Insulin resistance

Arab Journal of Gastroenterology(2010)

引用 0|浏览6
暂无评分
摘要
Insulin resistance (IR) is the pathogenic foundation underlying metabolic syndrome, steatosis and cirrhotic-NASH, and possibly HCC. The interplay between genetic and environmental risk factors ultimately leads to the development of IR. Obesity is considered a major risk factor, with dysregulation of levels of secreted adipokines from distended adipose tissue playing a major role in IR. HCV-induced IR may be due to the HCV core protein inducing proteasomal degradation of IRS 1 and 2, impairing expression of IRS 1 and 2 and blocking intracellular insulin signalling. The latter is mediated by increased levels of both TNF-α and suppressor of cytokine signalling 3 (SOC-3). IR, through different mechanisms, plays a role in the development of steatosis and its progression to steatohepatitis, cirrhosis, and even HCC. In addition, it has a role in impairing IFN signalling cascade (JAK → STAT → IFN genes), as insulin activates PI3K thus blocking STAT-1 translocation, avoiding the antiviral effect of interferon. Introduction Insulin resistance is a condition in which a higher than normal insulin concentration is needed to achieve normal metabolic responses. The overall prevalence of IR is significant; some have reported that 10–25% of the population is affected by the condition [1] , others have reported that approximately 1/3 of the apparently healthy population are insulin resistant [2,3] . 30–70% of patients with CHC display some form of IR [4] . Worldwide, 47 million patients may have HCV-associated DM [5] . Calculation of IR • HOMA-IR = Fasting insulin (μU/L) × fasting glucose (mmol/L)/22.5. • Convert glucose in mg/dL → mmol/L, divide glucose by 18. Values >2 are consistent with IR. Types of insulin resistance Insulin resistance may be either peripheral (which refers to diminished insulin-mediated uptake of glucose by skeletal muscle and depends primarily on the failure of glucose transporter type 4 (GLUT4) expression and translocation to the plasma membrane) or hepatic insulin resistance (which describes impaired suppression of hepatic glucose production, and largely accounts for hyperglycemia and glucose intolerance). Phases of insulin resistance [6] 1. Prereceptor: due to abnormal insulin (mutations) or anti-insulin antibodies. 2. Receptor: due to – Decreased number of receptors and proteasomal degradation of insulin receptor substrate (IRS). – Abnormal insulin receptor (mutation). – Antibodies blocking insulin-receptor binding. – Activation of receptor on serine rather than tyrosine kinase. – Reactive oxygen species (ROS), which decreases tyrosine phosphorylation of IRS. 3. Postreceptor: – TNF-α, IL-6 and insulin inhibit insulin signalling via induction of SOCs 1, 3. – Mutations of GLUT4. 4. Combination of defects [6] . Risk factors of insulin resistance ( Fig. 1 ) • Gender : Increased visceral adiposity in men leads to a higher risk of IR than in women [7] . • Aging : Associated with increased levels of FFAs and TG, a decrease and mutation of GLUT4. • Obesity : Associated with decreased number and downregulation of insulin receptors and impairment of postreceptor signalling. Overflow of FFAs from adipose tissue interferes with intrahepatic insulin signalling pathway via increased levels of pro-inflammatory cytokines such as TNF-α [8,9] and proteasomal degradation of the insulin receptor substrate-1 and 2 (IRS-1 and 2) [10] . • Smoking : Induces the production of high levels of circulating catecholamines, which act as antagonists to insulin-action [11] . Smoking contributes to increased levels of TG and decreased HDL. • Alcohol : Doses higher than 50 g/day leads to higher risk of IR. • Increased production of insulin antagonists : Cushing syndrome, acromegaly, stress, diabetic ketoacidosis, severe infection, uremia, trauma, surgery, liver cirrhosis, etc. • Medications, particularly : Glucocorticoids, cyclosporine, growth hormones, thyroid hormones, sex hormones, protease inhibitors (HCV), nucleoside analogues (HBV), thiazides, β-blockers. • Exercise : Low physical activity and high energy intake leads to high risk of IR [12] . In addition it has been reported that IR improves after exercise [13] . • Cirrhosis : Induces IR due to diminished insulin extraction by the liver as well as portosystemic shunting of insulin, whilst increasing incontra-insulin hormones (glucogen, growth hormones, IGF, FFAs). Cirrhosis can downregulate insulin receptor or postreceptor function [14] . • HCV : Core proteins downregulate IRS and block intracellular insulin signalling. • Periodontal infection : Patients with severe periodontitis have six times greater risk of worsening glycaemic control over time than patients without periodontitis, a finding attributed to increased insulin resistance [15] . Recent findings suggest that pro-inflammatory proteins (e.g., Tumour Necrosis Factor TNF-α and Interleukin IL-1b) can induce insulin resistance by interfering with lipid metabolism. It appears that prevention and treatment of periodontal infections can have a beneficial effect on glycaemic control in diabetic patients [16] ( Fig. 1 ). Insulin resistance and metabolic syndrome MS is a set of risk factors that includes: abdominal obesity, a decreased ability to process glucose (increased blood glucose and/or insulin resistance), dyslipidaemia, and hypertension [17] . The functional defect in patients with MS is resistance to the cellular action of insulin, particularly to insulin stimulated glucose uptake [18] . Patients who have this syndrome have been shown to be at an increased risk of developing cardiovascular disease and/or type-2 diabetes. IR is the pathogenic foundation underlying metabolic syndrome (MS) [4] . IR may play an important role in the development of hyperinsulinemia, hyperglycemia (due to enhanced hepatic gluconeogenesis and glucose output), an increase in plasma FFAs, and dyslipidemia, which can further aggravate the degree of IR [19] . However, a substantial proportion of patients with MS do not exhibit evidence of IR. Role of adipokines in insulin resistance Adipokines are polypeptides secreted in the adipose tissue in a regulated manner. Adipolines include Adiponectin, Leptin, Resistin, Retinol binding protein 4, Visfatin, Omentin, Vaspin, Chemerin, Apelin, TNF-α, IL-6, and Monocyte chemoattractant protein-1 (MCP-1) [20] . Adiponectin Acts as anti-inflammatory, hepatoprotective adipokine [21] . Some of its beneficial effects include suppression of TNF-α and IL-6 expression [22] , leading to increased insulin sensitivity [23] , glucose uptake, FA oxidation and activation of PPAR-α, which leads improved insulin sensitivity [24] . Finally, adiponectin prevent hepatic steatosis [25] . Deficiency of adiponectin is associated with: obesity, insulin resistance, glucose intolerance, TG accumulation and steatosis, and metabolic syndrome [20] . Leptin Upregulation of leptin has been found to be associated with regulation of food intake and body weight (anorexigenic), and limited accumulation of TG in the liver and skeletal muscles, thus inhibiting steatosis. In addition to stimulation of fatty acid oxidation, improved insulin sensitivity and stimulation of PI3K signalling can lead to increased glucose uptake by hepatocytes [26] . On the other hand, leptin insufficiency is associated with increased body weight, increased FA synthesis, decreased FA oxidation, decreased TG excretion, increased steatosis, and impaired insulin sensitivity and secretion [20] . TNF-α Impairs phosphorylation of IRS on tyrosine kinase [27] . Triggers secretion of FFAs from adipose tissue, causing proteasomal degradation of IRS. Inhibits P-R signalling via induction of SOCs, which leads to inactivation of PI3K and inhibition of translocation of GLUT4 to the cell membrane, thus blocking glucose entry [27] . Inactivation of both IRS and post receptor signalling generate IR, with subsequent development of hepatic steatosis and activation of HSCs, eventually leading to fibrosis. On the other hand, the nuclear receptor peroxisome proliferator-activated receptor-γ (PPAR-γ) has an anti-inflammatory role, involved in the regulation of glucose and lipid homeostasis and may have insulin sensitising effects. Upregulation of PPAR-γ regulates FA uptake by the liver. Control of TG and VLDL synthesis prevent hepatic steatosis. PPAR-α is known to be activated by insulin sensitisers (thiazolidinedione (TDZ)). Alternatively, downregulation of PPAR-γ occurs in cases of IR due to impaired expression of PPAR-γ, leading to retention of TG with subsequent development of fatty liver and upregulation of NFκ−β, which can accelerate HCV replication. HCV core protein also inhibits PPAR-α and γ promoting IRS-1 degradation, thereby generating IR [28] . HCV-induced insulin resistance HCV generates necroinflammation of cell membranes with consequent reduction of insulin receptors. HCV-Core protein induces IR by: proteasomal degradation of IRS 1 and 2, impairing expression of IRS 1 and 2, blocking intracellular insulin signalling mediated by increased levels of pro-inflammatory cytokine TNF-α [9,10] ; increased suppression of cytokine signalling 3 (SOC-3) [8] ; inhibition of phosphorylation of PI3K, leading to decreased translocation of GLUT4 to the cell membrane and glucose entry. HCV genotype non-3 induces IR via downregulation of IRS [29–31] through phosphorylation of the receptor on serine. HCV genotype-3 may induce a cytopathic effect and auto-immune aggression on b-cells of the pancreas [32] . Data suggest that either HCV per se or its inflammatory response in the liver plays a central role in the development of IR in patients with CHC [33] . Role of insulin resistance in steatosis development It is well known that insulin resistance causes impaired metabolic clearance of glucose. In addition, it also increases lipolysis, leading to increased plasma FFAs. Increased hepatic uptake of FFAs impairs b-oxidation and decreases excretion of TG-Apo B100 (VLDL), resulting in steatosis. IR impairs expression of anti-inflammatory PPAR-γ, resulting in TG retention. IR also reduces adiponectin and induces a loss of adiponectin receptors, resulting in TG retention with subsequent development of fatty liver [34–38] . The mechanisms leading NAFLD to NASH and cirrhosis Increased FFA flux to the liver is caused by excessive adipose tissue lipolysis, resulting from peripheral IR. In patients that fail to adequately incorporate FFAs into TG (“bad fat storers”) NASH develops as a result of direct lipotoxicity and generation of ROS. Individuals that efficiently store FFA as TG (“good fat storers”) have a benign course unless additional hepatic injury occurs (“second hit”). Progression to cirrhosis depends on fibrogenic factors [39] . Progression of metabolic cirrhosis to HCC Metabolic fibrosis or cirrhosis is associated with the following: oxidative stress, cell cycle stimulation, apoptosis inhibition, mutagenesis. Each is capable of developing hepatic cancer. Insulin-induced IFN resistance ( Fig. 2 ) IR-induced steatosis increases the risk of fibrosis and reduces antiviral activity. In addition, HCV-induces necroinflammation, with consequent reduction and malfunction of IFN receptors. Steatosis/NASH/fibrosis decreases the contact between IFN and receptor [40] , impairing IFN signalling cascade (JAK → STAT → IFN genes) [41] . Moreover, PI3K, activated by insulin, seems to be responsible for the block of STAT-1 translocation, eventually inhibiting production of ISG (Mx, 2′5′ OAS, PKR) ( Fig. 2 ). Insulin resistance can be improved by regulation of food intake and body weight, and exercise reduces peripheral IR. Pharmacological intervention with agents that target insulin resistance (metformin, thiazolidinedione (TDZ)) can also be used. In conclusion, IR plays a key role in the pathogenesis of metabolic syndrome, steatosis and cirrhotic-NASH, and perhaps HCC. Steatosis independently increases the risk of HCC. IR and steatosis reduce SVR to antiviral therapy. Diet control, exercise, and insulin sensitisers improve insulin sensitivity. Pentoxifylline also reduces TNF-α and may improve insulin sensitivity. References [1] E. Ferrannini A. Natali B. Capaldo M. Lehtovirta S. Jacob H. Yki-Jarvinen Insulin resistance, hyperinsulinemia, and blood pressure: role of age and obesity. European group for the study of insulin resistance (EGIR) Hypertension 30 5 1997 1144 1149 [2] F.S. Facchini N. Hua F. Abbasi G.M. Reaven Insulin resistance as a predictor of age-related diseases J Clin Endocrinol Metab 86 8 2001 3574 3578 [3] J. Yip F.S. Facchini G.M. Reaven Resistance to insulin-mediated glucose disposal as a predictor of cardiovascular disease J Clin Endocrinol Metab 83 8 1998 2773 2776 [4] S.A. Harrison Insulin resistance among patients with chronic hepatitis C: etiology and impact on treatment Clin Gastroenterol Hepatol 6 8 2008 864 876 [5] H. Knobler T. Zhornicky A. Sandler N. Haran Y. Ashur A. Schattner Tumor necrosis factor-alpha-induced insulin resistance may mediate the hepatitis C virus-diabetes association Am J Gastroenterol 98 12 2003 2751 2756 [6] S.J. Hunter W.T. Garvey Insulin action and insulin resistance: diseases involving defects in insulin receptors, signal transduction, and the glucose transport effector system Am J Med 105 4 1998 331 345 [7] C. Couillard N. Bergeron D. Prud’homme J. Bergeron A. Tremblay C. Bouchard Gender difference in postprandial lipemia: importance of visceral adipose tissue accumulation Arterioscl Thromb Vasc Biol 19 10 1999 2448 2455 [8] H. Knobler A. Schattner TNF-{alpha}, chronic hepatitis C and diabetes: a novel triad QJM 98 1 2005 1 6 [9] Y. Shintani H. Fujie H. Miyoshi T. Tsutsumi K. Tsukamoto S. Kimura Hepatitis C virus infection and diabetes: direct involvement of the virus in the development of insulin resistance Gastroenterology 126 3 2004 840 848 [10] T. Kawaguchi T. Yoshida M. Harada T. Hisamoto Y. Nagao T. Ide Hepatitis C virus down-regulates insulin receptor substrates 1 and 2 through up-regulation of suppressor of cytokine signaling 3 Am J Pathol 165 5 2004 1499 1508 [11] I. Lager S. Attvall B.M. Eriksson S.H. von U. Smith Studies on the insulin-antagonistic effect of catecholamines in normal man. Evidence for the importance of beta 2-receptors Diabetologia 29 7 1986 409 416 [12] V.S. Conn A.R. Hafdahl D.R. Mehr J.W. LeMaster S.A. Brown P.J. Nielsen Metabolic effects of interventions to increase exercise in adults with type 2 diabetes Diabetologia 50 5 2007 913 921 [13] G.D. Cartee D.A. Young M.D. Sleeper J. Zierath H. Wallberg-Henriksson J.O. Holloszy Prolonged increase in insulin-stimulated glucose transport in muscle after exercise Am J Physiol 256 4 Pt 1 1989 E494 E499 [14] P.S. Del F. Leonetti D.C. Simonson P. Sheehan M. Matsuda R.A. DeFronzo Effect of sustained physiologic hyperinsulinaemia and hyperglycaemia on insulin secretion and insulin sensitivity in man Diabetologia 37 10 1994 1025 1035 [15] G.W. Taylor B.A. Burt M.P. Becker R.J. Genco M. Shlossman W.C. Knowler Severe periodontitis and risk for poor glycemic control in patients with non-insulin-dependent diabetes mellitus J Periodontol 67 Suppl. 10 1996 1085 1093 [16] G.W. Taylor The effects of periodontal treatment on diabetes J Am Dent Assoc 134 Spec. No. 2003 41S 48S [17] R.H. Eckel S.M. Grundy P.Z. Zimmet The metabolic syndrome Lancet 365 9468 2005 1415 1428 [18] R.K. Avramoglu H. Basciano K. Adeli Lipid and lipoprotein dysregulation in insulin resistant states Clin Chim Acta 368 1–2 2006 1 19 [19] N. Mikhail The metabolic syndrome: insulin resistance Curr Hypertens Rep 11 2 2009 156 158 [20] K. Rabe M. Lehrke K.G. Parhofer U.C. Broedl Adipokines and insulin resistance Mol Med 14 11–12 2008 741 751 [21] N. Ouchi R. Shibata K. Walsh Targeting adiponectin for cardioprotection Expert Opin Ther Targets 10 4 2006 573 581 [22] C. Tsatsanis V. Zacharioudaki A. Androulidaki E. Dermitzaki I. Charalampopoulos V. Minas Adiponectin induces TNF-alpha and IL-6 in macrophages and promotes tolerance to itself and other pro-inflammatory stimuli Biochem Biophys Res Commun 335 4 2005 1254 1263 [23] A.H. Berg T.P. Combs X. Du M. Brownlee P.E. Scherer The adipocyte-secreted protein Acrp30 enhances hepatic insulin action Nat Med 7 8 2001 947 953 [24] N. Maeda M. Takahashi T. Funahashi S. Kihara H. Nishizawa K. Kishida PPARgamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein Diabetes 50 9 2001 2094 2099 [25] E.E. Kershaw J.S. Flier Adipose tissue as an endocrine organ J Clin Endocrinol Metab 89 6 2004 2548 2556 [26] S. Valtuena F. Numeroso D. Ardigo M. Pedrazzoni L. Franzini P.M. Piatti Relationship between leptin, insulin, body composition and liver steatosis in non-diabetic moderate drinkers with normal transaminase levels Eur J Endocrinol 153 2 2005 283 290 [27] G.S. Hotamisligil P. Peraldi A. Budavari R. Ellis M.F. White B.M. Spiegelman IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance Science 271 5249 1996 665 668 [28] G.A. De V. Pazienza P. Pugnale F. Bruttin L. Rubbia-Brandt C.E. Juge-Aubry Peroxisome proliferator-activated receptor-alpha and -gamma mRNA levels are reduced in chronic hepatitis C with steatosis and genotype 3 infection Aliment Pharmacol Therap 23 1 2006 107 114 [29] P. Cavallo-Perin M. Cassader C. Bozzo A. Bruno P. Nuccio A.M. Dall’omo Mechanism of insulin resistance in human liver cirrhosis. Evidence of a combined receptor and postreceptor defect J Clin Invest 75 5 1985 1659 1665 [30] P. Cavallo-Perin A. Bruno P. Nuccio M. Goria G. Pagano G. Lenti Feedback inhibition of insulin secretion is altered in cirrhosis J Clin Endocrinol Metab 63 4 1986 1023 1027 [31] A. Nygren N. Adner L. Sundblad K.L. Wiechel Insulin uptake by the human alcoholic cirrhotic liver Metabolism 34 1 1985 48 52 [32] A.R. El-Zayadi O.E. Selim H. Hamdy H. Dabbous A. Ahdy S.A. Moniem Association of chronic hepatitis C infection and diabetes mellitus Trop Gastroenterol 19 4 1998 141 144 [33] A. Zekry J.G. McHutchison A.M. Diehl Insulin resistance and steatosis in hepatitis C virus infection Gut 54 7 2005 903 906 [34] S. Chitturi S. Abeygunasekera G.C. Farrell J. Holmes-Walker J.M. Hui C. Fung NASH and insulin resistance. Insulin hypersecretion and specific association with the insulin resistance syndrome Hepatology 35 2 2002 373 379 [35] G. Marchesini M. Brizi A.M. Morselli-Labate G. Bianchi E. Bugianesi A.J. McCullough Association of nonalcoholic fatty liver disease with insulin resistance Am J Med 107 5 1999 450 455 [36] G. Pagano G. Pacini G. Musso R. Gambino F. Mecca N. Depetris Nonalcoholic steatohepatitis, insulin resistance, and metabolic syndrome: further evidence for an etiologic association Hepatology 35 2 2002 367 372 [37] V. Pazienza S. Clement P. Pugnale S. Conzelman M. Foti A. Mangia The hepatitis C virus core protein of genotypes 3a and 1b downregulates insulin receptor substrate 1 through genotype-specific mechanisms Hepatology 45 5 2007 1164 1171 [38] I.R. Willner B. Waters S.R. Patil A. Reuben J. Morelli C.A. Riely Ninety patients with nonalcoholic steatohepatitis: insulin resistance, familial tendency, and severity of disease Am J Gastroenterol 96 10 2001 2957 2961 [39] M. Cave I. Deaciuc C. Mendez Z. Song S. Joshi-Barve S. Barve Nonalcoholic fatty liver disease: predisposing factors and the role of nutrition J Nutr Biochem 18 3 2007 184 195 [40] M. Romero-Gomez M.V. Del R.J. Andrade J. Salmeron M. Diago C.M. Fernandez-Rodriguez Insulin resistance impairs sustained response rate to peginterferon plus ribavirin in chronic hepatitis C patients Gastroenterology 128 3 2005 636 641 [41] K. Kaserer R. Fiedler P. Steindl C.H. Muller F. Wrba P. Ferenci Liver biopsy is a useful predictor of response to interferon therapy in chronic hepatitis C Histopathology 32 5 1998 454 461
更多
查看译文
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要