3,738
Views
98
CrossRef citations to date
0
Altmetric
Review Article

Biomarkers of insulin sensitivity and insulin resistance: Past, present and future

, &
Pages 180-190 | Received 01 Sep 2014, Accepted 16 Feb 2015, Published online: 04 Jun 2015

References

  • Kahn SE, Cooper ME, Del Prato S. Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future. Lancet 2014;383:1068–83
  • Rask-Madsen C, Kahn CR. Tissue-specific insulin signaling, metabolic syndrome, and cardiovascular disease. Arterioscler Thromb Vasc Biol 2012;32:2052–9
  • Tam CS, Xie W, Johnson WD, et al. Defining insulin resistance from hyperinsulinemic-euglycemic clamps. Diabetes Care 2012;35:1605–10
  • Matthews DR, Hosker JP, Rudenski AS, et al. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 1985;28:412–19
  • Katz A, Nambi SS, Mather K, et al. Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab 2000;85:2402–10
  • Echouffo-Tcheugui JB, Dieffenbach SD, Kengne AP. Added value of novel circulating and genetic biomarkers in type 2 diabetes prediction: a systematic review. Diabetes Res Clin Pract 2013;101:255–69
  • Brooks-Worrell B, Narla R, Palmer JP. Biomarkers and immune-modulating therapies for type 2 diabetes. Trends Immunol 2012;33:546–53
  • Bluher M. Adipokines – removing road blocks to obesity and diabetes therapy. Mol Metab 2014;3:230–40
  • Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol 2012;8:457–65
  • Stefan N, Haring HU. The role of hepatokines in metabolism. Nat Rev Endocrinol 2013;9:144–52
  • Knights AJ, Funnell AP, Pearson RC, et al. Adipokines and insulin action: a sensitive issue. Adipocyte 2014;3:88–96
  • Wang Y, Lam KS, Yau MH, Xu A. Post-translational modifications of adiponectin: mechanisms and functional implications. Biochem J 2008;409:623–33
  • Dadson K, Liu Y, Sweeney G. Adiponectin action: a combination of endocrine and autocrine/paracrine effects. Front Endocrinol 2011;2:62
  • Ye R, Scherer PE. Adiponectin, driver or passenger on the road to insulin sensitivity? Mol Metab 2013;2:133–41
  • Cheng KK, Lam KS, Wang B, Xu A. Signaling mechanisms underlying the insulin-sensitizing effects of adiponectin. Best Pract Res Clin Endocrinol Metab 2014;28:3–13
  • Combs TP, Marliss EB. Adiponectin signaling in the liver. Rev Endocrine Metab Disord 2014;15:137–47
  • Yamauchi T, Kadowaki T. Adiponectin receptor as a key player in healthy longevity and obesity-related diseases. Cell Metab 2013;17:185–96
  • Kadowaki T, Yamauchi T, Okada-Iwabu M, Iwabu M. Adiponectin and its receptors: implications for obesity-associated diseases and longevity. Lancet Diabetes Endocrinol 2014;2:8–9
  • Liu Y, Retnakaran R, Hanley A, et al. Total and high molecular weight but not trimeric or hexameric forms of adiponectin correlate with markers of the metabolic syndrome and liver injury in Thai subjects. J Clin Endocrinol Metab 2007;92:4313–18
  • Tschritter O, Fritsche A, Thamer C, et al. Plasma adiponectin concentrations predict insulin sensitivity of both glucose and lipid metabolism. Diabetes 2003;52:239–43
  • Li FY, Lam KS, Xu A. Therapeutic perspectives for adiponectin: an update. Curr Med Chem 2012;19:5513–23
  • Okada-Iwabu M, Yamauchi T, Iwabu M, et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature 2013;503:493–9
  • Qi Q, Yu Z, Ye X, et al. Elevated retinol-binding protein 4 levels are associated with metabolic syndrome in Chinese people. J Clin Endocrinol Metab 2007;92:4827–34
  • Cho YM, Youn BS, Lee H, et al. Plasma retinol-binding protein-4 concentrations are elevated in human subjects with impaired glucose tolerance and type 2 diabetes. Diabetes Care 2006;29:2457–61
  • Lim S, Yoon JW, Choi SH, et al. Combined impact of adiponectin and retinol-binding protein 4 on metabolic syndrome in elderly people: the Korean Longitudinal Study on Health and Aging. Obesity (Silver Spring) 2010;18:826–32
  • Alkharfy KM, Al-Daghri NM, Vanhoutte PM, et al. Serum retinol-binding protein 4 as a marker for cardiovascular disease in women. PLoS One 2012;7:e48612
  • Yang Q, Graham TE, Mody N, et al. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 2005;436:356–62
  • Abel ED, Peroni O, Kim JK, et al. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 2001;409:729–33
  • Ulgen F, Herder C, Kuhn MC, et al. Association of serum levels of retinol-binding protein 4 with male sex but not with insulin resistance in obese patients. Arch Physiol Biochem 2010;116:57–62
  • Promintzer M, Krebs M, Todoric J, et al. Insulin resistance is unrelated to circulating retinol binding protein and protein C inhibitor. J Clin Endocrinol Metab 2007;92:4306–12
  • Kotnik P, Fischer-Posovszky P, Wabitsch M. RBP4: a controversial adipokine. Eur J Endocrinol 2011;165:703–11
  • Park SE, Lee NS, Park JW, et al. Association of Urinary RBP4 with insulin resistance, inflammation, and microalbuminuria. Eur J Endocrinol 2014 ;171:443--9
  • Bonventre JV, Vaidya VS, Schmouder R, et al. Next-generation biomarkers for detecting kidney toxicity. Nat Biotechnol 2010;28:436–40
  • Norden AG, Lapsley M, Unwin RJ. Urine retinol-binding protein 4: a functional biomarker of the proximal renal tubule. Adv Clin Chem 2014;63:85–122
  • Roman AA, Parlee SD, Sinal CJ. Chemerin: a potential endocrine link between obesity and type 2 diabetes. Endocrine 2012;42:243–51
  • Zabel BA, Allen SJ, Kulig P, et al. Chemerin activation by serine proteases of the coagulation, fibrinolytic, and inflammatory cascades. J Biol Chem 2005;280:34661–6
  • Sell H, Laurencikiene J, Taube A, et al. Chemerin is a novel adipocyte-derived factor inducing insulin resistance in primary human skeletal muscle cells. Diabetes 2009;58:2731–40
  • Tonjes A, Fasshauer M, Kratzsch J, et al. Adipokine pattern in subjects with impaired fasting glucose and impaired glucose tolerance in comparison to normal glucose tolerance and diabetes. PLoS One 2010;5:e13911
  • Ouwens DM, Bekaert M, Lapauw B, et al. Chemerin as biomarker for insulin sensitivity in males without typical characteristics of metabolic syndrome. Arch Physiol Biochem 2012;118:135–8
  • Rourke JL, Dranse HJ, Sinal CJ. Towards an integrative approach to understanding the role of chemerin in human health and disease. Obes Rev 2013;14:245–62
  • Bozaoglu K, Segal D, Shields KA, et al. Chemerin is associated with metabolic syndrome phenotypes in a Mexican-American population. J Clin Endocrinol Metab 2009;94:3085–8
  • El-Mesallamy HO, El-Derany MO, Hamdy NM. Serum omentin-1 and chemerin levels are interrelated in patients with Type 2 diabetes mellitus with or without ischaemic heart disease. Diabet Med 2011;28:1194–200
  • Weigert J, Neumeier M, Wanninger J, et al. Systemic chemerin is related to inflammation rather than obesity in type 2 diabetes. Clin Endocrinol 2010;72:342–8
  • Takahashi M, Takahashi Y, Takahashi K, et al. Chemerin enhances insulin signaling and potentiates insulin-stimulated glucose uptake in 3T3-L1 adipocytes. FEBS Lett 2008;582:573–8
  • Kralisch S, Weise S, Sommer G, et al. Interleukin-1beta induces the novel adipokine chemerin in adipocytes in vitro. Regul Pept 2009;154:102–6
  • Bozaoglu K, Bolton K, McMillan J, et al. Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology 2007;148:4687–94
  • Rourke JL, Muruganandan S, Dranse HJ, et al. Gpr1 is an active chemerin receptor influencing glucose homeostasis in obese mice. J Endocrinol 2014;222:201–15
  • Makowski L, Hotamisligil GS. Fatty acid binding proteins-the evolutionary crossroads of inflammatory and metabolic responses. J Nutr 2004;134:2464S–8S
  • Chmurzynska A. The multigene family of fatty acid-binding proteins (FABPs): function, structure and polymorphism. J Appl Genet 2006;47:39–48
  • Cabre A, Lazaro I, Girona J, et al. Fatty acid binding protein 4 is increased in metabolic syndrome and with thiazolidinedione treatment in diabetic patients. Atherosclerosis 2007;195:e150–8
  • Kralisch S, Fasshauer M. Adipocyte fatty acid binding protein: a novel adipokine involved in the pathogenesis of metabolic and vascular disease? Diabetologia 2013;56:10–21
  • Lan H, Cheng CC, Kowalski TJ, et al. Small-molecule inhibitors of FABP4/5 ameliorate dyslipidemia but not insulin resistance in mice with diet-induced obesity. J Lipid Res 2011;52:646–56
  • Hui X, Li H, Zhou Z, et al. Adipocyte fatty acid-binding protein modulates inflammatory responses in macrophages through a positive feedback loop involving c-Jun NH2-terminal kinases and activator protein-1. J Biol Chem 2010;285:10273–80
  • Horakova D, Pastucha D, Stejskal D, et al. Adipocyte fatty acid binding protein and C-reactive protein levels as indicators of insulin resistance development. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2011;155:355–9
  • Stefanska A, Sypniewska G, Blaszkiewicz B, et al. Comparison between C-reactive protein and adipocyte fatty acid-binding protein as a component of metabolic syndrome in middle-aged women. Clin Biochem 2011;44:304–6
  • Xu A, Wang Y, Xu JY, et al. Adipocyte fatty acid-binding protein is a plasma biomarker closely associated with obesity and metabolic syndrome. Clin Chem 2006;52:405–13
  • Simon I, Escote X, Vilarrasa N, et al. Adipocyte fatty acid-binding protein as a determinant of insulin sensitivity in morbid-obese women. Obesity (Silver Spring) 2009;17:1124–8
  • Park SE, Rhee EJ, Lee WY, et al. The role of serum adipocyte fatty acid-binding protein on the development of metabolic syndrome is independent of pro-inflammatory cytokines. Nutr Metab Cardiovasc Dis 2012;22:525–32
  • Xu A, Tso AW, Cheung BM, et al. Circulating adipocyte-fatty acid binding protein levels predict the development of the metabolic syndrome: a 5-year prospective study. Circulation 2007;115:1537–43
  • Mohlig M, Weickert MO, Ghadamgadai E, et al. Adipocyte fatty acid-binding protein is associated with markers of obesity, but is an unlikely link between obesity, insulin resistance, and hyperandrogenism in polycystic ovary syndrome women. Eur J Endocrinol 2007;157:195–200
  • Engl J, Ciardi C, Tatarczyk T, et al. A-FABP – a biomarker associated with the metabolic syndrome and/or an indicator of weight change? Obesity (Silver Spring) 2008;16:1838–42
  • Reinehr T, Stoffel-Wagner B, Roth CL. Adipocyte fatty acid-binding protein in obese children before and after weight loss. Metab Clin Exp 2007;56:1735–41
  • Itoh N. FGF21 as a hepatokine, adipokine, and myokine in metabolism and diseases. Front Endocrinol 2014;5:107
  • Dostalova I, Haluzikova D, Haluzik M. Fibroblast growth factor 21: a novel metabolic regulator with potential therapeutic properties in obesity/type 2 diabetes mellitus. Physiol Res 2009;58:1–7
  • Woo YC, Xu A, Wang Y, Lam KS. Fibroblast growth factor 21 as an emerging metabolic regulator: clinical perspectives. Clin Endocrinol 2013;78:489–96
  • Iglesias P, Selgas R, Romero S, Diez JJ. Biological role, clinical significance, and therapeutic possibilities of the recently discovered metabolic hormone fibroblastic growth factor 21. Eur J Endocrinol 2012;167:301–9
  • Suzuki M, Uehara Y, Motomura-Matsuzaka K, et al. betaKlotho is required for fibroblast growth factor (FGF) 21 signaling through FGF receptor (FGFR) 1c and FGFR3c. Mol Endocrinol 2008;22:1006–14
  • Ogawa Y, Kurosu H, Yamamoto M, et al. BetaKlotho is required for metabolic activity of fibroblast growth factor 21. Proc Natl Acad Sci USA 2007;104:7432–7
  • So WY, Cheng Q, Chen L, et al. High glucose represses beta-klotho expression and impairs fibroblast growth factor 21 action in mouse pancreatic islets: involvement of peroxisome proliferator-activated receptor gamma signaling. Diabetes 2013;62:3751–9
  • Mashili FL, Austin RL, Deshmukh AS, et al. Direct effects of FGF21 on glucose uptake in human skeletal muscle: implications for type 2 diabetes and obesity. Diabetes Metab Res Rev 2011;27:286–97
  • Arner P, Pettersson A, Mitchell PJ, et al. FGF21 attenuates lipolysis in human adipocytes – a possible link to improved insulin sensitivity. FEBS Lett 2008;582:1725–30
  • Lin Z, Tian H, Lam KS, et al. Adiponectin mediates the metabolic effects of FGF21 on glucose homeostasis and insulin sensitivity in mice. Cell Metab 2013;17:779–89
  • Holland WL, Adams AC, Brozinick JT, et al. An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice. Cell Metab 2013;17:790–7
  • Chen C, Cheung BM, Tso AW, et al. High plasma level of fibroblast growth factor 21 is an Independent predictor of type 2 diabetes: a 5.4-year population-based prospective study in Chinese subjects. Diabetes Care 2011;34:2113–15
  • Zhang X, Yeung DC, Karpisek M, et al. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes 2008;57:1246–53
  • Semba RD, Sun K, Egan JM, et al. Relationship of serum fibroblast growth factor 21 with abnormal glucose metabolism and insulin resistance: the Baltimore Longitudinal Study of Aging. J Clin Endocrinol Metab 2012;97:1375–82
  • Chavez AO, Molina-Carrion M, Abdul-Ghani MA, et al. Circulating fibroblast growth factor-21 is elevated in impaired glucose tolerance and type 2 diabetes and correlates with muscle and hepatic insulin resistance. Diabetes Care 2009;32:1542–6
  • Cheng X, Zhu B, Jiang F, Fan H. Serum FGF-21 levels in type 2 diabetic patients. Endocr Res 2011;36:142–8
  • Li H, Fang Q, Gao F, et al. Fibroblast growth factor 21 levels are increased in nonalcoholic fatty liver disease patients and are correlated with hepatic triglyceride. J Hepatol 2010;53:934–40
  • Li H, Dong K, Fang Q, et al. High serum level of fibroblast growth factor 21 is an independent predictor of non-alcoholic fatty liver disease: a 3-year prospective study in China. J Hepatol 2013;58:557–63
  • Xiao Y, Xu A, Law LS, et al. Distinct changes in serum fibroblast growth factor 21 levels in different subtypes of diabetes. J Clin Endocrinol Metab 2012;97:E54–8
  • Kharitonenkov A, Adams AC. Inventing new medicines: the FGF21 story. Mol Metab 2014;3:221–9
  • Hui E, Xu A, Bo Yang H, Lam KS. Obesity as the common soil of non-alcoholic fatty liver disease and diabetes: role of adipokines. J Diabetes Investig 2013;4:413–25
  • Stefan N, Hennige AM, Staiger H, et al. Alpha2-Heremans-Schmid glycoprotein/fetuin-A is associated with insulin resistance and fat accumulation in the liver in humans. Diabetes Care 2006;29:853–7
  • Yilmaz Y, Yonal O, Kurt R, et al. Serum fetuin A/alpha2HS-glycoprotein levels in patients with non-alcoholic fatty liver disease: relation with liver fibrosis. Ann Clin Biochem 2010;47:549–53
  • Ix JH, Shlipak MG, Brandenburg VM, et al. Association between human fetuin-A and the metabolic syndrome: data from the Heart and Soul Study. Circulation 2006;113:1760–7
  • Ix JH, Wassel CL, Kanaya AM, et al. Fetuin-A and incident diabetes mellitus in older persons. JAMA 2008;300:182–8
  • Vionnet N, Hani EH, Dupont S, et al. Genomewide search for type 2 diabetes-susceptibility genes in French whites: evidence for a novel susceptibility locus for early-onset diabetes on chromosome 3q27-qter and independent replication of a type 2-diabetes locus on chromosome 1q21-q24. Am J Hum Genet 2000;67:1470–80
  • Ishibashi A, Ikeda Y, Ohguro T, et al. Serum fetuin-A is an independent marker of insulin resistance in Japanese men. J Atheroscler Thromb 2010;17:925–33
  • Rasul S, Ilhan A, Reiter MH, et al. Relations of adiponectin to levels of metabolic parameters and sexual hormones in elderly type 2 diabetic patients. Gend Med 2011;8:93–102
  • Mori K, Emoto M, Yokoyama H, et al. Association of serum fetuin-A with insulin resistance in type 2 diabetic and nondiabetic subjects. Diabetes Care 2006;29:468
  • Mori K, Emoto M, Inaba M. Fetuin-A and the cardiovascular system. Adv Clin Chem 2012;56:175–95
  • Ix JH, Sharma K. Mechanisms linking obesity, chronic kidney disease, and fatty liver disease: the roles of fetuin-A, adiponectin, and AMPK. J Am Soc Nephrol 2010;21:406–12
  • Pal D, Dasgupta S, Kundu R, et al. Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med 2012;18:1279–85
  • Hennige AM, Staiger H, Wicke C, et al. Fetuin-A induces cytokine expression and suppresses adiponectin production. PLoS One 2008;3:e1765
  • Jung TW, Youn BS, Choi HY, et al. Salsalate and adiponectin ameliorate hepatic steatosis by inhibition of the hepatokine fetuin-A. Biochem Pharmacol 2013;86:960–9
  • Stefan N, Sun Q, Fritsche A, et al. Impact of the adipokine adiponectin and the hepatokine fetuin-A on the development of type 2 diabetes: prospective cohort- and cross-sectional phenotyping studies. PLoS One 2014;9:e92238
  • Trayhurn P, Drevon CA, Eckel J. Secreted proteins from adipose tissue and skeletal muscle – adipokines, myokines and adipose/muscle cross-talk. Arch Physiol Biochem 2011;117:47–56
  • Liu Y, Chewchuk S, Lavigne C, et al. Functional significance of skeletal muscle adiponectin production, changes in animal models of obesity and diabetes, and regulation by rosiglitazone treatment. Am J Physiol Endocrinol Metab 2009;297:E657–64
  • Raschke S, Eckel J. Adipo-myokines: two sides of the same coin – mediators of inflammation and mediators of exercise. Mediat Inflamm 2013;2013:320724
  • Pedersen BK. Muscle as a secretory organ. Comprehens Physiol 2013;3:1337–62
  • Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006;444:860–7
  • Hu P, Han Z, Couvillon AD, et al. Autocrine tumor necrosis factor alpha links endoplasmic reticulum stress to the membrane death receptor pathway through IRE1alpha-mediated NF-kappaB activation and down-regulation of TRAF2 expression. Mol Cell Biol 2006;26:3071–84
  • Fischer CP. Interleukin-6 in acute exercise and training: what is the biological relevance? Exerc Immunol Rev 2006;12:6–33
  • Carey AL, Steinberg GR, Macaulay SL, et al. Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes 2006;55:2688–97
  • Weigert C, Hennige AM, Lehmann R, et al. Direct cross-talk of interleukin-6 and insulin signal transduction via insulin receptor substrate-1 in skeletal muscle cells. J Biol Chem 2006;281:7060–7
  • White PJ, St-Pierre P, Charbonneau A, et al. Protectin DX alleviates insulin resistance by activating a myokine-liver glucoregulatory axis. Nature Med 2014;20:664–9
  • Wolsk E, Mygind H, Grondahl TS, et al. IL-6 selectively stimulates fat metabolism in human skeletal muscle. Am J Physiol Endocrinol Metab 2010;299:E832–40
  • Yang J. Enhanced skeletal muscle for effective glucose homeostasis. Progr Mol Biol Transl Sci 2014;121:133–63
  • Elliott B, Renshaw D, Getting S, Mackenzie R. The central role of myostatin in skeletal muscle and whole body homeostasis. Acta Physiol (Oxf) 2012;205:324–40
  • Hittel DS, Berggren JR, Shearer J, et al. Increased secretion and expression of myostatin in skeletal muscle from extremely obese women. Diabetes 2009;58:30–8
  • Zhao B, Wall RJ, Yang J. Transgenic expression of myostatin propeptide prevents diet-induced obesity and insulin resistance. Biochem Biophys Res Commun 2005;337:248–55
  • Gonzalez-Cadavid NF, Bhasin S. Role of myostatin in metabolism. Curr Opin Clin Nutr Metab Care 2004;7:451–7
  • Morissette MR, Cook SA, Buranasombati C, et al. Myostatin inhibits IGF-I-induced myotube hypertrophy through Akt. Am J Physiol Cell Physiol 2009;297:C1124–32
  • Wilkes JJ, Lloyd DJ, Gekakis N. Loss-of-function mutation in myostatin reduces tumor necrosis factor alpha production and protects liver against obesity-induced insulin resistance. Diabetes 2009;58:1133–43
  • Hittel DS, Axelson M, Sarna N, et al. Myostatin decreases with aerobic exercise and associates with insulin resistance. Med Sci Sports Exerc 2010;42:2023–9
  • Ehehalt S, Schweizer R, Blumenstock G, et al. Investigation of myostatin serum levels before and after a 6-month lifestyle intervention program in obese children. Exp Clin Endocrinol Diabetes 2011;119:238–42
  • Hofmann T, Elbelt U, Stengel A. Irisin as a muscle-derived hormone stimulating thermogenesis – a critical update. Peptides 2014;54:89–100
  • Bostrom P, Wu J, Jedrychowski MP, et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012;481:463–8
  • Timmons JA, Baar K, Davidsen PK, Atherton PJ. Is irisin a human exercise gene? Nature 2012;488:E9–10; discussion E-1
  • Huh JY, Panagiotou G, Mougios V, et al. FNDC5 and irisin in humans: I. Predictors of circulating concentrations in serum and plasma and II. mRNA expression and circulating concentrations in response to weight loss and exercise. Metab: Clin Exp 2012;61:1725–38
  • Park KH, Zaichenko L, Brinkoetter M, et al. Circulating irisin in relation to insulin resistance and the metabolic syndrome. J Clin Endocrinol Metab 2013;98:4899–907
  • Choi YK, Kim MK, Bae KH, et al. Serum irisin levels in new-onset type 2 diabetes. Diabetes Res Clin Pract 2013;100:96–101
  • Nicholson JK, Lindon JC. Systems biology: metabonomics. Nature 2008;455:1054–6
  • Kurland IJ, Accili D, Burant C, et al. Application of combined omics platforms to accelerate biomedical discovery in diabesity. Ann N Y Acad Sci 2013;1287:1–16
  • Di Girolamo F, Del Chierico F, Caenaro G, et al. Human serum proteome analysis: new source of markers in metabolic disorders. Biomark Med 2012;6:759–73
  • Zhao YY, Lin RC. UPLC-MS(E) application in disease biomarker discovery: the discoveries in proteomics to metabolomics. Chemico-Biol Interact 2014;215:7–16
  • Kim OY, Lee JH, Sweeney G. Metabolomic profiling as a useful tool for diagnosis and treatment of chronic disease: focus on obesity, diabetes and cardiovascular diseases. Expert Rev Cardiovasc Therapy 2013;11:61–8
  • Milburn MV, Lawton KA. Application of metabolomics to diagnosis of insulin resistance. Ann Rev Med 2013;64:291–305
  • Batch BC, Shah SH, Newgard CB, et al. Branched chain amino acids are novel biomarkers for discrimination of metabolic wellness. Metab: Clin Exp 2013;62:961–9
  • Scholtens DM, Muehlbauer MJ, Daya NR, et al. Metabolomics reveals broad-scale metabolic perturbations in hyperglycemic mothers during pregnancy. Diabetes Care 2014;37:158–66
  • Newgard CB, An J, Bain JR, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab 2009;9:311–26
  • Huffman KM, Shah SH, Stevens RD, et al. Relationships between circulating metabolic intermediates and insulin action in overweight to obese, inactive men and women. Diabetes Care 2009;32:1678–83
  • Liu Y, Turdi S, Park T, et al. Adiponectin corrects high-fat diet-induced disturbances in muscle metabolomic profile and whole-body glucose homeostasis. Diabetes 2013;62:743–52
  • Billings LK, Florez JC. The genetics of type 2 diabetes: what have we learned from GWAS? Ann N Y Acad Sci 2010;1212:59–77
  • Rhee EP, Clish CB, Ghorbani A, et al. A combined epidemiologic and metabolomic approach improves CKD prediction. J Am Soc Nephrol 2013;24:1330–8
  • Shin SY, Fauman EB, Petersen AK, et al. An atlas of genetic influences on human blood metabolites. Nat Genet 2014;46:543–50
  • Karlsson F, Tremaroli V, Nielsen J, Backhed F. Assessing the human gut microbiota in metabolic diseases. Diabetes 2013;62:3341–9
  • Cani PD, Geurts L, Matamoros S, et al. Glucose metabolism: focus on gut microbiota, the endocannabinoid system and beyond. Diabet Metab 2014 ;40:246--57
  • Moreno-Indias I, Cardona F, Tinahones FJ, Queipo-Ortuno MI. Impact of the gut microbiota on the development of obesity and type 2 diabetes mellitus. Front Microbiol 2014;5:190
  • Musso G, Gambino R, Cassader M. Obesity, diabetes, and gut microbiota: the hygiene hypothesis expanded? Diabetes Care 2010;33:2277–84
  • Tilg H, Kaser A. Gut microbiome, obesity, and metabolic dysfunction. J Clin Investig 2011;121:2126–32
  • Membrez M, Blancher F, Jaquet M, et al. Gut microbiota modulation with norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J 2008;22:2416–26
  • Andoh A, Benno Y, Kanauchi O, Fujiyama Y. Recent advances in molecular approaches to gut microbiota in inflammatory bowel disease. Curr Pharmaceut Design 2009;15:2066–73
  • Hermann-Bank ML, Skovgaard K, Stockmarr A, et al. The Gut Microbiotassay: a high-throughput qPCR approach combinable with next generation sequencing to study gut microbial diversity. BMC Genom 2013;14:788
  • Bergstrom A, Licht TR, Wilcks A, et al. Introducing GUt low-density array (GULDA): a validated approach for qPCR-based intestinal microbial community analysis. FEMS Microbiol Lett 2012;337:38–47
  • Tottey W, Denonfoux J, Jaziri F, et al. The human gut chip “HuGChip”, an explorative phylogenetic microarray for determining gut microbiome diversity at family level. PLoS One 2013;8:e62544
  • Rajpal DK, Brown JR. Modulating the human gut microbiome as an emerging therapeutic paradigm. Sci Progr 2013;96:224–36
  • Bajaj M, Defronzo RA. Metabolic and molecular basis of insulin resistance. J Nucl Cardiol 2003;10:311–23

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.