Category: Health

Insulin sensitivity and insulin receptors

Insulin sensitivity and insulin receptors

Because Insulin sensitivity and insulin receptors signaling insilin involved insuoin multiple vital cellular functions, the protein level of IR needs to be tightly controlled. Antibodies and reagents. Chromosomal instability and aneuploidy in cancer: from yeast to man.

Insulin sensitivity and insulin receptors -

Many hormones can induce insulin resistance including cortisol , [14] growth hormone , and human placental lactogen. Cortisol counteracts insulin and can lead to increased hepatic gluconeogenesis , reduced peripheral utilization of glucose, and increased insulin resistance.

Based on the significant improvement in insulin sensitivity in humans after bariatric surgery and rats with surgical removal of the duodenum, [19] [20] it has been proposed that some substance is produced in the mucosa of that initial portion of the small intestine that signals body cells to become insulin resistant.

If the producing tissue is removed, the signal ceases and body cells revert to normal insulin sensitivity. No such substance has been found as yet, and the existence of such a substance remains speculative. Leptin is a hormone produced from the ob gene and adipocytes. Polycystic ovary syndrome [24] and non-alcoholic fatty liver disease NAFLD are associated with insulin resistance.

Hepatitis C also makes people three to four times more likely to develop type 2 diabetes and insulin resistance. Multiple studies involving different methodology suggest that impaired function of mitochondria might play a pivotal role in the pathogenesis of insulin resistance.

Acute or chronic inflammation, such as in infections, can cause insulin resistance. TNF-α is a cytokine that may promote insulin resistance by promoting lipolysis , disrupting insulin signaling, and reducing the expression of GLUT4.

Several genetic loci have been determined to be associated with insulin insensitivity. This includes variation in loci near the NAT2, GCKR, and IGFI genes associated with insulin resistance. Further research has shown that loci near the genes are linked to insulin resistance.

In normal metabolism, the elevated blood glucose instructs beta β cells in the Islets of Langerhans , located in the pancreas , to release insulin into the blood. The insulin makes insulin-sensitive tissues in the body primarily skeletal muscle cells, adipose tissue, and liver absorb glucose which provides energy as well as lowers blood glucose.

In an insulin-resistant person, normal levels of insulin do not have the same effect in controlling blood glucose levels. When the body produces insulin under conditions of insulin resistance, the cells are unable to absorb or use it as effectively and it stays in the bloodstream.

Certain cell types such as fat and muscle cells require insulin to absorb glucose and when these cells fail to respond adequately to circulating insulin, blood glucose levels rise. The liver normally helps regulate glucose levels by reducing its secretion of glucose in the presence of insulin.

However, in insulin resistance, this normal reduction in the liver's glucose production may not occur, further contributing to elevated blood glucose. Insulin resistance in fat cells results in reduced uptake of circulating lipids and increased hydrolysis of stored triglycerides.

This leads to elevated free fatty acids in the blood plasma and can further worsen insulin resistance. In states of insulin resistance, beta cells in the pancreas increase their production of insulin.

This causes high blood insulin hyperinsulinemia to compensate for the high blood glucose. During this compensated phase of insulin resistance, beta cell function is upregulated, insulin levels are higher, and blood glucose levels are still maintained.

If compensatory insulin secretion fails, then either fasting impaired fasting glucose or postprandial impaired glucose tolerance glucose concentrations increase. Eventually, type 2 diabetes occurs when glucose levels become higher as the resistance increases and compensatory insulin secretion fails.

Insulin resistance is strongly associated with intestinal-derived apoB production rate in insulin-resistant subjects and type 2 diabetics.

With respect to visceral adiposity, a great deal of evidence suggests two strong links with insulin resistance. In numerous experimental models, these proinflammatory cytokines disrupt normal insulin action in fat and muscle cells and may be a major factor in causing the whole-body insulin resistance observed in patients with visceral adiposity.

Second, visceral adiposity is related to an accumulation of fat in the liver, a condition known as non-alcoholic fatty liver disease NAFLD. The result of NAFLD is an excessive release of free fatty acids into the bloodstream due to increased lipolysis , and an increase in hepatic breakdown of glycogen stores into glucose glycogenolysis , both of which have the effect of exacerbating peripheral insulin resistance and increasing the likelihood of Type 2 diabetes mellitus.

The excessive expansion of adipose tissue that tends to occur under sustainedly positive energy balance as in overeating has been postulated by Vidal-Puig to induce lipotoxic and inflammatory effects that may contribute to causing insulin resistance and its accompanying disease states.

Also, insulin resistance often is associated with a hypercoagulable state impaired fibrinolysis and increased inflammatory cytokine levels. From a broader perspective, however, sensitivity tuning including sensitivity reduction is a common practice for an organism to adapt to the changing environment or metabolic conditions.

This can be achieved through raising the response threshold i. Insulin resistance has been proposed to be a reaction to excess nutrition by superoxide dismutase in cell mitochondria that acts as an antioxidant defense mechanism. This link seems to exist under diverse causes of insulin resistance.

It also is based on the finding that insulin resistance may be reversed rapidly by exposing cells to mitochondrial uncouplers, electron transport chain inhibitors, or mitochondrial superoxide dismutase mimetics. During a glucose tolerance test GTT , which may be used to diagnose diabetes mellitus, a fasting patient takes a 75 gram oral dose of glucose.

Then blood glucose levels are measured over the following two hours. Interpretation is based on WHO guidelines.

After two hours a glycemia less than 7. An oral glucose tolerance test OGTT may be normal or mildly abnormal in simple insulin resistance.

Often, there are raised glucose levels in the early measurements, reflecting the loss of a postprandial peak after the meal in insulin production.

Extension of the testing for several more hours may reveal a hypoglycemic "dip," that is a result of an overshoot in insulin production after the failure of the physiologic postprandial insulin response. The gold standard for investigating and quantifying insulin resistance is the "hyperinsulinemic euglycemic clamp," so-called because it measures the amount of glucose necessary to compensate for an increased insulin level without causing hypoglycemia.

The test is rarely performed in clinical care, but is used in medical research, for example, to assess the effects of different medications. The rate of glucose infusion commonly is referred to in diabetes literature as the GINF value. The procedure takes about two hours. Through a peripheral vein , insulin is infused at 10— mU per m 2 per minute.

The rate of glucose infusion is determined by checking the blood sugar levels every five to ten minutes. The rate of glucose infusion during the last thirty minutes of the test determines insulin sensitivity.

If high levels 7. Very low levels 4. Levels between 4. This basic technique may be enhanced significantly by the use of glucose tracers. Glucose may be labeled with either stable or radioactive atoms.

Commonly used tracers are 3- 3 H glucose radioactive , 6,6 2 H-glucose stable and 1- 13 C Glucose stable. Prior to beginning the hyperinsulinemic period, a 3h tracer infusion enables one to determine the basal rate of glucose production.

During the clamp, the plasma tracer concentrations enable the calculation of whole-body insulin-stimulated glucose metabolism, as well as the production of glucose by the body i.

Another measure of insulin resistance is the modified insulin suppression test developed by Gerald Reaven at Stanford University. The test correlates well with the euglycemic clamp, with less operator-dependent error.

This test has been used to advance the large body of research relating to the metabolic syndrome. Patients initially receive 25 μg of octreotide Sandostatin in 5 mL of normal saline over 3 to 5 minutes via intravenous infusion IV as an initial bolus, and then, are infused continuously with an intravenous infusion of somatostatin 0.

Blood glucose is checked at zero, 30, 60, 90, and minutes, and thereafter, every 10 minutes for the last half-hour of the test. These last four values are averaged to determine the steady-state plasma glucose level SSPG.

Given the complicated nature of the "clamp" technique and the potential dangers of hypoglycemia in some patients , alternatives have been sought to simplify the measurement of insulin resistance. The first was the Homeostatic Model Assessment HOMA , [52] and more recent methods include the Quantitative insulin sensitivity check index QUICKI [53] and SPINA-GR , a measure for insulin sensitivity.

Maintaining a healthy body weight and being physically active can help reduce the risk of developing insulin resistance.

The primary treatment for insulin resistance is exercise and weight loss. Metformin is approved for prediabetes and type 2 diabetes and has become one of the more commonly prescribed medications for insulin resistance. The Diabetes Prevention Program DPP showed that exercise and diet were nearly twice as effective as metformin at reducing the risk of progressing to type 2 diabetes.

Furthermore, physical training has also generally been seen to be an effective antagonist of insulin resistance in obese or overweight children and adolescents under the age of Resistant starch from high-amylose corn, amylomaize , has been shown to reduce insulin resistance in healthy individuals, in individuals with insulin resistance, and in individuals with type 2 diabetes.

Some types of polyunsaturated fatty acids omega-3 may moderate the progression of insulin resistance into type 2 diabetes, [62] [63] [64] however, omega-3 fatty acids appear to have limited ability to reverse insulin resistance, and they cease to be efficacious once type 2 diabetes is established.

The concept that insulin resistance may be the underlying cause of diabetes mellitus type 2 was first advanced by Professor Wilhelm Falta and published in Vienna in , [66] and confirmed as contributory by Sir Harold Percival Himsworth of the University College Hospital Medical Centre in London in ; [67] however, type 2 diabetes does not occur unless there is concurrent failure of compensatory insulin secretion.

Some scholars go as far as to claim that neither insulin resistance, nor obesity really are metabolic disorders per se , but simply adaptive responses to sustained caloric surplus, intended to protect bodily organs from lipotoxicity unsafe levels of lipids in the bloodstream and tissues : "Obesity should therefore not be regarded as a pathology or disease, but rather as the normal, physiologic response to sustained caloric surplus As a consequence of the high level of lipid accumulation in insulin target tissues including skeletal muscle and liver, it has been suggested that exclusion of glucose from lipid-laden cells is a compensatory defense against further accumulation of lipogenic substrate.

Potts, J. Sex and ethnic group differences in fat distribution in young United Kingdom South Asians and Europids. Ehtisham, S. Ethnic differences in insulin resistance and body composition in United Kingdom adolescents.

Lear, S. Ethnic variation in fat and lean body mass and the association with insulin resistance. Mason, C. Dietary weight loss and exercise effects on insulin resistance in postmenopausal women.

Med 41 , — Mikusova, V. Insulin resistance and need for a lifestyle change to eliminate it. Listy , — orpeleijn, E.

Metabolic flexibility in the development of insulin resistance and type 2 diabetes: effects of lifestyle. Shigeta, H. Lifestyle, obesity, and insulin resistance. Diabetes Care 24 , Oosterman, J. The circadian clock, shift work, and tissue-specific insulin resistance. Endocrinology , bqaa McAuley, K.

Intensive lifestyle changes are necessary to improve insulin sensitivity: a randomized controlled trial. Diabetes care 25 , — Bergman, B. Novel and reversible mechanisms of smoking-induced insulin resistance in humans.

Diabetes 61 , — Kan, C. A systematic review and meta-analysis of the association between depression and insulin resistance. Diabetes care 36 , — Sung, C.

Role of vitamin D in insulin resistance. Ardabili, H. Vitamin D supplementation has no effect on insulin resistance assessment in women with polycystic ovary syndrome and vitamin D deficiency. Pasieka, A.

Impact of glucocorticoid excess on glucose tolerance: clinical and preclinical evidence. Metabolites 6 , 24 Article PubMed Central CAS Google Scholar. Rizza, R. Cortisol-induced insulin resistance in man: impaired suppression of glucose production and stimulation of glucose utilization due to a postreceptor defect of insulin action.

Effects of growth hormone on insulin action in man: mechanisms of insulin resistance, impaired suppression of glucose production, and impaired stimulation of glucose utilization.

Diabetes 31 , — Barbour, L. A Cellular mechanisms for insulin resistance in normal pregnancy and gestational diabetes. Diabetes Care 30 , S—S Parichatikanond, W.

Prolonged stimulation of β2-adrenergic receptor with β2-agonists impairs insulin actions in H9c2 cells. Walli, R. Treatment with protease inhibitors associated with peripheral insulin resistance and impaired oral glucose tolerance in HIVinfected patients.

AIDS 12 , F—F Murata, H. The mechanism of insulin resistance caused by HIV protease inhibitor therapy. Teff, K. Antipsychotic-induced insulin resistance and postprandial hormonal dysregulation independent of weight gain or psychiatric disease.

Diabetes 62 , — Bittencourt, M. Insulin therapy in insulin resistance: could it be part of a lethal pathway? Elbein, S. Heritability of pancreatic beta-cell function among nondiabetic members of Caucasian familial type 2 diabetic kindreds.

Shulman, G. Cellular mechanisms of insulin resistance. Knauf, C. Brain glucagon-like peptide-1 increases insulin secretion and muscle insulin resistance to favor hepatic glycogen storage. Petersen, M. Regulation of hepatic glucose metabolism in health and disease.

Matsumoto, M. Dual role of transcription factor FoxO1 in controlling hepatic insulin sensitivity and lipid metabolism.

Shimomura, I. Cell 6 , 77—86 Petersen, K. Mechanism by which glucose and insulin inhibit net hepatic glycogenolysis in humans. Henriksen, E. Role of glycogen synthase kinase-3 in insulin resistance and type 2 diabetes. Drug Targets 7 , — Karim, S.

Hepatic expression and cellular distribution of the glucose transporter family. World J. Rencurel, F. Requirement of glucose metabolism for regulation of glucose transporter type 2 GLUT2 gene expression in liver. Thorens, B. Diabetologia 58 , — Eberlé, D.

SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 86 , — Horton, J. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Ferré, P. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c.

Diabetes Obes. Tobe, K. Dentin, R. 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 87 , 81—86 Hepatic glucokinase is required for the synergistic action of ChREBP and SREBP-1c on glycolytic and lipogenic gene expression. Herman, M. A novel ChREBP isoform in adipose tissue regulates systemic glucose metabolism.

Iizuka, K. Deficiency of carbohydrate response element-binding protein ChREBP reduces lipogenesis as well as glycolysis. Natl Acad.

Jaworski, K. Regulation of triglyceride metabolism. Hormonal regulation of lipolysis in adipose tissue. Liver Physiol. Vaughan, M. Hormone-sensitive lipase and monoglyceride lipase activities in adipose tissue.

Zmuda-Trzebiatowska, E. Role of PDE3B in insulin-induced glucose uptake, GLUT-4 translocation and lipogenesis in primary rat adipocytes. Cell Signal 18 , — Choi, Y. Alterations in regulation of energy homeostasis in cyclic nucleotide phosphodiesterase 3B—null mice.

Martinez-Botas, J. Genet 26 , — Tansey, J. Perilipin ablation results in a lean mouse with aberrant adipocyte lipolysis, enhanced leptin production, and resistance to diet-induced obesity. USA 98 , — Mechanisms of Insulin Action and Insulin Resistance.

Kimball, S. Regulation of protein synthesis by insulin. Pösö, A. Multifunctional control of amino acids of deprivation-induced proteolysis in liver. Role of leucine. Marshall, S. New insights into the metabolic regulation of insulin action and insulin resistance: role of glucose and amino acids.

Rudrappa, S. Human skeletal muscle disuse atrophy: effects on muscle protein synthesis, breakdown, and insulin resistance-a qualitative review.

Front Physiol. Medeiros, C. Antuna-Puente, B. Adipokines: the missing link between insulin resistance and obesity. Diabetes Metab. Rabe, K. Adipokines and insulin resistance. Adipokines mediate inflammation and insulin resistance. Lausanne 4 , 71 Li, S.

Adiponectin levels and risk of type 2 diabetes: a systematic review and meta-analysis. JAMA , — Hotta, K. 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.

Diabetes 50 , — Takahashi, M. Chemerin enhances insulin signaling and potentiates insulin-stimulated glucose uptake in 3T3-L1 adipocytes. FEBS Lett. Yamauchi, T. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions.

Li, L. Changes and relations of circulating visfatin, apelin, and resistin levels in normal, impaired glucose tolerance, and type 2 diabetic subjects.

Diabetes , — Soriguer, F. Apelin levels are increased in morbidly obese subjects with type 2 diabetes mellitus. Yue, P. Apelin is necessary for the maintenance of insulin sensitivity. American journal of physiology. Apelin decreases lipolysis via G q , G i , and AMPK-dependent mechanisms.

Endocrinology , 59—68 Segal, K. Relationship between insulin sensitivity and plasma leptin concentration in lean and obese men. Amitani, M. The role of leptin in the control of insulin-glucose axis. Front Neurosci. Article PubMed PubMed Central CAS Google Scholar.

Ceddia, R. Analysis of paradoxical observations on the association between leptin and insulin resistance. Covey, S. The pancreatic beta cell is a key site for mediating the effects of leptin on glucose homeostasis.

Seufert, J. Leptin effects on pancreatic beta-cell gene expression and function. Diabetes 53 , S—S Myers, M. Mechanisms of leptin action and leptin resistance. Burguera, B. Obesity is associated with a decreased leptin transport across the blood-brain barrier in rats.

Gainsford, T. Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells. USA 93 , — Scherer, P. Adipose tissue: from lipid storage compartment to endocrine organ.

Diabetes 55 , — Saponaro, C. The subtle balance between lipolysis and lipogenesis: a critical point in metabolic homeostasis. Nutrients 7 , — Frayn, K.

Adipose tissue and the insulin resistance syndrome. Roden, M. Mechanism of free fatty acid-induced insulin resistance in humans. Invest 97 , — Goossens, G. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. RANDLE, P. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus.

Lancet 1 , — Randle, P. The glucose fatty acid cycle in obesity and maturity onset diabetes mellitus. Y Acad. Brechtel, K. Fast elevation of the intramyocellular lipid content in the presence of circulating free fatty acids and hyperinsulinemia: a dynamic 1H-MRS study.

Reson Med 45 , — Boden, G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 46 , 3—10 Rothman, D. USA 92 , — Cline, G. Impaired glucose transport as a cause of decreased insulin-stimulated muscle glycogen synthesis in type 2 diabetes.

Dresner, A. Effects of free fatty acids on glucose transport and IRSassociated phosphatidylinositol 3-kinase activity. Yu, C. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 IRS-1 -associated phosphatidylinositol 3-kinase activity in muscle.

Erion, D. Diacylglycerol-mediated insulin resistance. Kim, J. PKC-theta knockout mice are protected from fat-induced insulin resistance. Nagai, Y. The role of peroxisome proliferator-activated receptor gamma coactivator-1 beta in the pathogenesis of fructose-induced insulin resistance.

Yen, C. The triacylglycerol synthesis enzyme DGAT1 also catalyzes the synthesis of diacylglycerols, waxes, and retinyl esters. Lipid Res. Timmers, S. Muscular diacylglycerol metabolism and insulin resistance.

Liu, L. Upregulation of myocellular DGAT1 augments triglyceride synthesis in skeletal muscle and protects against fat-induced insulin resistance. Sokolowska, E. The Role of Ceramides in Insulin Resistance. Lausanne 10 , Schubert, K. Chaurasia, B. Trends Endocrinol. Stiban, J. Ceramide synthases: roles in cell physiology and signaling.

Med Biol. Stöckli, J. Metabolomic analysis of insulin resistance across different mouse strains and diets. Blachnio-Zabielska, A. The crucial role of CCer in fat-induced skeletal muscle insulin resistance. Biochem 40 , — Raichur, S. CerS2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance.

Turpin, S. Obesity-induced CerS6-dependent C ceramide production promotes weight gain and glucose intolerance. Holland, W. An FGFadiponectin-ceramide axis controls energy expenditure and insulin action in mice. Mente, A. Causal relationship between adiponectin and metabolic traits: a Mendelian randomization study in a multiethnic population.

PLoS One 8 , e Brown, A. Genetics of Insulin Resistance and the Metabolic Syndrome. Chen, Z. Functional Screening of Candidate Causal Genes for Insulin Resistance in Human Preadipocytes and Adipocytes. Shakhanova, A. Association of polymorphism genes LPL , ADRB2 , AGT and AGTR1 with risk of hyperinsulinism and insulin resistance in the Kazakh population.

Henkin, L. Genetic epidemiology of insulin resistance and visceral adiposity. The IRAS Family Study design and methods. Parks, B. Genetic architecture of insulin resistance in the mouse. Xue, A. Genome-wide association analyses identify risk variants and putative regulatory mechanisms for type 2 diabetes.

Yousef, A. IRS-1 genetic polymorphism r. Appl Clin. Genet 11 , 99— Hashemian, L. The role of the PPARG Pro12Ala common genetic variant on type 2 diabetes mellitus risk. Zeng, Q. Association Between Insulin-like Growth Factor-1 rs Polymorphism and Type 2 Diabetes Mellitus Susceptibility: A Meta-Analysis.

Front Genet 12 , Knowles, J. Identification and validation of N-acetyltransferase 2 as an insulin sensitivity gene. Invest , Richards, J. A genome-wide association study reveals variants in ARL15 that influence adiponectin levels.

PLoS Genet 5 , e Eichler, E. Missing heritability and strategies for finding the underlying causes of complex disease. Genet 11 , — Flannick, J. Exome sequencing of 20, cases of type 2 diabetes and 24, controls.

Nature , 71—76 Viñuela, A. Genetic variant effects on gene expression in human pancreatic islets and their implications for T2D.

Metz, S. The Arg82Cys polymorphism of the protein nepmucin implies a role in HDL metabolism. Moltke, I. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes.

Huypens, P. Epigenetic germline inheritance of diet-induced obesity and insulin resistance. Genet 48 , — Ling, C. Epigenetics in human obesity and type 2 diabetes. Ahmed, S. The role of DNA methylation in the pathogenesis of type 2 diabetes mellitus.

Epigenetics 12 , 1—23 Cierzniak, A. DNA methylation in adipocytes from visceral and subcutaneous adipose tissue influences insulin-signaling gene expression in obese individuals. Int J. Epigenetic regulation of insulin action and secretion-role in the pathogenesis of type 2 diabetes.

Intern Med , — Zhao, J. Global DNA methylation is associated with insulin resistance: a monozygotic twin study. Zhou, Z. DNA methylation landscapes in the pathogenesis of type 2 diabetes mellitus.

DNA methylation Landsc. pathogenesis type 2 diabetes Mellit. Liu, J. An integrative cross-omics analysis of DNA methylation sites of glucose and insulin homeostasis. Commu 10 , 1—11 CAS Google Scholar. Gu, T. DNA methylation analysis of the insulin-like growth factor-1 IGF1 gene in Swedish men with normal glucose tolerance and type 2 diabetes.

Wittenbecher, C. Insulin-like growth factor binding protein 2 IGFBP-2 and the risk of developing type 2 diabetes. Diabetes 68 , — Haywood, N. Crujeiras, A. Genome-wide DNA methylation pattern in visceral adipose tissue differentiates insulin-resistant from insulin-sensitive obese subjects.

Res , 13—24 Ma, X. Functional implications of DNA methylation in adipose biology. Gao, H. Long non-coding RNAs associated with metabolic traits in human white adipose tissue. EBioMedicine 30 , — Altered adipose tissue DNA methylation status in metabolic syndrome: relationships between global DNA methylation and specific methylation at adipogenic, lipid metabolism and inflammatory candidate genes and metabolic variables.

Article CAS PubMed Central Google Scholar. Samblas, M. DNA methylation markers in obesity, metabolic syndrome, and weight loss. Epigenetics 14 , — Naidoo, V. Cell-and tissue-specific epigenetic changes associated with chronic inflammation in insulin resistance and type 2 diabetes mellitus.

Ali, M. DNA methylation profile of genes involved in inflammation and autoimmunity correlates with vascular function in morbidly obese adults. Epigenetics 17 , 93— Panchal, S.

DNA methylation in adipose tissue and metabolic syndrome. Liu J, Ibi D, Taniguchi K et al Inflammation improves glucose homeostasis through IKKbeta-XBP1s interaction. Cell 4 — e Raciti M, Lotti LV, Valia S, Pulcinelli FM, Di Renzo L JNK2 is activated during ER stress and promotes cell survival.

Cell Death Dis 3:e Cummings NE, Williams EM, Kasza I et al Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol 4 — 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 9 4 — Saad MJ, Santos A, Prada PO Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology 31 4 — Diabetologia 55 10 — Diabetes 64 12 — Thomou T, Mori MA, Dreyfuss JM et al Adipose-derived circulating miRNAs regulate gene expression in other tissues.

Ying W, Riopel M, Bandyopadhyay G et al Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity.

Cell 2 — e Bouzakri K, Roques M, Gual P et al Reduced activation of phosphatidylinositol-3 kinase and increased serine phosphorylation of insulin receptor substrate-1 in primary culture of skeletal muscle cells from patients with type 2 diabetes.

Diabetes 52 6 — Diabetologia 51 3 — Gaster M, Brusgaard K, Handberg A, Hojlund K, Wojtaszewski JF, Beck-Nielsen H The primary defect in glycogen synthase activity is not based on increased glycogen synthase kinase-3alpha activity in diabetic myotubes.

Biochem Biophys Res Commun 4 — Gaster M, Petersen I, Hojlund K, Poulsen P, Beck-Nielsen H The diabetic phenotype is conserved in myotubes established from diabetic subjects: evidence for primary defects in glucose transport and glycogen synthase activity. Diabetes 51 4 — McIntyre EA, Halse R, Yeaman SJ, Walker M Cultured muscle cells from insulin-resistant type 2 diabetes patients have impaired insulin, but normal 5-aminoimidazolecarboxamide riboside-stimulated, glucose uptake.

J Clin Endocrinol Metab 89 7 — Burkart AM, Tan K, Warren L et al Insulin resistance in human iPS cells reduces mitochondrial size and function. Sci Rep Iovino S, Burkart AM, Kriauciunas K et al Genetic insulin resistance is a potent regulator of gene expression and proliferation in human iPS cells.

Diabetes 63 12 — Iovino S, Burkart AM, Warren L, Patti ME, Kahn CR Myotubes derived from human-induced pluripotent stem cells mirror in vivo insulin resistance. Proc Natl Acad Sci U S A 7 — Gupta MK, Vethe H, Softic S et al Leptin receptor signaling regulates protein synthesis pathways and neuronal differentiation in pluripotent stem cells.

Stem Cell Rep 15 5 — Teo AK, Windmueller R, Johansson BB et al Derivation of human induced pluripotent stem cells from patients with maturity onset diabetes of the young. J Biol Chem 8 — Batista TM, Jayavelu AK, Wewer Albrechtsen NJ et al A cell-autonomous signature of dysregulated protein phosphorylation underlies muscle insulin resistance in type 2 diabetes.

Cell Metab — Taniguchi CM, Emanuelli B, Kahn CR Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol 7 2 — Barres R, Osler ME, Yan J et al Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density.

Cell Metab 10 3 — Ribel-Madsen R, Fraga MF, Jacobsen S et al Genome-wide analysis of DNA methylation differences in muscle and fat from monozygotic twins discordant for type 2 diabetes. PLoS One 7 12 :e Frobel J, Hemeda H, Lenz M et al Epigenetic rejuvenation of mesenchymal stromal cells derived from induced pluripotent stem cells.

Stem Cell Rep 3 3 — Vujkovic M, Keaton JM, Lynch JA et al Discovery of new risk loci for type 2 diabetes and related vascular outcomes among 1.

Nat Genet 52 7 — Miguel-Escalada I, Bonas-Guarch S, Cebola I et al Human pancreatic islet three-dimensional chromatin architecture provides insights into the genetics of type 2 diabetes.

Nat Genet 51 7 — Williams K, Ingerslev LR, Bork-Jensen J et al Skeletal muscle enhancer interactions identify genes controlling whole-body metabolism.

Nat Commun 11 1 Claussnitzer M, Dankel SN, Kim KH et al FTO obesity variant circuitry and adipocyte browning in humans. N Engl J Med 10 — Manning AK, Hivert MF, Scott RA et al A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance.

Nat Genet 44 6 — Nigi L, Grieco GE, Ventriglia G et al MicroRNAs as regulators of insulin signaling: research updates and potential therapeutic perspectives in type 2 diabetes. Int J Mol Sci 19 12 Article CAS PubMed Central Google Scholar.

Massart J, Sjogren RJO, Lundell LS et al Altered miR expression in type 2 diabetes influences glucose and lipid metabolism in skeletal muscle. Diabetes 66 7 — Katayama M, Wiklander OPB, Fritz T et al Circulating exosomal miRb-5p is elevated in type 2 diabetes and could impair insulin action in human skeletal muscle.

Mol Metab 6 7 — Yang L, Li P, Yang W et al Integrative transcriptome analyses of metabolic responses in mice define pivotal LncRNA metabolic regulators. Cell Metab 24 4 — Carter G, Miladinovic B, Patel AA, Deland L, Mastorides S, Patel NA Circulating long noncoding RNA GAS5 levels are correlated to prevalence of type 2 diabetes mellitus.

BBA Clin — Shi Y, Parag S, Patel R et al Stabilization of lncRNA GAS5 by a small molecule and its implications in diabetic adipocytes. Cell Chem Biol 26 3 — e Pihlajamaki J, Lerin C, Itkonen P et al Expression of the splicing factor gene SFRS10 is reduced in human obesity and contributes to enhanced lipogenesis.

Cell Metab 14 2 — Weyer C, Funahashi T, Tanaka S et al Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia.

J Clin Endocrinol Metab 86 5 — Lynes MD, Leiria LO, Lundh M et al The cold-induced lipokine 12,diHOME promotes fatty acid transport into brown adipose tissue. Nat Med 23 5 — Yore MM, Syed I, Moraes-Vieira PM et al Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects.

Cell 2 — Download references. Iacocca Professorship to CRK. The authors declare that there are no relationships or activities that might bias, or be perceived to bias, their work. Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.

Thiago M. Ronald Kahn. You can also search for this author in PubMed Google Scholar. TMB proposed the outline, conceptualised and designed figures and wrote the paper.

NH helped with outline and figure design and wrote the paper. CRK proposed the outline, conceptualised figures and wrote and edited the paper.

All authors approved the version to be published. Correspondence to C. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. The original online version of this article was revised: The funding statement was inadvertently omitted.

Reprints and permissions. Batista, T. Defining the underlying defect in insulin action in type 2 diabetes. Diabetologia 64 , — Download citation.

Received : 29 December Accepted : 29 January Published : 17 March Issue Date : May 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 Insulin resistance is one of the earliest defects in the pathogenesis of type 2 diabetes. Graphical abstract.

Insights into pancreatic islet cell dysfunction from type 2 diabetes mellitus genetics Article 25 February Mechanisms of Type 2 Diabetes Risk Loci Article 24 July Use our pre-submission checklist Avoid common mistakes on your manuscript.

Introduction The ground-breaking discovery of insulin years ago [ 1 ] turned diabetes from a death sentence into a manageable condition. Defining the insulin signalling network Although key components involved in insulin signal transduction are present in virtually every cell, the biological outcomes following activation or disruption of this pathway are highly dependent on the cell type and physiological context Fig.

Full size image. Insulin resistance as a central component of type 2 diabetes and the metabolic syndrome Type 2 diabetes affects more than million adults worldwide and its prevalence continues to increase at epidemic rates, thus posing one of the greatest public health challenges to society [ 57 ].

Extrinsic factors in the pathogenesis of insulin resistance In type 2 diabetes, most attention has focused on extrinsic factors contributing to insulin resistance, including the role of adipose tissue, circulating metabolites, inflammatory signals and the gut microbiome [ 66 , 67 , 68 ] Fig.

Intrinsic factors and cell-autonomous insulin resistance In vitro approaches, where cells are cultured under controlled conditions, provides an opportunity to minimise the influence of extrinsic factors and isolate cell-autonomous determinants of insulin resistance, which are more closely linked to the genetic and epigenetic alterations underlying type 2 diabetes.

Conclusions and perspectives Insulin and IGF-1 signalling is present in virtually every cell of the body and plays a central role in the control of metabolism, growth and differentiation.

References Macleod JJR Treatment of diabetes mellitus by pancreatic extracts. Can Med Assoc J 12 6 — Google Scholar Himsworth HP Diabetes mellitus: its differentiation into insulin-sensitive and insulin-insensitive types.

db Article CAS PubMed Google Scholar Perry RJ, Camporez JP, Kursawe R et al Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes.

M Article CAS PubMed Google Scholar Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. C Article CAS PubMed Google Scholar Humphrey SJ, Azimifar SB, Mann M High-throughput phosphoproteomics reveals in vivo insulin signaling dynamics.

Proc Natl Acad Sci U S A 92 4 — Article CAS PubMed PubMed Central Google Scholar Cline GW, Petersen KF, Krssak M et al Impaired glucose transport as a cause of decreased insulin-stimulated muscle glycogen synthesis in type 2 diabetes.

Annu Rev Immunol — Article Google Scholar Newgard CB Interplay between lipids and branched-chain amino acids in development of insulin resistance. Diabetes 51 7 — Article CAS PubMed Google Scholar Lyu K, Zhang Y, Zhang D et al A membrane-bound diacylglycerol species induces PKC-mediated hepatic insulin resistance.

M Article CAS PubMed Google Scholar Weigert C, Hennige AM, Brischmann T et al The phosphorylation of Ser of insulin receptor substrate 1 is not per se inhibitory in skeletal muscle cells but is necessary to trigger the attenuation of the insulin-stimulated signal.

J Biol Chem 45 — Article CAS PubMed Google Scholar Paz K, Hemi R, LeRoith D et al A molecular basis for insulin resistance. x Article CAS Google Scholar Liu J, Ibi D, Taniguchi K et al Inflammation improves glucose homeostasis through IKKbeta-XBP1s interaction.

Author information Authors and Affiliations Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA Thiago M. Ronald Kahn Authors Thiago M. Batista View author publications.

View author publications. Supplementary Information. Slideset of figures PPTX 1. Rights and permissions Reprints and permissions. About this article. Cite this article Batista, T. Copy to clipboard. search Search by keyword or author Search.

Amd Weight loss plateau Molecular Sensitivoty of Weight loss plateau Resistance: Serine Phosphorylation of Nutritional deficiencies Receptor Substrate-1 and Increased Expression of p85α : The Receptorrs Sides of a Coin. Diabetes 1 August ; 55 8 insuiln — Initial attempts to unravel the molecular mechanism of insulin resistance have strongly suggested that a defect responsible for insulin resistance in the majority of patients lies at the postreceptor level of insulin signaling. However, the nature of the triggering event s remains largely enigmatic. Two separate, but likely, complementary mechanisms have recently emerged as a potential explanation. First, it became apparent that serine phosphorylation of IRS proteins can reduce their ability to attract PI 3-kinase, thereby minimizing its activation. Weight gain for seniors you for visiting nature. You Insylin using a browser innsulin with Weight loss plateau support Weight loss plateau CSS. To obtain the best experience, we recommend you use 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. Insulin signaling controls cell growth and metabolic homeostasis.

Insulin sensitivity and insulin receptors -

Insulin, Blood Sugar, and Type 2 Diabetes Insulin is a key player in developing type 2 diabetes. Here are the high points: The food you eat is broken down into blood sugar. Blood sugar enters your bloodstream, which signals the pancreas to release insulin. Insulin also signals the liver to store blood sugar for later use.

Blood sugar enters cells, and levels in the bloodstream decrease, signaling insulin to decrease too. But this finely tuned system can quickly get out of whack, as follows: A lot of blood sugar enters the bloodstream. The pancreas pumps out more insulin to get blood sugar into cells.

The pancreas keeps making more insulin to try to make cells respond. Do You Have Insulin Resistance? What Causes Insulin Resistance? How to Reverse Insulin Resistance If you have insulin resistance, you want to become the opposite—more insulin sensitive cells are more effective at absorbing blood sugar so less insulin is needed.

Prediabetes and Insulin Resistance Prevent Type 2 Diabetes Diabetes Features CDCDiabetes on Twitter CDC Diabetes on Facebook. Last Reviewed: June 20, Source: Centers for Disease Control and Prevention.

Facebook Twitter LinkedIn Syndicate. Tm-treated iIR-KO mice experienced severe lipoatrophy because of loss of IR in adipose tissue and showed a mild increase of circulating free fatty acids FFAs compared with the healthy controls. Yet AAV-hIR was able to counter such minor increase and induce significant inhibition of lipolysis Supplemental Figure 3.

Protein quantification revealed that AAV-hIR injection led to a significant increase of IR expression only in the liver of Tm-treated iIR-KO mice Figure 2B , not in skeletal muscle Supplemental Figure 2B or eWAT Supplemental Figure 2C. The higher IR protein level in liver was associated with an elevation of insulin-stimulated hepatic Akt phosphorylation Figure 2C , indicating AAV-mediated IR expression enhanced downstream signal transduction.

Injection of AAV-hIR rescued diabetic phenotype in Tm-treated iIR-KO mice. A Blood glucose levels were measured 3 months after AAV-hIR or control virus injection at ad lib—fed condition. GAPDH was applied as an endogenous control.

Glucose AUCs of E and H were calculated according to data in D and G , respectively. Liver tissues were also subjected to F TG extraction and quantification. Blood was collected and subjected to I plasma TG measurement. Injection of AAV-hIR in corn oil vehicle—treated iIR-KO mice i.

Consistent with the stable IR protein levels, AAV-hIR administration did not significantly affect any of the metabolic parameters examined in the vehicle-treated iIR-KO mice Figure 2. These results demonstrated that liver-tropic IR restoration by AAV-hIR injection was sufficient to rescue the diabetic phenotype of Tm-treated iIR-KO mice, which have IR deficiency in multiple tissues.

This approach works only under pathologically diabetic conditions and has minimal impact in nondiabetic mice. IR protein level reduction in insulin-sensitive tissues of diabetic animal models. IR restoration rescued the diabetic phenotype in Tm-treated iIR-KO mice and provided proof of concept about the potential therapeutic value of AAV-hIR in animals with impaired IR.

Whole-tissue lysates were prepared from the liver, skeletal muscle, and eWAT of these animals. Meanwhile, membrane-bound proteins were extracted from the same samples Figure 3A.

IR protein levels were quantified by Western blotting using both the whole-tissue lysates and the membrane protein fractions. Studies in other models, such as diet-induced obese DIO mice, yielded similar results Supplemental Figure 4.

Collectively, these observations indicated that the reduction of IR level is a common phenomenon and may be the unifying pathophysiology underlying the defects of multiple T2DM models. Membrane-bound fraction was extracted from whole-tissue lysate A. IR levels were analyzed by Western blot using samples from B whole-tissue lysate or C membrane-bound fraction, respectively.

Data presented as mean ± SEM. These parameters were significantly reduced e. Injection of AAV-hIR rescued diabetic phenotype in mice. Glucose AUCs F and G were calculated according to data in D and E , respectively. To assess AAV-hIR—induced IR expression, we analyzed IR protein levels in insulin-sensitive tissues, including liver, skeletal muscle, and eWAT.

The normalization of IR protein level improved downstream insulin signaling, as indicated by the enhancement of insulin-stimulated Akt phosphorylation Figure 5C.

Corresponding to improved Akt phosphorylation, the expression of sterol regulatory element—binding protein 1 SREBP1 a key gene that regulates hepatic de novo lipogenesis [DNL] was significantly elevated Supplemental Figure 6. In line with the WT mouse results described above e. A Liver tissues were collected and whole-tissue lysates were subjected to Western blot for protein level analysis.

insulin injection. The ratio of phosphorylated Akt to total Akt was calculated according to Western blot results. The insulin signaling pathway participates in critical physiological functions, including cell proliferation, differentiation, growth, and metabolism 7.

IR is the first step in this signaling cascade. Its important role in development is supported by the observation that whole-body deletion of IR gives rise to early postnatal death from diabetic ketoacidosis The scientific debate has been ongoing regarding the primary site of defect leading to insulin resistance, i.

It has been well documented that signaling events, such as phosphorylation of IR, IRS, PI3K, and Akt, as well as GLUT4-mediated glucose uptake, all play crucial roles in the regulation of energy metabolism, yet the primary node of insulin resistance thus the effective targeting strategy has not been mapped out Although deletion of FoxO1 in the liver rescued systematic glucose and insulin sensitivity in the absence of the hepatic IR, the normal state of glucose metabolism requires intact function of IR and insulin signaling from other tissues 20 , which indicates the importance of IR in the whole-body metabolism.

Supporting the vital role of IR in this process, aberrant IR content and kinase activity have been reported in human subjects with obesity and diabetes 9 , 10 , 21 , 22 , which may be attributed to chronic hyperinsulinemia-induced IR downregulation 23 — Similar observations have been made in other T2DM animals with insulin resistance.

This suggests that downregulation of IR protein level may be a common mechanism contributing to the dysregulated glucose and lipid metabolism in these animals. Enhancing IR expression in different tissues via transgenic approaches has been attempted previously, the results of which were supportive of potential corrective value of restoring IR expression.

However, such an approach has not been considered as a therapeutic method because in many of those studies animals bearing IR transgene did not achieve normal IR expression and were only partially rescued from diabetes, in addition to having potentially higher developmental risks 11 , 13 , 14 , 26 , Thus, these studies did not provide definitive conclusions on the therapeutic value of restoring IR in adult-onset diabetes as a potential cure.

In the present study, AAV9 and a truncated chicken β-actin promoter with CMV enhancer were used to deliver hIR to achieve an adequate efficiency of IR expression. AAVs drive long-lasting protein expression and cause low levels of immune response AAV9 surpasses other serotypes of AAVs because of its broad genome distribution and high level of expression CB promoter has been shown to deliver potent gene expression in a ubiquitous fashion Restricted by the large size of the IR sequence and the limited AAV packaging capacity, a short CB promoter without the intron was applied.

The iIR-KO mouse, a model of insulin resistance due to IR deletion in multiple insulin-sensitive tissues in adult mice, was tested in the present study. This proof-of-principle experiment of IR restoration in the diabetic iIR-KO mouse by AAV delivery provided evidence about the effectiveness, tissue specificity, and safety of the current strategy.

The successful rescue in these mice by AAV-hIR provides strong support of the current approach to treat type 2 diabetics. Although IR is widely expressed, results from the tissue-specific KO mouse study showed that loss of IR in the liver led to severer systematic phenotype of diabetes 30 compared with IR deficiency in other tissues 31 — This is in agreement with data supporting the liver as the primary organ of insulin action responsible for glucose and lipid metabolism 38 , The liver-tropic IR expression may be attributed to the modified CB promoter applied in the current study, which limited viral expression in tissues other than liver.

In line with these findings, restoration of IR in the liver, but not in other tissues, by a transgenic approach achieved the most robust effects in rescuing the diabetic phenotype and extending the life span of iIR-KO mice 11 — 13 , These results support a prevailing role of hepatic IR over that of other tissues in the pathology of T2DM and highlight the importance of restoring IR-mediated liver metabolism via gene therapy.

In the liver, insulin suppresses hepatic glucose production and promotes lipid synthesis There has been a concern that enhancing liver insulin signaling may exacerbate the hypertriglyceridemia observed in animal models and patients with T2DM.

Corresponding to the enhanced Akt phosphorylation, AAV-hIR led to increased SREBP1 expression, which indicates upregulated hepatic DNL. This is possibly due to reduced influx of circulating FFAs. The liver adapts 3 major lipid sources: esterified fatty acid from TG hydrolysis, nonesterified fatty acid by adipocyte lipolysis, and DNL synthesized by hepatocytes As the main source of liver TGs, circulating FFAs primarily come from adipose lipolysis Although AAV-hIR induced liver-tropic IR expression, it improved both hepatic and systematic insulin sensitivity, which should alleviate the insulin resistance in eWAT and slow down the FFA influx into the liver as indicated by the normalization of circulating FFA levels Figure 4J.

These data demonstrated the correlation between glycemia improvement and adipocyte lipolysis inhibition. The decrease of ectopic lipid accumulation mitigates inflammation in liver and forms a positive feedback loop to improve insulin sensitivity A recent study provided evidence that increased liver lipid accumulation may be attributable to insulin resistance of the peripheral tissues 44 because hepatic TG synthesis is more dependent on increased plasma fatty acid esterification than hepatic insulin signaling Insulin promotes hepatic TG synthesis and inhibits lipolysis in WAT.

This suggests, in the current models, the inhibitory role of lipolysis in WAT prevails over hepatic TG synthesis with regard to hepatic TG regulation by insulin signaling. Services Alert me when this article is cited Alert me if a correction is posted Similar articles in this journal Similar articles in Web of Science Similar articles in PubMed Download to citation manager Permissions.

Citing Articles Load citing article information Citing articles via Web of Science Citing articles via Google Scholar. Google Scholar Articles by Boucher, J. Articles by Kahn, C. Search for related content. Subject Collections Signaling by Receptor Tyrosine Kinases.

Share CiteULike Delicious Digg Facebook Reddit Twitter What's this? Interview Click to see an interview with subject collection editor Tom Misteli. Interview Click to see an interview with subject collection editor Tom Cech. Interview Click to see an interview with subject collection editor Lucy Shapiro.

Interview Click to see an interview with subject collection editor Paolo Sassone-Corsi. Interview Click to see an interview with subject collection editor Richard Morimoto.

Invisible changes in the body begin long receptosr a person is diagnosed receptogs Weight loss plateau 2 diabetes. One of the most important unseen changes? Insulin resistance. Insulin is a key player in developing type 2 diabetes. Here are the high points:.

Author: Zulkizahn

5 thoughts on “Insulin sensitivity and insulin receptors

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com