non-insulin dependent glucose transport to the liver both contribute to hepatic lipid accumulation, which in turn may further deteriorate insulin signalling.

Multiple organs, such as skeletal muscle, adipose tissue and the liver, are affected by insulin-resistant states and there has been considerable progress in identifying molecular pathways and potential factors involved.

Skeletal muscle and fat tissue are relatively easily accessible for biopsy in humans. However, despite many similarities between molecular pathways in different tissues, there are also important differences.

Thus, results obtained in other tissues cannot necessarily be transferred to the liver. Elucidating molecular pathways in human liver is more problematic due to the potential hazards involved in performing liver biopsies.

It is therefore important to note that most studies that have investigated molecular pathways in the liver were performed in animal models or in vitro. Because molecular functions differ even between rodent species [ ], it should be noted that, whilst results obtained in animal models provide valuable insights, they cannot necessarily be extrapolated to other species.

Biochem Soc Trans — Article PubMed 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.

Biochem J — Dentin R, Girard J, Postic C Carbohydrate responsive element binding protein ChREBP and sterol regulatory element binding protein-1c SREBP-1c : two key regulators of glucose metabolism and lipid synthesis in liver. Biochimie — Ishii S, Iizuka K, Miller BC, Uyeda K Carbohydrate response element binding protein directly promotes lipogenic enzyme gene transcription.

Proc Natl Acad Sci USA — Steffensen KR, Gustafsson JA Putative metabolic effects of the liver X receptor LXR. Diabetes 53 Suppl 1 :S36—S PubMed CAS Google Scholar. Marchesini G, Brizi M, Bianchi G et al Nonalcoholic fatty liver disease: a feature of the metabolic syndrome.

Diabetes — Angulo P Nonalcoholic fatty liver disease. N Engl J Med — Seppala-Lindroos A, Vehkavaara S, Hakkinen AM et al Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men.

J Clin Endocrinol Metab — Sutinen J, Hakkinen AM, Westerbacka J et al Rosiglitazone in the treatment of HAART-associated lipodystrophy—a randomized double-blind placebo-controlled study. Antivir Ther — Gavrilova O, Marcus-Samuels B, Graham D et al Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice.

J Clin Invest — J Biol Chem — Samuel VT, Liu ZX, Qu X et al Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease.

Petersen KF, Dufour S, Befroy D et al Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes.

Gastaldelli A, Toschi E, Pettiti M et al Effect of physiological hyperinsulinemia on gluconeogenesis in nondiabetic subjects and in type 2 diabetic patients.

Boden G Effects of free fatty acids on gluconeogenesis and glycogenolysis. Life Sci — Roden M, Bernroider E Hepatic glucose metabolism in humans—its role in health and disease.

Best Pract Res Clin Endocrinol Metab — Nordlie RC, Foster JD, Lange AJ Regulation of glucose production by the liver. Annu Rev Nutr — Pilkis SJ, Granner DK Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis. Annu Rev Physiol — Lam TK, Gutierrez-Juarez R, Pocai A, Rossetti L Regulation of blood glucose by hypothalamic pyruvate metabolism.

Science — Pocai A, Obici S, Schwartz GJ, Rossetti L A brain—liver circuit regulates glucose homeostasis. Cell Metab — Horton JD, Shah NA, Warrington JA et al Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes.

Randle PJ Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes Metab Rev — Ruderman NB, Saha AK, Vavvas D, Witters LA Malonyl-CoA, fuel sensing, and insulin resistance.

Am J Physiol E1—E McGarry JD Banting lecture dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Vaulont S, Vasseur-Cognet M, Kahn A Glucose regulation of gene transcription.

Koo SH, Dutcher AK, Towle HC Glucose and insulin function through two distinct transcription factors to stimulate expression of lipogenic enzyme genes in liver.

Mol Cell — Najjar SM, Yang Y, Fernstrom MA et al Insulin acutely decreases hepatic fatty acid synthase activity.

Wolfrum C, Asilmaz E, Luca E, Friedman JM, Stoffel M Foxa2 regulates lipid metabolism and ketogenesis in the liver during fasting and in diabetes. Nature — Kalaany NY, Gauthier KC, Zavacki AM et al LXRs regulate the balance between fat storage and oxidation. Gerin I, Dolinsky VW, Shackman JG et al LXRbeta is required for adipocyte growth, glucose homeostasis, and beta cell function.

Repa JJ, Mangelsdorf DJ The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol — Tobin KA, Ulven SM, Schuster GU et al Liver X receptors as insulin-mediating factors in fatty acid and cholesterol biosynthesis. Cao G, Liang Y, Broderick CL et al Antidiabetic action of a liver x receptor agonist mediated by inhibition of hepatic gluconeogenesis.

Stulnig TM, Oppermann U, Steffensen KR, Schuster GU, Gustafsson JA Liver X receptors downregulate 11beta-hydroxysteroid dehydrogenase type 1 expression and activity.

Stulnig TM, Steffensen KR, Gao H et al Novel roles of liver X receptors exposed by gene expression profiling in liver and adipose tissue. Mol Pharmacol — Glass CK Differential recognition of target genes by nuclear receptor monomers, dimers, and heterodimers.

Endocr Rev — Chen G, Liang G, Ou J, Goldstein JL, Brown MS Central role for liver X receptor in insulin-mediated activation of Srebp-1c transcription and stimulation of fatty acid synthesis in liver.

Yoshikawa T, Ide T, Shimano H et al Cross-talk between peroxisome proliferator-activated receptor PPAR alpha and liver X receptor LXR in nutritional regulation of fatty acid metabolism. PPARs suppress sterol regulatory element binding protein-1c promoter through inhibition of LXR signaling.

Mol Endocrinol — Ide T, Shimano H, Yoshikawa T et al Cross-talk between peroxisome proliferator-activated receptor PPAR alpha and liver X receptor LXR in nutritional regulation of fatty acid metabolism.

LXRs suppress lipid degradation gene promoters through inhibition of PPAR signaling. Miyata KS, McCaw SE, Patel HV, Rachubinski RA, Capone JP The orphan nuclear hormone receptor LXR alpha interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling.

Anderson SP, Dunn C, Laughter A, et al Overlapping transcriptional programs regulated by the nuclear receptors peroxisome proliferator-activated receptor alpha, retinoid X receptor, and liver X receptor in mouse liver.

Grefhorst A, Elzinga BM, Voshol PJ et al Stimulation of lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles. Am J Physiol Endocrinol Metab E—E Miyazaki M, Ntambi JM Role of stearoyl-coenzyme A desaturase in lipid metabolism.

Prostaglandins Leukot Essent Fatty Acids — Vessby B, Unsitupa M, Hermansen K et al Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy men and women: The KANWU Study.

Diabetologia — Hu T, Foxworthy P, Siesky A et al Hepatic peroxisomal fatty acid beta oxidation is regulated by liver X receptor alpha. Endocrinology — Gibbons G Old fat, make way for new fat. Nat Med — Lemberger T, Braissant O, Juge-Aubry C et al PPAR tissue distribution and interactions with other hormone-signaling pathways.

Ann N Y Acad Sci — Browning JD, Horton JD Molecular mediators of hepatic steatosis and liver injury. Lee CH, Chawla A, Urbiztondo N et al Transcriptional repression of atherogenic inflammation: modulation by PPARdelta. Tanaka T, Yamamoto J, Iwasaki S et al Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome.

Reddy JK, Hashimoto T Peroxisomal beta-oxidation and peroxisome proliferator-activated receptor alpha: an adaptive metabolic system. Vidal-Puig AJ, Considine RV, Jimenez-Linan M et al Peroxisome proliferator-activated receptor gene expression in human tissues.

Effects of obesity, weight loss, and regulation by insulin and glucocorticoids. Bernal-Mizrachi C, Weng S, Feng C et al Dexamethasone induction of hypertension and diabetes is PPAR-alpha dependent in LDL receptor-null mice.

Koo SH, Satoh H, Herzig S et al PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB Kersten S, Seydoux J, Peters JM, et al Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting.

Linton MF, Fazio S Re-emergence of fibrates in the management of dyslipidemia and cardiovascular risk. Curr Atheroscler Rep — Bays H, Stein EA Pharmacotherapy for dyslipidaemia—current therapies and future agents.

Expert Opin Pharmacother — Hermanowski-Vosatka A, Gerhold D, Mundt SS et al PPARalpha agonists reduce 11beta-hydroxysteroid dehydrogenase type 1 in the liver. Biochem Biophys Res Commun — Guerre-Millo M, Gervois P, Raspe E et al Peroxisome proliferator-activated receptor alpha activators improve insulin sensitivity and reduce adiposity.

Staels B, Fruchart JC Therapeutic roles of peroxisome proliferator-activated receptor agonists. Rao MS, Reddy JK Hepatocarcinogenesis of peroxisome proliferators. Knight B, Yeap BB, Yeoh GC, Olynyk JK Inhibition of adult liver progenitor oval cell growth and viability by an agonist of the peroxisome proliferator activated receptor PPAR family member gamma, but not alpha or delta.

Carcinogenesis — Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells.

Horton JD, Goldstein JL, Brown MS SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Yamamoto T, Shimano H, Nakagawa Y et al SREBP-1 interacts with hepatocyte nuclear factor-4 alpha and interferes with PGC-1 recruitment to suppress hepatic gluconeogenic genes.

Chakravarty K, Cassuto H, Reshef L, Hanson RW Factors that control the tissue-specific transcription of the gene for phosphoenolpyruvate carboxykinase-C. Crit Rev Biochem Mol Biol — Shimano H, Horton JD, Shimomura I et al Isoform 1c of sterol regulatory element binding protein is less active than isoform 1a in livers of transgenic mice and in cultured cells.

Foretz M, Guichard C, Ferre P, Foufelle F Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes.

Ribaux PG, Iynedjian PB Analysis of the role of protein kinase B cAKT in insulin-dependent induction of glucokinase and sterol regulatory element-binding protein 1 SREBP1 mRNAs in hepatocytes. Mol Cell Biol — Zhou G, Myers R, Li Y et al Role of AMP-activated protein kinase in mechanism of metformin action.

Jump DB, Botolin D, Wang Y et al Fatty Acid regulation of hepatic gene transcription. J Nutr — Puigserver P, Rhee J, Donovan J et al Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction.

Barthel A, Schmoll D Novel concepts in insulin regulation of hepatic gluconeogenesis. Ide T, Shimano H, Yahagi N et al SREBPs suppress IRSmediated insulin signalling in the liver. Nat Cell Biol — Hasty AH, Shimano H, Yahagi N et al Sterol regulatory element-binding protein-1 is regulated by glucose at the transcriptional level.

Matsuzaka T, Shimano H, Yahagi N et al Insulin-independent induction of sterol regulatory element-binding protein-1c expression in the livers of streptozotocin-treated mice.

Metbolism Lipid metabolism and glucose utilization for visiting nature. You are Lipid metabolism and glucose utilization a browser version Mindful eating practices limited utiluzation for CSS. To obtain the best experience, we recommend you gpucose a more up to date browser or turn off compatibility mode in Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. The epidemic of type 2 diabetes and impaired glucose tolerance is one of the main causes of morbidity and mortality worldwide.

Video

Fats - biochemistry

Author: Kijas

0 thoughts on “Lipid metabolism and glucose utilization

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com