Category: Moms

Metabolism and insulin sensitivity

Metabolism and insulin sensitivity

Sensigivity 7— Hoogeveen RC, Inzulin JW, Sun W, Dodge RC, Crosby JR, Jiang Sensitifity, Couper D, Virani SS, Kathiresan S, Boerwinkle Sensitovity, Recovery nutrition strategies for endurance athletes al. Bonora E, Kiechl S, Willeit J, Oberhollenzer F, Egger G, Targher G, Alberiche M, Bonadonna RC, Muggeo M. Dysregulated lipid storage and its relationship with insulin resistance and cardiovascular risk factors in non-obese asian patients with type 2 diabetes. Valenti L, Bugianesi E, Pajvani U, Targher G. Chemistry of insulin.

Insullin Yoshino, Paloma Almeda-Valdes, Bruce W. Patterson, Adewole L. The mechanism Metabolusm responsible for diurnal variations in insulin sensitivity of glucose metabolism Metaabolism healthy people are Metabooism.

The objective of the study was insuljn evaluate whether Antioxidant-rich chia seeds variations in whole-body and cellular fatty acid metabolism Metabolizm contribute to evening Metabplism resistance in metabolically normal people.

We measured plasma nisulin free fatty acid FFA concentration, palmitate ajd, and skeletal muscle Metbolism of genes sensiyivity in fatty acid metabolism at insjlin am and dinner unsulin in 13 overweight body mass sebsitivity Sensiitvity, adipose tissue lipolytic activity was not different in the evening Metabklism in the morning.

Metabolically normal Nutrient timing for recovery nutrition demonstrate diurnal variations in fatty acid metabolism, manifested sensitivitty an increase in insulon FFAs, presumably derived from previous meal consumption rather than lipolysis of adipose tissue triglycerides, and Metabolizm shift Metabollism muscle Meabolism acid metabolism Healthy fat burning oxidation zensitivity lipogenesis.

These metabolic alterations could be responsible for the known evening decline in insulin sensitivity. Many metabolic pathways sensitivjty functions vary isulin to the time insylin day 1.

In healthy sensiivity, insulin sensitivity with respect to glucose metabolism is lower in the evening than in the morning sesnitivity — 4. Consequently, sensitlvity or meal ingestion results Metabolims a greater Autophagy and LC in senxitivity glucose concentration in sensitovity evening than in the morning 3 — 5.

The mechanism s responsible isulin diurnal variation in senaitivity action is not known but could be Recovery nutrition strategies for endurance athletes to alterations in Arthritis supplements and vitamins free fatty acid EMtabolism availability and muscle fatty acid metabolism.

Increased FFA availability from plasma can cause insulin Metabklism in skeletal Metabolizm 6. Anr, clock genes, which regulate circadian rhythm, could contribute to diurnal variation in muscle insulin Diabetes and eye health because they have been shown to regulate Mrtabolism sensitivity and fatty acid metabolic pathways in rodent models 1.

The purpose of the present Metabolism and insulin sensitivity was ineulin test the hypothesis that diurnal variations Metabolksm clock gene expression, Metablism FFA availability, Glucose storage muscle fatty acid metabolism are associated with diurnal Metaabolism in insulin-mediated glucose metabolism.

Accordingly, we evaluated the effect of consuming identical breakfast and Metaboliwm meals on plasma glucose, indulin, and FFA Energy drinks for busy professionals, adipose sensitivit lipolytic rate, and sensktivity variation in jnsulin of genes associated with Metabolism and insulin sensitivity tissue lipolytic activity and insuln muscle insullin acid metabolism in metabolically normal women.

Thirteen women participated in this study Supplemental Inxulin 1. All subjects were considered to be metabolically normal, based on a history and physical examination, an oral glucose tolerance test, and one or fewer metabolic syndrome criteria 7. Written informed an was obtained before Recovery nutrition strategies for endurance athletes subjects senditivity in the study, which was approved by the Institutional Review Board of Metabolism and insulin sensitivity Sensitiity School of Medicine.

Total body fat mass insjlin fat-free mass FFM were determined by using dual-energy X-ray absorptiometry. Intraabdominal adipose tissue volume and intrahepatic triglyceride content were quantified by using magnetic amd imaging senistivity magnetic resonance spectroscopy 8.

Resting energy expenditure was determined by measuring the imsulin gas exchange TrueOne ; ParvoMedicsand Fat metabolism pills Recovery nutrition strategies for endurance athletes daily energy requirement was Metabolism and insulin sensitivity as 1.

At pmWebsite performance improvement were Metaboliism into ssensitivity antecubital Injury rehab nutrition guide for palmitate innsulin infusion and a contralateral radial artery Blood pressure monitor blood sampling.

Nad consumed anx identical liquid mixed meals at am breakfast sensitivuty, pm lunchand pm dinner. Subjects rested in bed to avoid the influence of physical activity on Performance nutrition tips outcome measures.

Blood samples were obtained 10 minutes and immediately before and at 20, 40, 60, sesitivity,Fat distribution and body positivity,and eensitivity after starting breakfast and insylin. Subcutaneous insluin adipose tissue and Anti-cancer news muscle vastus insuiln biopsies were Metabollsm at Metabolisj and pmas described previously The second fat biopsy Metabbolism Metabolism and insulin sensitivity from the opposite side anf the senssitivity of the first biopsy in sfnsitivity 13 subjects.

The sensitivify muscle biopsy Recovery nutrition strategies for endurance athletes obtained from the opposite Weight loss and diabetes management of the first biopsy data were ahd for six subjects.

Plasma glucose, insulin, Metabolissm FFA concentrations and palmitate tracer to tracee sensktivity were determined Protein supplements for athletes previously described Meetabolism cortisol was measured by using an immunoassay Elecsys; Roche Diagnostics GmbH.

Gene expression insulun determined by using Metaboliem PCR primer insuoin in Supplemental Table 3 based on their cycle threshold CT values relative to glyceraldehydephosphate dehydrogenase GAPDHas previously described Plasma substrate and hormone concentration total areas under the curve AUCs and incremental AUC iAUC from baseline values before and for 4 hours after breakfast and dinner were calculated by using the trapezoid method.

The palmitate rate of appearance Ra of total FFA Ra in plasma was calculated as previously described The differences between single values obtained at breakfast and dinner were evaluated by using the paired Student's t test.

Repeated-measures ANOVA was used to compare changes in substrate kinetics and concentrations induced by breakfast and dinner. Results are presented as means ± SD, unless otherwise stated. Plasma glucose iAUC Figure 1 A and total AUC These findings suggest that insulin sensitivity with respect to glucose metabolism was lower in the evening than in the morning, which is consistent with results from previous studies conducted in healthy people 2 — 4.

Plasma glucose iAUC after breakfast am and dinner pm A. Plasma FFA concentration after breakfast and dinner Bthe percentage of total plasma FFA as C palmitate and C oleate right before breakfast and dinner Cthe FFA Ra Dand palmitate Ra E after breakfast and dinner, and adipose tissue gene expression of ATGL and HSL at am before breakfast and pm before dinner F are shown.

Expression of ATGL and HSL was normalized to GAPDH expression. Data are means ± SEM. The contribution of C palmitate to total FFA concentration decreased from Meal ingestion rapidly reduced plasma FFA concentration by approximately fold after both breakfast and dinner, but plasma FFA concentrations Figure 1 B and the FFA AUC 0.

Adipose tissue triglyceride lipase ATGL was the same, and hormone sensitive lipase HSL was slightly lower at pm before dinner than at am before breakfast Figure 1 F. Expression of genes involved in muscle fatty acid oxidation [pyruvate dehydrogenase kinase 4 PDK4uncoupling protein-3 UCP3and carnitine palmitoyltransferase 1A CPT1A ] were lower at pm than at am Figure 2 A.

In contrast, the expressions of genes involved in de novo lipogenesis [sterol regulatory element binding protein-1c SREBP-1c and fatty acid synthase FAS ] were greater at pm than at am Figure 2 B.

We also found diurnal variations in the core clock genes circadian locomotor output cycles kaput CLOCKbrain, and muscle Arnt-like protein 1 BMAL1period 1 and 2 PER1 and PER2cryptochrome 1 CRY1and D site of albumin promoter albumin D-box binding protein DBP in skeletal muscle Figure 2 C.

CRY2 expression in the morning was not different from that in the evening data not shown. The expression of genes of interest was normalized to GAPDH expression. Although insulin sensitivity and glucose tolerance are often worse in the evening than in the morning in healthy people 2 — 5the mechanism s responsible for diurnal variation in glucose homeostasis are not clear.

We investigated whether there are diurnal variations in fatty acid metabolism that could contribute to this phenomenon in metabolically normal women and found that insulin resistance with respect to glucose metabolism in the evening was accompanied by increased plasma FFA availability.

The increased FFA availability was likely derived from the hydrolysis of chylomicron triglycerides from previous meals, not from an increase in adipose tissue lipolytic activity, because palmitate Ra and adipose tissue gene expression of lipolytic enzymes were the same or lower in the evening than in the morning.

In addition, the percentage of total plasma FFAs as palmitate was lower at dinner than breakfast, and the percentage of total plasma FFAs as oleate was higher at dinner than breakfast, suggesting an increased contribution of FFAs from ingested meals, which contained predominantly oleate and little palmitate.

These data demonstrate a plausible mechanism for a decrease in insulin sensitivity in the evening in healthy people because an increase in circulating FFAs can cause insulin resistance 6. We also found a diurnal variation in skeletal muscle expression of genes involved in regulating fatty acid metabolism; the expression of genes that regulate fatty acid oxidation was lower, whereas the expression of genes involved in de novo lipogenesis was higher, at pm before dinner than at am before breakfast.

These data suggest a shift from muscle fatty acid oxidation toward lipogenesis in the evening, which could lead to insulin resistance by producing specific fatty acid metabolites that impair insulin action The mechanism s responsible for this diurnal variability is not clear but could be related to the expression of core clock genes, which oscillate in adipose tissue and muscle in people 16 — 18 and regulate fatty acid metabolic pathways 1 It is also possible that the differences in the duration of fasting before breakfast 12 h fast and dinner 6.

Nonetheless, our data represent the normal diurnal variations in metabolic pathways in people consuming a typical daily meal pattern. However, the morning-to-evening direction of the variation in muscle clock gene expression in people is opposite from the direction observed in nocturnal mice 1719 Taken together, the data from our study and previous studies conducted in people and rodents support the notion that the core molecular clock machinery is involved in regulating both diurnal variations in fatty acid metabolism and insulin action.

In conclusion, the present study demonstrates that insulin resistance in the evening is associated with both an increase in circulating FFAs and alterations in cellular metabolic pathways associated with skeletal muscle fatty acid metabolism and core clock genes in metabolically normal women.

However, our study is not able to prove a direct cause-and-effect relationship between diurnal variations in fatty acid metabolism and insulin resistance. Further studies are needed to evaluate the complex mechanistic relationships among clock genes and metabolic pathways in people.

We thank Martha Hessler for help with subject recruitment; Janine Kampelman, Jennifer Shew, Freida Custodio, Anna C. Moseley, Kelly L. Stromsdorfer, and Ioana Gruchevska for technical assistance; the staff of the Clinical Research Unit for their help in performing the studies; and the study subjects for their participation.

This study was registered at clinicaltrials. gov as trial number NCT This study was supported by National Institutes of Health Grants DK and DK to the Washington University School of Medicine Nutrition Obesity Research CenterGrant DK to the Washington University School of Medicine Diabetes Research CenterGrant RR to the Washington University Biomedical Mass Spectrometry ResourceGrant UL1 TR to the Washington University School of Medicine Clinical Translational Science Award including KL2 Subaward TR, and the Central Society for Clinical and Translational Research Early Career Development Award.

Disclosure Summary: S. is a shareholder and consultant for Aspire Bariatrics and serves on the Scientific Advisory Boards for NovoNordisk, Takeda Pharmaceuticals, the Egg Nutrition Council, and NuSi.

The other authors have nothing to declare. Maury ERamsey KMBass J. Circadian rhythms and metabolic syndrome: from experimental genetics to human disease. Circ Res. Google Scholar. Morgan LMAspostolakou FWright JGama R. Diurnal variations in peripheral insulin resistance and plasma non-esterified fatty acid concentrations: a possible link?

Ann Clin Biochem. Saad ADalla Man Cet al. Diurnal pattern to insulin secretion and insulin action in healthy individuals. Lee AAder MBray GABergman RN. Diurnal variation in glucose tolerance. Cyclic suppression of insulin action and insulin secretion in normal-weight, but not obese, subjects.

Van Cauter EShapiro ETTillil HPolonsky KS. Circadian modulation of glucose and insulin responses to meals: relationship to cortisol rhythm.

Am J Physiol. Roden MPrice TBPerseghin Get al. Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest. Third Report of the National Cholesterol Education Program NCEP Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults Adult Treatment Panel III final report.

Frimel TNDeivanayagam SBashir AO'Connor RKlein S. Assessment of intrahepatic triglyceride content using magnetic resonance spectroscopy. J Cardiometab Syndr.

James WPMcNeill GRalph A. Metabolism and nutritional adaptation to altered intakes of energy substrates.

: Metabolism and insulin sensitivity

REVIEW article If you know you have at least one component Recovery nutrition strategies for endurance athletes metabolic ssnsitivity, ask annd doctor whether you need testing for other components of the syndrome. Metabolic syndrome: focus on dyslipidemia. Role of microRNAs in obesity and obesity-related diseases. Try these simple, delicious recipes for breakfast, lunch, and…. Insulin resistance, hyperglycemia, and atherosclerosis.
Understanding Insulin Resistance

Research has identified at least compounds contained in a variety of herbs and spices that may contribute to reducing insulin resistance Several studies have found that drinking green tea can help increase insulin sensitivity and reduce blood sugar 27 , These beneficial effects of green tea could be due to its powerful antioxidant epigallocatechin gallate EGCG , which helps increase insulin sensitivity Vinegar could help increase insulin sensitivity by reducing blood sugar and improving the effectiveness of insulin It also appears to delay the stomach from releasing food into the intestines, giving the body more time to absorb sugar into the bloodstream Unlike other fats, trans fats provide no health benefits and increase the risk of many diseases Evidence on the effects of high trans-fat intake on insulin resistance appears to be mixed.

Some human studies have found it harmful, while others have not 33 , Many different supplements can help increase insulin sensitivity, including vitamin C , probiotics , and magnesium. That said, many other supplements, such as zinc, folate, and vitamin D, do not appear to have this effect, according to research As with all supplements, there is a risk they may interact with any current medication you may be taking.

Insulin is an important hormone that has many roles in the body. When your insulin sensitivity is low, it puts pressure on your pancreas to increase insulin production to clear sugar from your blood. Low insulin sensitivity is also called insulin resistance.

Insulin sensitivity describes how your cells respond to insulin. Symptoms develop when your cells are resistant to insulin.

Insulin resistance can result in chronically high blood sugar levels, which are thought to increase your risk of many diseases, including diabetes and heart disease. Insulin resistance is bad for your health, but having increased insulin sensitivity is good.

It means your cells are responding to insulin in a healthier way, which reduces your chance of developing diabetes. Consider trying some of the suggestions in this article to help increase your insulin sensitivity and lower your risk of disease but be sure to talk with a healthcare professional first before making changes, especially adding supplements to your treatment regimen.

Read this article in Spanish. Our experts continually monitor the health and wellness space, and we update our articles when new information becomes available. VIEW ALL HISTORY. This article is based on scientific evidence, written by experts and fact checked by experts.

Our team of licensed nutritionists and dietitians strive to be objective, unbiased, honest and to present both sides of the argument. This article contains scientific references.

The numbers in the parentheses 1, 2, 3 are clickable links to peer-reviewed scientific papers. Has taking insulin led to weight gain for you? Learn why this happens, plus how you can manage your weight once you've started insulin treatment.

When it comes to managing diabetes, adding the right superfoods to your diet is key. Try these simple, delicious recipes for breakfast, lunch, and…. A Quiz for Teens Are You a Workaholic?

How Well Do You Sleep? Health Conditions Discover Plan Connect. Nutrition Evidence Based Top Natural Ways to Improve Your Insulin Sensitivity. Medically reviewed by Kelly Wood, MD — By Ryan Raman, MS, RD — Updated on October 30, Get more sleep. Exercise more. Explore our top resources.

Reduce stress. Lose a few pounds. Discover more about Type 2 Diabetes. Eat health-promoting foods. Frequently asked questions. The bottom line. How we reviewed this article: History.

Oct 30, Written By Ryan Raman. Sep 18, Medically Reviewed By Kelly Wood, MD. Circulating palmitoleic acid and risk of metabolic abnormalities and new-onset diabetes. Nestel, P. Effects of increasing dietary palmitoleic acid compared with palmitic and oleic acids on plasma lipids of hypercholesterolemic men.

Yore, M. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. This study reports the discovery of a novel class of lipids, branched FAHFAs, and shows that FAHFAs have anti-diabetic and anti-inflammatory effects.

Ma, Y. Kuda, O. Docosahexaenoic acid-derived fatty acid esters of hydroxy fatty acids FAHFAs with anti-inflammatory properties. Diabetes 65 , — Syed, I. Palmitic acid hydroxystearic acids activate GPR40, which is involved in their beneficial effects on glucose homeostasis.

e4 Article CAS PubMed PubMed Central Google Scholar. Lee, J. Branched fatty acid esters of hydroxy fatty acids FAHFAs protect against colitis by regulating gut innate and adaptive immune responses.

Parsons, W. AIG1 and ADTRP are atypical integral membrane hydrolases that degrade bioactive FAHFAs. Kolar, M. Branched fatty acid esters of hydroxy fatty acids are preferred substrates of the MODY8 protein carboxyl ester lipase. Biochemistry 55 , — Raeder, H. Mutations in the CEL VNTR cause a syndrome of diabetes and pancreatic exocrine dysfunction.

Nelson, A. Stereochemistry of endogenous palmitic acid ester of 9-hydroxystearic acid and relevance of absolute configuration to regulation. Shulman, G. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease.

This is a comprehensive review on the roles of ectopic lipids, especially DAGs, in insulin resistance, dyslipidaemia and cardiometabolic disease. Erion, D. Diacylglycerol-mediated insulin resistance. Samuel, V. Lipid-induced insulin resistance: unravelling the mechanism.

Lancet , — Szendroedi, J. Role of diacylglycerol activation of PKCtheta in lipid-induced muscle insulin resistance in humans. Natl Acad. USA , — Yu, C. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 IRS-1 -associated phosphatidylinositol 3-kinase activity in muscle.

Kim, J. PKC-theta knockout mice are protected from fat-induced insulin resistance. Li, Y. Protein kinase C Theta inhibits insulin signaling by phosphorylating IRS1 at Ser Petersen, M. Insulin receptor Thr phosphorylation mediates lipid-induced hepatic insulin resistance.

Finck, B. Does diacylglycerol accumulation in fatty liver disease cause hepatic insulin resistance? This review article discusses the controversies on the roles of DAGs in insulin resistance.

Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. Ferris, H. Unraveling the paradox of selective insulin resistance in the liver: the brain-liver connection. Brown, J.

CGI knockdown in mice causes hepatic steatosis but prevents diet-induced obesity and glucose intolerance. Turpin, S. Adipose triacylglycerol lipase is a major regulator of hepatic lipid metabolism but not insulin sensitivity in mice.

Diabetologia 54 , — Amati, F. Skeletal muscle triglycerides, diacylglycerols, and ceramides in insulin resistance: another paradox in endurance-trained athletes? Diabetes 60 , — Kishimoto, A. Activation of calcium and phospholipid-dependent protein kinase by diacylglycerol, its possible relation to phosphatidylinositol turnover.

Hannun, Y. Sphingolipids and their metabolism in physiology and disease. Park, J. Ceramide synthases as potential targets for therapeutic intervention in human diseases. Acta , — Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin.

Peraldi, P. Tumor necrosis factor TNF -alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase.

A ceramide-centric view of insulin resistance. Ussher, J. Inhibition of de novo ceramide synthesis reverses diet-induced insulin resistance and enhances whole-body oxygen consumption.

Diabetes 59 , — Deevska, G. Acid sphingomyelinase deficiency prevents diet-induced hepatic triacylglycerol accumulation and hyperglycemia in mice.

Blouin, C. Plasma membrane subdomain compartmentalization contributes to distinct mechanisms of ceramide action on insulin signaling.

Chalfant, C. Long chain ceramides activate protein phosphatase-1 and protein phosphatase-2A. Activation is stereospecific and regulated by phosphatidic acid.

Bourbon, N. Ceramide-induced inhibition of Akt is mediated through protein kinase Czeta: implications for growth arrest.

Hajduch, E. Targeting of PKCzeta and PKB to caveolin-enriched microdomains represents a crucial step underpinning the disruption in PKB-directed signalling by ceramide.

Powell, D. Roles of diacylglycerols and ceramides in hepatic insulin resistance. Trends Pharmacol. Merrill, A. Jr Sphingolipid and glycosphingolipid metabolic pathways in the era of sphingolipidomics.

Bergman, B. Muscle sphingolipids during rest and exercise: a C signature for insulin resistance in humans.

Diabetologia 59 , — Chung, J. Intramyocellular ceramides: subcellular concentrations and fractional de novo synthesis in postabsorptive humans. Diabetes 66 , — Montgomery, M. Regulation of glucose homeostasis and insulin action by ceramide acyl-chain length: a beneficial role for very long-chain sphingolipid species.

Raichur, S. CerS2 haploinsufficiency inhibits beta-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance.

Obesity-induced CerS6-dependent C ceramide production promotes weight gain and glucose intolerance. Randle, P. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus.

Lancet 1 , — Goodpaster, B. Metabolic flexibility in health and disease. This comprehensive review discusses the mechanisms for insulin resistance induced by metabolic inflexibility in muscle and adipose tissue. Kelley, D. Skeletal muscle fatty acid metabolism in association with insulin resistance, obesity, and weight loss.

Fuel selection in human skeletal muscle in insulin resistance: a reexamination. Diabetes 49 , — References and show the evidence for and describe the concept of metabolic inflexibility in the development of insulin resistance.

Guilherme, A. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Perry, R. Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes.

This study shows that adipose inflammation-stimulated lipolysis increases the influx of acetyl-CoA into the liver, which activates pyruvate carboxylase and promotes hepatic gluconeogenesis, leading to hyperglycaemia.

This occurs in insulin-resistant states such as obesity and T2DM. Zhang, H. Tumor necrosis factor-alpha stimulates lipolysis in differentiated human adipocytes through activation of extracellular signal-related kinase and elevation of intracellular cAMP.

Diabetes 51 , — Grant, R. Fat in flames: influence of cytokines and pattern recognition receptors on adipocyte lipolysis. Adipocyte-specific overexpression of retinol-binding protein 4 causes hepatic steatosis in mice.

Hepatology 64 , — Yang, Q. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Morigny, P. Adipocyte lipolysis and insulin resistance. Biochimie , — Aguer, C. Acylcarnitines: potential implications for skeletal muscle insulin resistance.

Koves, T. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. This paper provides evidence of increased incomplete β-oxidation of lipids in states of insulin resistance and obesity, resulting in the accumulation of acylcarnitines.

Muoio, D. Lipid-induced mitochondrial stress and insulin action in muscle. Muscle-specific deletion of carnitine acetyltransferase compromises glucose tolerance and metabolic flexibility. Liepinsh, E. Decreased acylcarnitine content improves insulin sensitivity in experimental mice models of insulin resistance.

Bene, J. Role of carnitine and its derivatives in the development and management of type 2 diabetes. Diabetes 8 , 8 Nurjhan, N. Increased lipolysis and its consequences on gluconeogenesis in non-insulin-dependent diabetes mellitus.

Best, C. The effects of cholesterol and choline on liver fat. Li, Z. Phosphatidylcholine and choline homeostasis. Raubenheimer, P. A choline-deficient diet exacerbates fatty liver but attenuates insulin resistance and glucose intolerance in mice fed a high-fat diet.

Diabetes 55 , — Meikle, P. Sphingolipids and phospholipids in insulin resistance and related metabolic disorders. van der Veen, J. The critical role of phosphatidylcholine and phosphatidylethanolamine metabolism in health and disease.

Article CAS Google Scholar. A nuclear-receptor-dependent phosphatidylcholine pathway with antidiabetic effects. Liu, S. A diurnal serum lipid integrates hepatic lipogenesis and peripheral fatty acid use. Rong, X. LXRs regulate ER stress and inflammation through dynamic modulation of membrane phospholipid composition.

Singh, A. Identification of hepatic lysophosphatidylcholine acyltransferase 3 as a novel target gene regulated by peroxisome proliferator-activated receptor delta. Cash, J. Liver-specific overexpression of LPCAT3 reduces postprandial hyperglycemia and improves lipoprotein metabolic profile in mice.

Diabetes 6 , e Drazic, A. The world of protein acetylation. Menzies, K. Protein acetylation in metabolism — metabolites and cofactors.

Moussaieff, A. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Akt-dependent metabolic reprogramming regulates tumor cell histone acetylation. Donohoe, D. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation.

Cell 48 , — Cai, L. Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes.

Cell 42 , — Wellen, K. ATP-citrate lyase links cellular metabolism to histone acetylation. Carrer, A. Impact of a high-fat diet on tissue acyl-coA and histone acetylation levels. Lerin, C. GCN5 acetyltransferase complex controls glucose metabolism through transcriptional repression of PGC-1alpha.

Rodgers, J. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Sakai, M. The GCN5-CITED2-PKA signalling module controls hepatic glucose metabolism through a cAMP-induced substrate switch.

Acetylation of glucokinase regulatory protein decreases glucose metabolism by suppressing glucokinase activity. Fang, S. The p acetylase is critical for ligand-activated farnesoid X receptor FXR induction of SHP. Kemper, J.

FXR acetylation is normally dynamically regulated by p and SIRT1 but constitutively elevated in metabolic disease states. Ma, K. Farnesoid X receptor is essential for normal glucose homeostasis.

Houtkooper, R. Sirtuins as regulators of metabolism and healthspan. Banks, A. SirT1 gain of function increases energy efficiency and prevents diabetes in mice. Pfluger, P. Sirt1 protects against high-fat diet-induced metabolic damage.

Purushotham, A. Systemic SIRT1 insufficiency results in disruption of energy homeostasis and steroid hormone metabolism upon high-fat-diet feeding. Xu, F. Chalkiadaki, A. High-fat diet triggers inflammation-induced cleavage of SIRT1 in adipose tissue to promote metabolic dysfunction.

Gillum, M. SirT1 regulates adipose tissue inflammation. White, A. Skeletal muscle-specific overexpression of SIRT1 does not enhance whole-body energy expenditure or insulin sensitivity in young mice.

Diabetologia 56 , — High-fat diet-induced impairment of skeletal muscle insulin sensitivity is not prevented by SIRT1 overexpression.

Qiang, L. Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of Ppargamma. Hepatic overexpression of SIRT1 in mice attenuates endoplasmic reticulum stress and insulin resistance in the liver.

Wang, R. Daitoku, H. Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity. Matsuzaki, H. Acetylation of Foxo1 alters its DNA-binding ability and sensitivity to phosphorylation.

Fasting-dependent glucose and lipid metabolic response through hepatic sirtuin 1. Hirschey, M. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Cell 44 , — Ahn, B. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis.

Lombard, D. Mammalian Sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation. Lantier, L. SIRT3 is crucial for maintaining skeletal muscle insulin action and protects against severe insulin resistance in high-fat-fed mice.

Fernandez-Marcos, P. Muscle or liver-specific Sirt3 deficiency induces hyperacetylation of mitochondrial proteins without affecting global metabolic homeostasis. Hancock, C. High-fat diets cause insulin resistance despite an increase in muscle mitochondria.

Holloszy, J. References and provide evidence that impaired mitochondrial function may not be a causative factor for insulin resistance. Holloway, G. Regulation of skeletal muscle mitochondrial fatty acid metabolism in lean and obese individuals.

Ryu, D. A SIRT7-dependent acetylation switch of GABPbeta1 controls mitochondrial function. Shin, J. SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease. Yoshizawa, T. SIRT7 controls hepatic lipid metabolism by regulating the ubiquitin-proteasome pathway.

Canto, C. Feng, D. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Sun, Z. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration.

Hong, S. Dissociation of muscle insulin sensitivity from exercise endurance in mice by HDAC3 depletion. Montgomery, R. Maintenance of cardiac energy metabolism by histone deacetylase 3 in mice.

Diet-induced lethality due to deletion of the Hdac3 gene in heart and skeletal muscle. Wang, S. Insulin and mTOR pathway regulate HDAC3-mediated deacetylation and activation of PGK1. PLoS Biol. Yang, X.

Protein O-GlcNAcylation: emerging mechanisms and functions. Metabolic regulation by lysine malonylation, succinylation, and glutarylation. Cell Proteomics 14 , — Choudhary, C.

The growing landscape of lysine acetylation links metabolism and cell signalling. Resh, M. Fatty acylation of proteins: the long and the short of it.

Guan, X. Understanding protein palmitoylation: biological significance and enzymology. China Chem. Yalovsky, S. Lipid modifications of proteins - slipping in and out of membranes.

Trends Plant Sci. Ren, W. Proteomic analysis of protein palmitoylation in adipocytes. Adipocyte 2 , 17—28 Du, K.

DHHC7 palmitoylates glucose transporter 4 Glut4 and regulates Glut4 membrane translocation. Glut4 palmitoylation at Cys plays a critical role in Glut4 membrane trafficking.

Wei, X. De novo lipogenesis maintains vascular homeostasis through endothelial nitric-oxide synthase eNOS palmitoylation. Spinelli, M. Brain insulin resistance impairs hippocampal synaptic plasticity and memory by increasing GluA1 palmitoylation through FoxO3a.

Turnbaugh, P. An obesity-associated gut microbiome with increased capacity for energy harvest. Schwiertz, A. Microbiota and SCFA in lean and overweight healthy subjects. Obesity Silver Spring 18 , — Article Google Scholar. Ridaura, V. Gut microbiota from twins discordant for obesity modulate metabolism in mice.

Science , References and provide evidence of altered gut microbiota in obesity and insulin resistance and show that this increases the propensity to develop obesity and insulin resistance.

Vrieze, A. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology , — e Todesco, T. Propionate lowers blood glucose and alters lipid metabolism in healthy subjects.

Venter, C. Effects of dietary propionate on carbohydrate and lipid metabolism in healthy volunteers. Chambers, E. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults.

Gut 64 , — De Vadder, F. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell , 84—96 den Besten, G. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARgamma-dependent switch from lipogenesis to fat oxidation.

Frost, G. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58 , — Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic syndrome.

Ang, Z. GPR41 and GPR43 in obesity and inflammation — protective or causative? Brown, A. The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids.

Canfora, E. Short-chain fatty acids in control of body weight and insulin sensitivity. Karaki, S. Expression of the short-chain fatty acid receptor, GPR43, in the human colon. Thangaraju, M. GPRA is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon.

Cancer Res. Jiang, L. Increased brain uptake and oxidation of acetate in heavy drinkers. Al-Lahham, S. Regulation of adipokine production in human adipose tissue by propionic acid. Xiong, Y. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR Freeland, K.

Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumour necrosis factor-alpha. Psichas, A. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Tolhurst, G. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2.

Diabetes 61 , — Aberdein, N. Sodium acetate decreases phosphorylation of hormone sensitive lipase in isoproterenol-stimulated 3T3-L1 mature adipocytes.

Adipocyte 3 , — Ge, H. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Arpaia, N. Metabolites produced by commensal bacteria promote peripheral regulatory T cell generation.

Maslowski, K. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects.

Liu, T. Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-kappaB pathway in RAW Inflammation 35 , — Cox, M.

Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E 2 and cytokines. World J. Li, G. Short-chain fatty acids enhance adipocyte differentiation in the stromal vascular fraction of porcine adipose tissue.

Dewulf, E. Evaluation of the relationship between GPR43 and adiposity in human. Hong, Y. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR Priyadarshini, M. An acetate-specific GPCR, FFAR2, regulates insulin secretion.

Felig, P. Plasma amino acid levels and insulin secretion in obesity. Cheng, S. Adipose tissue dysfunction and altered systemic amino acid metabolism are associated with non-alcoholic fatty liver disease.

Iwasa, M. Elevation of branched-chain amino acid levels in diabetes and NAFL and changes with antidiabetic drug treatment.

Bhattacharya, S. Validation of the association between a branched chain amino acid metabolite profile and extremes of coronary artery disease in patients referred for cardiac catheterization. Atherosclerosis , — Shah, S. Association of a peripheral blood metabolic profile with coronary artery disease and risk of subsequent cardiovascular events.

Newgard, C. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance.

Wurtz, P. Branched-chain and aromatic amino acids are predictors of insulin resistance in young adults.

Diabetes Care 36 , — Metabolomics and metabolic diseases: where do we stand? This comprehensive review discusses the emerging roles of metabolites, especially BCAAs, in insulin resistance.

Integrated metabolomics and genomics: systems approaches to biomarkers and mechanisms of cardiovascular disease. Interplay between lipids and branched-chain amino acids in development of insulin resistance. She, P. Obesity-related elevations in plasma leucine are associated with alterations in enzymes involved in branched-chain amino acid metabolism.

Wang, T. Metabolite profiles and the risk of developing diabetes. Lackey, D. Regulation of adipose branched-chain amino acid catabolism enzyme expression and cross-adipose amino acid flux in human obesity.

Herman, M. Adipose tissue branched chain amino acid BCAA metabolism modulates circulating BCAA levels. Burrill, J. Inflammation and ER stress regulate branched-chain amino acid uptake and metabolism in adipocytes. Zimmerman, H. Adipose transplant for inborn errors of branched chain amino acid metabolism in mice.

Shin, A. Brain insulin lowers circulating BCAA levels by inducing hepatic BCAA catabolism. Lefort, N. Increased reactive oxygen species production and lower abundance of complex I subunits and carnitine palmitoyltransferase 1B protein despite normal mitochondrial respiration in insulin-resistant human skeletal muscle.

White, P. Branched-chain amino acid restriction in Zucker-fatty rats improves muscle insulin sensitivity by enhancing efficiency of fatty acid oxidation and acyl-glycine export. Pedersen, H.

Human gut microbes impact host serum metabolome and insulin sensitivity. Lotta, L. Genetic predisposition to an impaired metabolism of the branched-chain amino acids and risk of type 2 diabetes: a Mendelian randomisation analysis.

Smith, G. Protein ingestion induces muscle insulin resistance independent of leucine-mediated mTOR activation. Macotela, Y. Dietary leucine — an environmental modifier of insulin resistance acting on multiple levels of metabolism. PLoS ONE 6 , e Zeanandin, G. Differential effect of long-term leucine supplementation on skeletal muscle and adipose tissue in old rats: an insulin signaling pathway approach.

Age Dordr 34 , — Xiao, F. Effects of individual branched-chain amino acids deprivation on insulin sensitivity and glucose metabolism in mice. Metabolism 63 , — Jang, C. A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance.

Roberts, L. beta-Aminoisobutyric acid induces browning of white fat and hepatic beta-oxidation and is inversely correlated with cardiometabolic risk factors. Sun, H. Catabolic defect of branched-chain amino acids promotes heart failure. Circulation , — Li, T. Defective branched-chain amino acid catabolism disrupts glucose metabolism and sensitizes the heart to ischemia-reperfusion injury.

Green, C. Branched-chain amino acid catabolism fuels adipocyte differentiation and lipogenesis. Su, X. Adipose tissue monomethyl branched-chain fatty acids and insulin sensitivity: effects of obesity and weight loss. Obesity Silver Spring 23 , — Malloy, V.

Methionine restriction decreases visceral fat mass and preserves insulin action in aging male Fischer rats independent of energy restriction. Aging Cell 5 , — Stone, K. Mechanisms of increased in vivo insulin sensitivity by dietary methionine restriction in mice. Diabetes 63 , — Wanders, D.

UCP1 is an essential mediator of the effects of methionine restriction on energy balance but not insulin sensitivity. FGF21 mediates the thermogenic and insulin-sensitizing effects of dietary methionine restriction but not its effects on hepatic lipid metabolism.

Epner, D. Nutrient intake and nutritional indexes in adults with metastatic cancer on a phase I clinical trial of dietary methionine restriction. Cancer 42 , — Mentch, S. Histone methylation dynamics and gene regulation occur through the sensing of one-carbon metabolism.

Chen, T. Tryptophan predicts the risk for future type 2 diabetes. PLoS ONE 11 , e Branched-chain amino acid levels are associated with improvement in insulin resistance with weight loss.

Laferrere, B. Differential metabolic impact of gastric bypass surgery versus dietary intervention in obese diabetic subjects despite identical weight loss.

Transl Med. Cotter, D. Ketogenesis prevents diet-induced fatty liver injury and hyperglycemia. Puchalska, P. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Taggart, A.

D -beta-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. Kimura, I. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 GPR Shimazu, T.

Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Goldberg, E. beta-Hydroxybutyrate deactivates neutrophil NLRP3 inflammasome to relieve gout flares. Youm, Y. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease.

Rheinheimer, J. Current role of the NLRP3 inflammasome on obesity and insulin resistance: A systematic review. Metabolism 74 , 1—9 Houstis, N. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Fisher, F. Understanding the physiology of FGF Foster, G. A randomized trial of a low-carbohydrate diet for obesity.

Chavez, A. Circulating fibroblast growth factor is elevated in impaired glucose tolerance and type 2 diabetes and correlates with muscle and hepatic insulin resistance. Diabetes Care 32 , — Fazeli, P. FGF21 and the late adaptive response to starvation in humans. Douris, N.

Beta-adrenergic receptors are critical for weight loss but not for other metabolic adaptations to the consumption of a ketogenic diet in male mice. Chavez-Talavera, O. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease.

Gastroenterology , — Preidis, G. Nutrient-sensing nuclear receptors PPARalpha and FXR control liver energy balance. Kawamata, Y. A G protein-coupled receptor responsive to bile acids. Maruyama, T. Identification of membrane-type receptor for bile acids M-BAR.

Broeders, E. The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Watanabe, M. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation.

Somm, E. beta-Klotho deficiency protects against obesity through a crosstalk between liver, microbiota, and brown adipose tissue. JCI Insight 2 , Fujisaka, S. Antibiotic effects on gut microbiota and metabolism are host dependent.

Kumar, D. Activation of transmembrane bile acid receptor TGR5 modulates pancreatic islet alpha cells to promote glucose homeostasis.

Thomas, C.

Insulin and Insulin Resistance: The Ultimate Guide

Since insulin is a pivotal hormone that regulates blood sugar, IR is closely associated with all stages of DM, including prediabetes, diabetes, and its complications.

Impaired β-cell compensation in response to increased IR is a pathophysiological factor associated with poor glucose tolerance, which contributes to the development of DM. Type 1 DM T1DM is caused by the primary loss of β-cells — the cells that release insulin — and the complex autoimmune process of continuous insulin deficiency.

Nevertheless, clinical and experimental evidence have shown that patients with T1DM exhibit IR 14 , which is a prominent feature in adolescents and adults 15 — 17 , mainly involving the liver, peripheral, and adipose tissue Insulin injections are currently the conventional treatment for T1DM, and prolonged overexposure to insulin itself is a trigger for insulin resistance.

patients with T1DM eventually also develop insulin resistance and other features of T2DM, such as cardiovascular disease Type 2 DM T2DM is characterized by defective insulin secretion from pancreatic beta cells.

Under normal conditions, increased insulin release by pancreatic β-cells is sufficient of insulin action and maintain normal glucose tolerance However, under the circumstances of IR combined with environmental factors and genetic factors related to T2D, persistent overnutrition sets up a vicious spiral of hyperinsulinemia and insulin resistance, ultimately leading to beta cell failure, possibly due to glucose and lipid toxicity and other factors leading to significant T2D There is a lot of evidence suggesting that both IR and T2D are associated with obesity, especially with high proportion of intra-abdominal and intra-hepatic fat, which is the most crucial factor contributes to the emergence of metabolic disease 22 , IR at the beta-cell level may play a role in the pathogenesis of insulin release defects.

Reduced insulin release may impair adipocyte metabolism, leading to increased lipolysis and elevated levels of non-esterified fatty acid NEFA.

Elevation of NEFA and glucose can work together to impair islet health and insulin action. Therefore, this process may slowly progress forward to develop T2D In addition, IR was independently associated with each of the chronic macrovascular and microvascular complications from diabetes Triglyceride-glucose index TyG index is a convenient measure of IR.

In a large Chinese inpatient cohort study, inpatients with elevated TyG index were shown to be at higher risk for lower extremity macrovascular stenosis, arterial stiffness and renal microvascular injury 25 , In particular, IR or hyperinsulinemia is responsible for the development of diabetic cardiomyopathy by pathophysiological mechanisms including impaired insulin signaling, cardiac mitochondrial dysfunction, endoplasmic reticulum stress, impaired autophagy, impaired myocardial calcium handling, abnormal coronary microcirculation, inappropriate neurohumoral activation and maladaptive immune responses 27 , Regarding chronic kidney disease, although this remains to be proven, IR is considered to be a factor contributing to the development and progression of diabetic nephropathy DN , as well as a consequence of DN.

IR is exacerbated during the development of DN, possibly due to some potentially modifiable changes in circulating hormones, neuroendocrine pathways, and chronic inflammation In recent years, a wealth of experimental, epidemiological and clinical evidence has suggested that IR and its compensatory hyperinsulinemia have a synergistic relationship with the development and progression of certain types of cancer, including breast, colorectal, prostate, pancreatic, adrenocortical and endometrial cancers 30 — To put it in perspective, IR and hyperinsulinemia, even in individuals without diabetes, are independently and positively associated with increased mortality from pancreatic cancer Besides, according to a large observational study, breast cancer incidence in women with high HOMA-IR is associated with all-cause mortality, especially in postmenopausal women Although the underlying mechanisms of the association between IR and tumor remain unclear, it may rely on several mechanisms and is not necessarily the same for different types of cancers.

On the other hand, IR is closely associated with visceral adipose dysfunction and systemic inflammation, both of which favor creating an environment conducive to tumorigenesis 38 , Additionally, epigenetic modifications which are triggered by IR and other environmental factors and chronic disease often involve in oncogenesis, such as DNA methylation, histone modifications, and non-coding RNA 35 , 40 , In addition to the mechanisms described above, recent studies indicate that gut microbiota may be a contributing factor in the relationship between IR and cancer, due to gut dysbiosis Therefore, increasing knowledge about the role of IR in cancer has important implications for cancer prevention and tumor growth inhibition.

IR is thought to be a key risk factor leading to cardiovascular and cerebrovascular diseases in different populations, whether normal or diabetic 43 — Increased plasma levels of fatty acids in patients with IR and dyslipidemia, with or without diabetes, may lead to the development of metabolism-related cardiomyopathy An example is diabetic cardiomyopathy, which is characterized by diastolic dysfunction and left ventricular hypertrophy in the absence of vascular defects.

Diabetic dyslipidemia and lipid accumulation in the myocardium are key pathologic features In animal experiments, mice have shown that when IR develops, insulin receptor substrate-1 IRS1 and insulin receptor substrate-2 IRS2 signaling will be impaired, resulting in impaired expression of cardiac energy metabolism genes and activation of p38α mitogen-activated protein kinase p38 , ultimately leading to abnormal cardiac function The strong association between IR and CVD may be due to the fact that the heart is a target organ for insulin, which requires greater energy consumption, yet when IR occurs, it impedes the normal function of the heart and increases the incidence of CVD 52 , Therefore, improving insulin sensitivity not only reduces plasma glucose concentrations in patients with T2DM, but also reduces the risk of cerebrovascular disease independent of the control of blood glucose levels 43 , The liver is one of the main organs controlling the metabolic balance and there is a close relationship between IR and NAFLD, which could be described as a two-way street 57 , NAFLD is characterized by excessive accumulation of lipids in hepatocytes.

Lipids and metabolites secreted by the liver, including lipoproteins, ketones, acylcarnitine and bile acids, may act as signaling molecules and regulate insulin action 59 , Hyperinsulinemia can drive hepatic lipogenesis and lipid accumulation directly as well as through indirect mechanisms, including excess circulating FFA, that impede the ability of insulin to inhibit hepatic glucose production High IR was found to be the most important predictor of NAFLD in both obese and lean subjects 62 , and studies have shown that serum insulin levels are strongly associated with hepatic lobular inflammation and histological progression such as ballooning Similarly, in patients with NAFLD, glycerol appearance and lipid oxidation were markedly increased, and IR also increased with the degree of steatosis 64 , A meta-analysis showed that compared with those without NAFLD, the risk of T2DM was more than two times higher in patients with NAFLD, with the highest risk particularly in patients with nonalcoholic steatohepatitis NASH In the condition of mildly active hepatic steatosis, IR is associated with hepatocellular injury and atherosclerotic dyslipidemia.

While in steatohepatitis, IR is combined with cytokine pro-inflammatory status and fibrosis indicators PCOS is a complex gynecologic endocrine disease, which is characterized by hyperandrogenism, menoxenia, ovulatory dysfunction and infertility.

A study of obese adolescent girls indicates that the PCOS phenotype with high androgen levels has the greatest degree of insulin resistance and inflammation Although the etiology and pathogenesis behind PCOS remain to be determined, IR and its compensatory hyperinsulinemia is considered to be an important pathological change that led to progression of PCOS and the main pathological basis for its reproductive dysfunction 69 — Excessive insulin secretion triggers insulin receptors in the pituitary gland, promoting androgen secretion from the ovaries and adrenal glands through the pituitary-ovary and adrenal axes, and increases free testosterone levels by inhibiting hepatic sex binding globulin SHBG synthesis 72 , Moreover, insulin, as a reproductive as well as metabolic hormone, has direct effect of stimulating ovarian androgen production by stimulating 17α-hydroxylase activity in the ovarian theca cells and enhance the activity of insulin-like growth factor-1 IGF-1 receptor in the ovary, thus increasing its free IGF level and promoting androgen production 74 , Also, IR has long-term and deleterious effects on the metabolism of women with polycystic ovary syndrome.

In addition to the diseases described above, IR is also associated with many other diseases of various systems throughout the body. This includes liver cirrhosis, which is associated with changes in glucose homeostasis, even in intact liver function. Essential features of the association between cirrhosis and IR include endocrine dysregulation, liver inflammation, changes in muscle mass and composition, changes in the gut microbiota, and permeability IR may also affect the association between insulinemia and bone mass, and Yi-Hsiu Fu et al.

Additionally, IR is a crucial risk factor for deterioration of renal function in non-diabetic chronic kidney disease CKD and hypertension We also noted the effect of IR in the studies related to postburn trauma 81 , postadolescent acne 82 , gastro-esophageal reflux disease GERD 83 and other diseases.

The pathogenesis of IR is the result of the interaction of environmental and genetic factors. Its mechanism of development mainly includes abnormalities in the internal environment, such as inflammation, hypoxia, lipotoxicity, immune environment abnormalities, and abnormal metabolic functions, including metabolic tissues and metabolites.

IR and metabolic disorders are commonly clustered in families, which is thought to be the result of an interaction of environmental and genetic factors, although the full genetic background of these conditions remains incomplete 84 , Genetic factors associated with IR can be classified as abnormal structure of insulin, genetic defects in the insulin signaling system, genetic defects related to substance metabolism, and other related genetic defects.

There are also rare mutations in insulin receptor genes leading to reduced number of cell surface receptors and defective insulin receptor pathways causing hereditary IR, which are found in patients with genetic syndromes of severe IR, such as type A syndrome of extreme IR, leprechaunism, Rabson-Mendenhall syndrome and Donohue syndrome 88 , More importantly, since many molecular pathways are involved in energy homeostasis and metabolism, IR is the result of a certain number of mutations in multiple genes, such as those related to type 4 glucose transporter GLUT4 , glucokinase, and Peroxisome proliferator-activated receptor PPAR nuclear receptor family, among others 90 , Mutations in lipid metabolic pathways, such as mutations in adipocyte-derived hormones such as leptin, adiponectin, resistin or their receptors, mutations in peroxisome proliferator-activated receptors α, γ, and δ, mutations in the lipoprotein lipase gene, and other mutations in genes related to adipose tissue formation can affect the development of glycolipid metabolism and IR The latest advances in high-throughput genetics have revealed the relationship between protein tyrosine phosphatase N1 PTPN1 and IR, and that the association is mediated by differences in DNA sequences outside the coding region of PTPN1 Healthy carriers of the T allele of TCF7L2 rs, may increase insulin secretion and lead to impaired β-cell function, which is associated with an increased risk of T2DM Obesity-induced IR is characterized by impaired insulin function that inhibits hepatic glucose output and promotes glucose uptake in adipose tissue and muscle It has been found that waist circumference is closely related to IR, and an increase in waist circumference corresponds to a decrease in glucose consumption or an increase in IR.

Hence, obesity, especially central obesity, may induce the development of IR due to the massive accumulation of adipose tissue inducing systemic insulin resistance, including endocrine dysregulation and inflammation In obese individuals, especially in those with abdominal obesity, the increase in adipose tissue tends to be more lipolytic, resulting in higher plasma free fatty acid FFA levels and intracellular lipid accumulation.

Elevated FFA can enhance the phosphorylation of serine residues of insulin receptor substrate IRS by activating a series of protein kinases such as c-Jun N-terminal kinase JNK , whose activity is abnormally increased in obese patients , Another mechanism linking obesity and IR is chronic inflammatory responses, including increased production and release of pro-inflammatory factors such as TNF-α, IL-6, and C-reactive protein, which cause insulin resistance in liver, skeletal muscle, and adipose tissue through insulin-interfering signaling pathways Several physiopathological factors and therapeutic causes, such as chronic hyperglycemia, high free fatty acidemia, certain drugs, such as glucocorticoids, pregnancy, and increased insulin-antagonistic hormones in the body all contribute to the occurrence of IR.

There is a pathophysiological relationship between chronic obstructive pulmonary disease COPD and IR, partly because the two conditions share common risk factors, such as smoking and lack of physical activity.

In addition, systemic effects deterioration of physical inactivity and sedentary behavior, inflammation and corticosteroid therapy in patients with COPD may also play a role Also, IR is a common condition after organ transplantation, which leads to new-onset diabetes and metabolic syndrome after transplantation, and subsequent hyperglycemia may significantly increase the morbidity and mortality of cardiovascular disease after kidney transplantation , This is due to post-transplant treatment with immunosuppressive agents such as sirolimus, cyclosporine, steroids, etc.

In both rodents and humans, exogenous synthetic glucocorticoids such as prednisolone and dexamethasone may induce a number of adverse effects when administered in excess or for prolonged periods, including the development of glucose intolerance, islet-cell dysfunction, IR, hyperglycemia, and dyslipidemia , In contrast, almost all morphophysiological changes induced by dexamethasone in the endocrine pancreas are reversed after cessation of treatment Advanced age is an important factor in increasing susceptibility to IR.

With increasing age, there is insufficient insulin secretion and a progressive decrease in glucose tolerance, as well as increasing IR due to sarcopenia, excess adiposity and osteoporosis , According to epidemiology, the prevalence of IR and T2DM is high in the elderly population , This is associated with an increased prevalence of central obesity and increased visceral fat in the aging population 99 , In addition to this, factors that increase the risk of IR in the elderly are free radicals that contribute to oxidative stress in old age, and mitochondrial dysfunction — The paper by Petersen et al.

published in the journal Science mentions that older subjects clearly showed reduced insulin-stimulated muscle glucose metabolism compared to younger subjects.

According to the result of an animal experiment, compared with young mice, aged mice are more susceptible to IR, due to reduced levels of glycolytic proteins and reduced flexible to diet, caused by reduced mitochondrial β-oxidation capacity However, these hypotheses still need to be further tested and further understanding of the metabolic changes associated with aging.

The balance of insulin action involves multiple processes in several glucose-utilizing organs or organs, including the liver, adipose tissue, skeletal muscle and kidneys. These metabolic processes receive complex signal regulation.

The etiology and pathogenesis of IR are complicated, and the main pathological mechanisms include abnormalities in receptor binding, environment inside the host, intracellular factors, autophagy and intestinal microecology.

It is noteworthy that the mechanisms of IR occur somewhat differently in different insulin receptor tissues, and IR appears in a different order, where the initial appearance of IR is in adipose tissue. However, they interact with each other and may eventually develop into systemic IR, a phenomenon verified in observational studies in humans — In-depth study of the pathogenesis of IR and multiple research directions have become the key to solving the challenges of IR and its related metabolic diseases today.

The effects of insulin signaling pathways and the effects of inflammatory cytokines and FFA on them are shown in Figure 2. Figure 2 A The insulin signaling pathway; B Abnormalities in the insulin signaling pathway caused by inflammatory cytokines, FFA, etc.

Insulin receptors INSR which is a tyrosine kinase, bind specifically to insulin and play a key role in insulin-mediated glucose homeostasis and cell growth , Impaired INSR binding mainly refers to a decrease in the affinity and number of target receptors on the cell membrane or structural abnormalities of the target receptors that affect insulin binding to the receptor The insulin receptor substrate protein is generally considered a node in the insulin signaling system, which is closely related to the development of insulin insensitivity.

At the molecular level, the crosstalk between the downstream nucleotide-binding oligomerization domain NOD 1 effector and the insulin receptor pathway may inhibit insulin signaling by reducing the action of insulin receptor substrates Insulin activates insulin receptor tyrosine kinases, which are capable of aggregating and phosphorylating various substrate docking proteins, such as the insulin receptor substrate IRS protein family.

Of the four mammalian IRS proteins IRS-1, IRS-2, IRS-3, IRS-4 , IRS1 and IRS2 play key roles in regulating growth and survival, metabolism and aging.

They are key substrates of insulin signaling and play an important role in insulin signaling by binding to PI3K and inducing downstream pathways.

At the molecular level, dysregulation of the signaling pathway by insulin receptor substrates IRS is one of the most common causes of this disease. For example the double-stranded RNA-dependent protein kinase PKR has also been shown to upregulate the inhibitory phosphorylation of IRS1 and the expression of IRS2 in liver and muscle cells, thereby regulating the insulin signaling pathway.

Mediated by two other protein kinases, JNK and IKK, PKR upregulated the phosphorylation of IRS1 at Ser and inhibited the tyrosine phosphorylation of IRS1 , IRS1 has also been shown to be a target of ceramide-induced Pbx regulating protein 1 Prep1 and p in muscle cells, and the Prep1-p axis also affects IRS-1 stability In addition, protein tyrosine phosphatase 1B PTP-1B , protein kinase C PKC and tyrosine residue receptor phosphorylation levels are involved in the regulation of receptor-insulin binding in target tissues.

It has been shown that inhibition of PTP1B, a main negative regulator of insulin receptor signaling, can improve glucose homeostasis and insulin signaling In the insulin receptor signaling cascade, protein tyrosine kinase amplifies the insulin signaling response, and phosphatase is necessary to regulate the rate and duration of the reaction IR occurs in a variety of tissues, including skeletal muscle, liver, kidney and adipose tissue, and its mechanisms are specific.

Among the target organs of insulin, bone, as an endocrine organ, can regulate energy homeostasis by altering insulin sensitivity, dietary behavior, and adipocytes There seems to be a bilateral relationship between bone and IR that binds them together in a biological partnership Among them, skeletal muscle estrogen receptor α plays a crucial role in maintaining systemic glucose homeostasis and insulin sensitivity It has been repeatedly demonstrated that skeletal muscle tissue plays an important role in the maintenance of systemic glucose homeostasis and overall metabolic health.

In addition, the crosstalk between muscle factors and adipokines leads to negative feedback, which in turn aggravates muscle reduction obesity and IR In the kidney, the effector cells of insulin are podocytes in which nucleotide-binding oligomerization domain 2 NOD2 is highly expressed. NOD2 is a major member of the NOD receptor family and is involved in the innate immune response.

It induces podocyte IR by activating the inflammatory response In terms of hepatic IR, IRA, one of the isoforms of the insulin receptor, whose expression in the liver of mice on a high-fat diet increase hepatic glucose uptake, decrease lipid accumulation, and reduce or at least delay the development of fatty liver and NASH.

This suggests that a gene therapy approach to hepatic IRA expression could act as a facilitator of glucose uptake in IR states — Insulin acts by binding to the INSR and activating downstream signaling pathways which have been extensively studied.

Although where the defect occurs in the insulin signaling pathway remains a matter of doubt, many key insulin signaling pathway components have been identified. IR is caused by defects in one or more of these signaling components Environment, such as diet and exercise, and genetics, as well as the interaction between the two, play a major role in the development of IR and metabolic disease.

Exercise and dietary habits may directly or indirectly drive changes in the host internal microenvironment. Current research suggests that extracellular influences such as inflammation, hypoxic environments, lipotoxicity or immune abnormalities can trigger intracellular stress in key metabolic target tissues, which impairs the normal metabolic function of insulin in these cells thereby causing the progression of whole-body IR Obesity characterized by a chronic, low-grade inflammatory state is closely associated with IR.

The mechanisms of inflammation leading to IR mainly include inflammatory factors acting on the insulin signaling system to interfere with INSR signal transduction. TNF-α and IL-1β are additional macrophage-derived pro-inflammatory mediators that directly affect insulin sensitivity , TNF-α stimulates insulin-resistant adipose tissue through IRS protein interference by abnormal signals on phosphorylated serine residues of IRS1 In addition, TNF-α could affect insulin signaling through serine phosphorylation and kinase pathway defects 99 , CRP is another marker of inflammation associated with IR and metabolic diseases and is a widely used clinical biomarker.

CRP binds to leptin, blocks leptin signaling and modulates its central action and hypothalamic signaling, thereby directly interfering with energy homeostasis, insulin sensitivity and glucose homeostasis , The above pro-inflammatory cytokines exert their effects by stimulating major intracellular inflammatory pathways, and the activation of these pathways also promotes increased expression of the inflammatory factors involved in IR.

Toll-like receptor TLR , especially TLR4, participates in IR-related inflammation by increasing the gene expression of IKKβ, NF-κB transcription factors, and pro-inflammatory mediators in adipose tissue macrophages — IKK is an enzyme complex that activates the NF-κB transcription factor It has also been shown that NF-κB receptor activator RANKL is a potent stimulator of NF-κB and that systemic or hepatic blockade of RANKL signaling leads to significant improvements in hepatic insulin sensitivity and prevents the development of diabetes And JNK signaling in adipocytes leads to an increase in circulating concentrations of hepatic factor fibroblast growth factor 21 FGF21 , which regulates systemic metabolism In the pathogenesis of IR and metabolic diseases, immune cells play a crucial role.

Adipose tissue contains most types of immune cells, which under conditions of obesity contribute to a complex network of inflammation and IR with activation and infiltration of pro-inflammatory immune cells in adipose tissue, including macrophages, neutrophils, eosinophils, mast cells, NK cells, MAIT cells, CD4 T cells, CD8 T cells, regulatory T cells and B cells, as well as high levels of pro-inflammatory molecules Among them, adipose tissue macrophages can be divided into M1 phenotype pro-inflammatory macrophages and M2 phenotype anti-inflammatory macrophages , representing the two extremes of macrophage polarization.

M1 macrophages are highly antimicrobial and antigen-presenting, producing pro-inflammatory cytokines, such as TNF-α, and reactive oxygen species ROS that worsen inflammation, mast cells, neutrophils and dendritic cells directly or indirectly exacerbate IR In contrast, M2 macrophages help maintain insulin sensitivity in lean adipose tissue, as well as eosinophils and innate lymphocytes appear to have a protective effect on glucose homeostasis and insulin sensitivity — Crosstalk between M1-M2 macrophage polarization plays an important role in IR through the shift from M1 to M2 phenotype and activation of transcription factors , Dysregulation of visceral adipose tissue macrophage ATM response to microenvironmental changes underlies the development of abnormal local and systemic inflammation and IR In the obese state, enhanced macrophage infiltration and secretion of various inflammatory cytokines in white adipose tissue activate JNK and NF-κB, causing local and systemic IR , Macrophages can alter their phenotype in response to changes in the microenvironment and macrophage differentiation.

In the past, more attention has been paid to the regulation of insulin sensitivity by innate immune cells, particularly macrophage mediated, which have been mentioned before. Cells of the adaptive immune system, B lymphocytes and T lymphocytes, and their respective subsets, are also thought to be important regulators of glucose homeostasis and play an important role in the immunopathogenesis of autoimmune diabetes , , Impaired through an adaptive immune response, IR can also be driven by inflammation and dysregulation of the gut microbiota, as in pathogen-induced periodontitis In addition, the intestinal immune system is an important regulator of glucose homeostasis and obesity-related IR in turn affects intestinal permeability and thus systemic IR Another essential part of the immune defense system is the complement system.

It plays an important role in activating innate and adaptive immune responses, promoting apoptosis, and eliminating damaged endogenous cells. Patients with obesity exhibit activation of the complement system in their adipose tissue, which is connected to changes in glucose metabolism and subclinical inflammation Adipose tissue hypoxia is causally related to obesity-induced IR, especially in high-fat diet HFD fed and early obese patients, as adipocyte respiration becomes uncoupled, resulting in a state of increased oxygen consumption and relative adipocyte hypoxia Clinically, obstructive sleep apnea OSA , characterized by intermittent hypoxia IH , is a widely prevalent respiratory disorder with a particularly high prevalence in obese patients and is associated with IR and metabolic diseases such as hypertension, cardiovascular risk and NAFLD , Not only in obese individuals, but an animal study found that IH cause acute IR in lean or healthy mice, which is related to reduced glucose utilization in oxidized muscle fibers.

As the glucose infusion rate decreased, hypoxia induced systemic IRA The key regulators of oxygen homeostasis in response to hypoxia are the hypoxia-inducible factors HIFs , a family of transcription factors activated by hypoxia. Adipocyte hypoxia could trigger HIF-1α induction causing adipose tissue inflammation and IR , HIFmediated activation of NOX4 transcription and the consequent increase in H2O2 led to intermittent hypoxia-induced pancreatic β-cell dysfunction In hypoxic adipocytes, HIF-1α activates the NLRP3 inflammasome pathway and stimulates IR by upregulating the expression of pla2g In obesity-induced intestinal hypoxia, HIF-2α increases the production of ceramide, to promote the expression of the key enzyme sialidase 3 encoding Neu3, which leads to the development of IR in obese mice induced by a high-fat diet While in skeletal muscle, hypoxia is a stimulus stimulating GLUT4 translocation via activation of AMPK, causing defects of glucose transport and this may counteract IR Insulin regulates lipid metabolism through the typical insulin signaling cascade, while metabolites can also directly regulate insulin sensitivity by modulating components of the insulin signaling pathway Lipids have multiple roles as signaling molecules, metabolic substrates and cell membrane components, and can also alter proteins that affect insulin sensitivity Lipotoxicity is when the storage capacity of adipose tissue is overloaded due to obesity, overnutrition, etc.

High concentrations of lipids and lipid derivatives cause deleterious effects on cells through mechanisms including oxidative stress, endoplasmic reticulum ER stress, c-Jun NH2-terminal kinase JNK -induced toxicity, and BH3-pure protein-induced mitochondrial and lysosomal dysfunction , Numerous studies have reported that Adipose tissue dysfunction and lipotoxicity play a role in metabolic disorders and IR , This is associated with a chronic elevation of free fatty acids FFA, also called non-esterified fatty acids in plasma due to adipose tissue dysfunction Adipose malnutrition or adipose tissue dysfunction can lead to pathologically elevated FFAs.

Chronically elevated FFAs appear to cause adipocyte production of inflammatory factors, decreased insulin biosynthesis, glucose-stimulated insulin secretion, and glucose sensitivity in β-cells.

The ER stress pathway is a key mediator of inflammation induced by serum excess FFA and IR in various cell types, and PERK and IKKβ are key signaling components The obesity-induced increase in adipocyte volume and tissue mass will lead to inflammation, additional disturbances in adipose tissue function, and ultimately adipose tissue fibrosis Adipose tissue macrophages are an abundant immune component of hypertrophy, which plays a key role in diet-induced T2DM and IR In renal ectopic lipid accumulation, lipotoxicity promotes podocyte injury, tubular injury, thylakoid proliferation, endothelial cell activation and macrophage-derived foam cell formation, which contribute to the development of renal IR and other renal diseases, especially diabetic nephropathy In skeletal muscle, sustained nutrient overload of L6 myotubes leads to lipotoxicity that promotes activation of the IKKβ-NFkB pathway in muscle cells, inducing increased cellular ROS and impaired insulin action in the myotubes Saturated fatty acids are known to increase the production of lipotoxic products such as ceramide and diacylglycerol, which disrupt islet beta-cell function, vascular reactivity and mitochondrial metabolism, and also play a key role in the induction of muscle IR — Similarly, defective fatty acid oxidation FAO and consequent lipotoxicity in cardiac cells induce a range of pathological responses, including oxidative stress, DNA damage, inflammation and insulin resistance.

The obesity-mediated atrial fibrillation and structural remodeling can be attenuated by promoting FAO, activating AMPK signaling and attenuating atrial lipotoxicity through levocarnitine LCA Lysophosphatidic acid LPA is an effective, biologically active lipid.

After binding to G protein-coupled receptors, it can profoundly affect cell signal transduction and function.

Metabolic and inflammatory disorders, including obesity and IR, are associated with modifications in LPA signaling as well as the production and function of autocrine motility factors Additionally, it has been discovered that the anti-adipogenic transcription factor GATA-3 is a possible molecular target that affects adipogenesis.

Those with obesity and IR exhibit increased GATA-3 expression when compared to insulin-sensitive individuals with BMI matches While lifestyle interventions such as physical activity have been confirmed to have a positive effect on insulin sensitivity in skeletal muscle, affecting lipid metabolism Ceramides are a family of lipid molecules consisting of sphingosine and a fatty acid.

The synthesis of de novo ceramides depends on the availability of free fatty acids, especially palmitate, whose over-intake may lead to an excessive accumulation of ceramides In addition to their function in lipid bilayers, these molecules are also thought to be biologically active agents involved in a variety of intracellular pathways, such as free radical production, release of inflammatory cytokines, apoptotic processes, and regulation of gene expression.

Ceramides are metabolic products that accumulate in individuals suffering from obesity or dyslipidemia and alter cellular processes in response to fuel overload ceramides accumulation over time modulates signaling and metabolic pathways that drive lipotoxicity and IR, causing tissue dysfunction Numerous studies have been conducted in recent years to confirm the critical role played by ceramides in glucose homeostasis and insulin signaling These evidence are particularly strong in skeletal muscle, while the data in liver and WA are somewhat more equivocal , Ceramides are synthesized by ceramide synthase CerS through N-acylation.

To date, six mammalian CerS have been identified CerS that show different affinities for the fatty acid acyl-CoA chain length used for sphingomyelin N-acylation.

CerS6 is specific for C14 and C16 acyl chain lengths, and CerS6 levels are significantly increased in obese adipose tissue , In addition, ceramide may cause IR by accumulating in mitochondria and causing mitochondrial reactive oxygen species ROS or by promoting the secretion of pro-inflammatory factors Another lipid metabolite closely associated with IR is DAG, whose accumulation in skeletal muscle, adipocytes and liver is thought to promote IR by altering cellular signaling at its specific location, due to increased serum FFA levels The DAG hypothesis of IR is that the interference of activated PKC, especially the novel PKC isoforms including δ, ϵ, ν, and θ, with insulin signaling is due to the accumulation of DAG in insulin-sensitive tissues , In particular, 1,2-DAG, which derives from esterification and accumulates mainly in the membranes, is clearly associated with PKC activation, and these isoforms then phosphorylate IRS1 serine with the result that decrease PI3K activation , It is worth noting that the role of intracellular ceramide and DAG in IR is controversial and that defects in these components are unlikely to be the sole cause of IR.

It is true that not all studies have confirmed a role for the DAG-PKC-insulin receptor pathway in IR; for example, some studies have shown that PKCϵ deficiency in the liver has no effect on systemic insulin sensitivity in mice , and there are also experiments in which acute knockout of PKCϵ in the liver protects rats from IR Therefore, more in-depth studies on proximal insulin signaling with DAG and ceramide are still needed.

Organelles, including the endoplasmic reticulum ER , mitochondria and endoplasmata, contribute to a range of cellular functions through their unique local environment and molecular composition.

Organelles can actively communicate and cooperate with each other through vesicle trafficking pathways and membrane contact points MCSs to maintain cellular homeostasis, which facilitates the exchange of metabolites and other information required for normal cellular physiology Imbalances in organelle interactions may lead to various pathological processes, such as imbalances in cellular energy metabolism Recent studies have shown that mitochondria could interact with various organelles , which are essential for energy metabolism and cell survival, and increasing evidence shows that mitochondrial dysfunction in skeletal muscle and mitochondrial overactivation may induce IR The production of mitochondrial ROS is thought to adjust skeletal muscle insulin sensitivity.

Mitochondrial quality control mechanisms are regulated by PGC-1α, which may affect age-related mitochondrial dysfunction and insulin sensitivity The continuous processes that occur in the skeletal muscle after excessive intake of a high-fat diet include the accumulation of cytosolic fatty acids, increased production of ROS, mutation, and aging.

The ensuing mitochondrial dysfunction could lead to decreased β-oxidation, respiratory function, and increased glycolipid toxicity.

Together, these events induce IR in the skeletal muscle The physical contact site between the mitochondria and endoplasmic reticulum ER is called the mitochondrial-associated membrane MAM. The imbalance of MAMs significantly leads to IR. ER stress may be the main mechanism by which MAM induces IR in the brain, especially in the hypothalamus , Exosome-like vesicles ELVs are the smallest type of extracellular vesicles released from cells that play a role in cell crosstalk because they regulate insulin signaling and β-cell quality, and released ELVs leading to IR or β-cell apoptosis PTEN is not only a tumor suppressor gene but also a metabolic regulator.

Under physiological and T2D conditions, PTEN also has a negative regulatory function in insulin signaling through its inhibition in the PI3K pathway , PTEN reduces the level of phosphatidylinositol-3, 4, 5-phosphate PIP3. This leads to impaired insulin signaling and promotion of IR in the pathogenesis of T2D.

The function of PTEN in regulating insulin signaling in different organs has been identified. The role of PTEN in the regulation of insulin action in many cell types has been elucidated through mouse models of lacking PTEN in metabolic organs and in vitro cell culture , Interventions targeting PTEN regulatory signaling may therefore be a promising target aimed at reversing insulin resistance.

In addition to its effects on skeleton, Vit D has significant effects on pancreatic β-cells function and metabolic syndrome including blood pressure, abdominal obesity, glucose metabolism associated with it, as calcitriol functions as a chemical messenger by interacting with calcium flux-regulating receptors on beta cells As the results of a meta-analysis showed, there was an inverse relationship between serum Vit D concentration and metabolic syndrome risk in the general adult population in cross-sectional studies Vitro studies showed that Vit D could regulate lipid and glucose metabolism in adipose tissue, skeletal muscle and liver, and pancreatic insulin secretion Minerals are essential micronutrients for the human body.

Deficiencies in certain micronutrients due to differences in diet composition may lead to imbalances in glucose homeostasis and IR Magnesium is a cofactor required for glucose access to cells and carbohydrate metabolism, and it has the function of regulating the electrical activity of pancreatic beta cells and insulin secretion Mechanistically explained, magnesium is a cofactor in the downstream action of the insulin cascade.

Low magnesium ion levels lead to defective tyrosine kinase activity, blocking intracellular insulin action and altered cellular glucose transport, thus promoting IR On the other hand, magnesium deficiency inhibits cellular defenses against oxidative damage and triggers chronic systemic inflammation that enhances IR.

As demonstrated in a longitudinal study, magnesium intake was also inversely associated with high-sensitivity CRP, IL-6 and fibrinogen levels, as well as HOMA-IR There is evidence suggesting that magnesium supplementation attenuates IR in patients with hypomagnesemia-associated IR Also, animal studies have shown that dietary magnesium supplementation to increase plasma magnesium concentrations reduces blood glucose levels, improves mitochondrial function, and reduces oxidative stress in diabetic mice However, new intervention studies are still needed to clarify the role of nutrients in the prevention of this metabolic disorder, as well as to standardize the type, dose, and timing of magnesium supplementation.

Zinc is an essential micronutrient for metabolism, which plays a particularly critical role in the islets.

Diabetes affects zinc homeostasis, and disturbances in zinc homeostasis have been associated with diabetes and IR Because zinc is an essential component of insulin, it regulates islet cell secretion and promotes its binding to hepatocyte membranes while maintaining phosphorylation and dephosphorylation levels of the receptor.

Zinc influx mediated by Slc39a5, a zinc exporter in pancreatic β-cells, plays a role in insulin processing and secretion by inducing Glut2 expression through Sirt1-mediated activation of Pgc-1α In addition, zinc acts as a pro-antioxidant to reduce the formation of ROS, which is particularly beneficial in aging and IR Mineral deficiencies are directly or indirectly associated with oxidative stress, which ultimately leads to IR or diabetes The brain is also an insulin-sensitive organ with a large number of insulin receptors distributed , The action of insulin in the brain produces a variety of behavioral and metabolic effects that influence eating behavior, peripheral metabolism, and cognitive performance Disturbances in the role of insulin in the brain reveal a possible link between metabolism and cognitive health.

The hypothalamus plays a fundamental role in the survival and control of physiological processes necessary for vital physical functions, including various endocrine functions. Injecting insulin via intranasal administration leads to an increase and subsequent decrease in plasma insulin, affecting peripheral metabolism, and a decrease in BOLD signaling and cerebral blood flow in the hypothalamus is observed , It appears that the effects of central insulin may have a biphasic effect on peripheral insulin sensitivity Insulin signaling has been shown to affect the molecular cascade of hippocampal plasticity, learning, and memory Furthermore, the insulin-responsive glucose transporter GluT4 has a key part in hippocampal memory processes, and reduced activation of this transporter may underlie IR-induced cognitive deficits Autophagy is a self-degrading process that is conserved in all eukaryotic cells and plays a crucial role in balancing energy sources during critical periods of development and in response to nutritional stress.

Autophagy also promotes cellular senescence and cell surface antigen presentation, prevents genomic instability and necrosis, and it is an important mechanism for a variety of physiological processes, such as cellular homeostasis, senescence, immunity, oxidation, differentiation, and cell death and survival Recent studies have shown that autophagy is an important regulator of organelle function and insulin signaling, and that loss of autophagy is a key component of defective insulin action in obesity, which may be specifically related to ER function It has been found that autophagy deficiency and its resulting mitochondrial dysfunction increase fibroblast growth factor 21 Fgf21 expression through the induction of Atf4.

The induction of Fgf21 promotes protection against diet-induced obesity and IR In addition, exercise induces autophagy through the regulator BCL2, which may contribute to beneficial metabolic effects and improve IR in muscle In addition to the aforementioned influences such as metabolites and cytokines, the trillion bacterial colonized gut microbiota can also contribute to IR , Patients with metabolic syndrome showed increased insulin sensitivity after six weeks of infusion of gut microbiota from lean individuals.

Levels of gut microbiota producing butyrate, which has been shown to prevent and treat diet-induced insulin resistance in mice by promoting energy expenditure and inducing mitochondrial function, were also increased , Dietary reasons for obesity may promote IR both through mechanisms independent of the gut microbiota and through mechanisms dependent on the bacterial community Intestinal dysbiosis is associated with the transfer of bacterial lipopolysaccharide LPS into the systemic circulation and its induction of metabolic endotoxemia, leading to a chronic subclinical inflammatory process and the development of IR through activation of toll-like receptor 4 TLR4 — In addition to the LPA mentioned above, branched-chain amino acids BCAAs are another harmful gut microbially regulated metabolite whose levels are increased in the serum metabolome of IR individuals.

Prevotella copri has been shown in mice experiments to induce IR, exacerbate glucose intolerance and increase circulating levels of BCAAs Moreover, gut microbiota-derived short-chain fatty acids SCFA may improve IR and prevent T2DM by reducing the secretion of pro-inflammatory cytokines and chemokines and decreasing local macrophage infiltration, as well as increasing the lipid storage capacity of white adipose tissue , , Taken together, targeting gut microbes may have the potential to reduce IR and decrease the incidence of related metabolic diseases.

This lifestyle triggers several mechanisms such as the development of IR that aggravate metabolic stress. Next, the contribution of non-pharmacological therapies, including exercise and diet, to the alleviation of IR will be elaborated.

Exercise is well known to improve metabolic disease by improving obesity and enhancing insulin sensitivity. A meta-analysis determined the effectiveness of a structured exercise intervention program for IR in T2DM, and the evidence highlights that regular exercise improves glycemic control and therefore can be recommended for reducing IR with a moderate level of evidence As we know, physical exercise increases the oxidative capacity and biogenesis of mitochondrial substrates in skeletal muscle.

It was shown that treadmill training modulates the increase in mitochondrial substrate oxidation in liver and skeletal muscle induced by a high-energy diet in mice, disconnecting it from pyruvate and acetyl CoA-driven lipid synthesis. This may help prevent the long-term deleterious effects of excessive nutritional intake on liver mitochondrial function and insulin sensitivity, thereby preventing the development of metabolic diseases such as fatty liver and NAFLD As described in the mechanism section, intermittent hypoxia leads to disturbances in the gut microbiota-circulating exosome pathway, disrupting adipocyte homeostasis and leading to metabolic dysfunction manifested as IR, whereas experiments have shown that such changes can be attenuated by physical activity, as regular non-strenuous activity will lead to substantial improvements in the gut microbiota-exosome pathway In addition, available data suggest that aerobic exercise can lead to increased insulin sensitivity and enhanced glucose metabolism through a variety of different molecular mechanisms, including upregulation of insulin transporters on cell membranes of insulin-dependent cells, reduction of adipokines, normalization of redox status, improvement of β-cell function, regulation of IRS-1 phosphorylation, reduction of ceramide plasma levels, and induction of angiogenesis, which may lead to a reduced incidence of diabetic complications, as well as other metabolic effects , Other forms of exercise, such as yoga, have also been shown to improve IR.

Several meta-analyses have shown that yoga is a safe and effective intervention to reduce waist circumference and systolic blood pressure in patients with metabolic syndrome, particularly in improving cardio-metabolic health , Some traditional Chinese health exercises, such as qigong and tai chi, have also been shown to have a measurable effect on weight, waist circumference, leg strength, increase HDL cholesterol, and result in significant improvements in IR , As mentioned above, high-fat diets and the obesity they induce are a major cause of IR.

Conversely, weight loss, when necessary, and dietary interventions such as intermittent fasting programs that reduce carbohydrates in the diet can significantly improve glycemic and insulin responses.

The Mediterranean diet is characterized by a wide range of cardio-protective nutrients, with beneficial effects on several outcomes related to metabolic health, and significant beneficial changes in metabolic risk factors, including HOMA-IR index — There are also RCT studies reporting that a high-protein diet is more effective in controlling IR and glycemic variability compared to a Mediterranean diet, which may be related to the satiety and increased metabolic rate associated with a high-protein, low-sugar diet In terms of dietary composition, a key dietary strategy for treating IR and improving glycemic control is to consume foods and meals that reduce the glucose fluctuations known to induce oxidative stress and beta cell damage The contribution of high-fat diets to obesity and IR is well known.

However, a single-minded approach to weight loss by replacing fat intake with carbohydrates is counterproductive because it could exacerbate IR. Researchers suggest that calorie restriction for weight loss and rationing of the macronutrient composition of the diet is important.

The possible mechanism for this is that calcium and vitamin D in supplemental dairy products may facilitate lipolysis and optimize glucose metabolism Carbohydrates are the main macro-nutrient influencing the glycemic response, especially after a meal. In recent years, some researchers have proposed that consumption of carbohydrates rich in dietary fiber and low glycemic index, such as whole grains, is beneficial in improving insulin sensitivity and metabolic flexibility, independent of gut hormones , A recent meta-analysis reported that increasing daily fiber intake by 15 or 35 grams compared to a low-fiber diet reduced homeostatic model assessment of insulin resistance HOMA-IR , leading to improvements in glycemic control, lipids, weight, and inflammation, as well as a reduction in premature mortality Not only is the amount of carbohydrate intake important, but the timing of major carbohydrate intake during the day is also a determining factor in the increase in glucose and insulin after meals and the improvement or otherwise of IR The results of some randomized controlled trial RCT studies suggest that it is advisable to consume at least half of the carbohydrates at lunch and to avoid consuming large amounts of carbohydrates at breakfast or dinner in order to control blood glucose spikes, which may be related to diurnal variations in insulin sensitivity — Results of another study showed that 10 hours of restrictive eating improved quality of life by reducing body weight and improving blood glucose, insulin sensitivity and related metabolic disorders Other dietary strategies have been shown to prevent high-fat diet-induced IR, such as the intake of flavonoid-rich natural products, like flavonoids, which upregulate the expression of related genes through cell surface G protein-coupled estrogen receptors Although lifestyle modification and weight loss are highly recommended to improve IR and its associated metabolic disorders, they have limited effectiveness, slow onset of action, and low feasibility.

Pharmacological treatments to increase insulin sensitivity will be described next. Currently, the main drugs that can effectively improve IR are anti-hyperglycemic drugs, including metformin, thiazolidinediones TZD , sodium glucose cotransporter SGLT -2 inhibitors SGLT2i , etc.

Metformin, the most commonly used insulin-sensitizing agent, has been a guideline-recommended first-line treatment for T2DM for decades and has recently found new applications in the prevention and treatment of various diseases, including metabolic disorders and cardiovascular diseases Metformin improves IR by modulating metabolic mechanisms and mitochondrial biogenesis through altering microRNAs levels by AMPK-dependent or AMPK-independent mechanisms TZDs, such as pioglitazone, are potent insulin sensitizers targeting PPARγ and PI3K, regulating the transcription of nuclear transcription factors, stimulating mainly white adipose tissue remodeling, and regulating lipid flux for insulin sensitization and beta cell protection , SGLT2i is a relatively new class of glucose-lowering drug that not only lowers blood glucose by inhibiting renal glucose reuptake, leading to increased urinary glucose excretion and lower blood glucose, but also improves insulin sensitivity in patients with T2DM by reducing body weight or glucose toxicity , And in a randomized, double-blind, placebo-controlled clinical trial, it was shown that 8 weeks of treatment with SGLT2i empagliflozin restored insulin sensitivity in the hypothalamus of patients with prediabetes Glucose-lowering drugs have also shown good, stacked effects in patients who do not have good response with one drug alone.

For example, the addition of rosiglitazone to metformin can be clinically important in improving glycemic control, insulin sensitivity and beta-cell function The addition of sitagliptin or metformin to pioglitazone monotherapy also leads to faster and better improvement in IR and inflammatory status parameters Other therapies, as well as some new drugs in clinical trials, such as anti-inflammatory drugs, drugs that target hepatic lipid and energy metabolism, renin-angiotensin-aldosterone system blockers, vitamin D, antioxidants, probiotics and fecal transplants, have also shown improvement in IR Among them, selected clinical trials in the last decade have been listed in Table 2.

As mentioned previously, low-grade chronic inflammation is associated with IR and metabolic disturbances. For example, in in vitro and in vivo mouse models of diet-induced hyperinsulinemia, low-dose naltrexone attenuates hyperinsulinemia-induced proinflammatory cytokine release and restores insulin sensitivity However, it is worth noting that corticosteroids can cause IR and hyperglycemia due to their metabolic effects, and statins also increase the risk of IR, although they can reduce circulating inflammatory markers TCM plays an equally critical role in the treatment of many acute and chronic diseases, especially its adeptness in restoring the dynamic balance of the body in systemic diseases.

Its main therapeutic measures include herbal medicine, acupuncture and Tui Na. Several classical herbal formulations have been widely used in the clinical treatment of T2DM and various other metabolic disorders. For example, GegenQinlian decoction improves IR in fat, liver and muscle tissue through a variety of compounds, targets, pathways and mechanisms Yi Qi Zeng Min Tang has been shown to improve IR in high-fat fed Sprague-Dawley rats without increasing body weight Because it reduced the expression of PI3K p85 mRNA and IRS1 protein, Fu Fang Zhen Zhu Tiao Zhi formula similarly improved IR in vitro and in rats with metabolic syndrome Gui Zhi Fu Ling Wan, Dingkun Pill and Liuwei Dihuang Pills are herbal formulas widely used in the treatment of gynecological disorders and have the effect of harmonizing Qi and blood or dispelling blood stasis in Chinese medical theory.

In addition, the efficacy of acupuncture in improving IR is equally impressive, as a recent meta-analysis showed that acupuncture improved HOMA-IR and ISI as well as fasting blood glucose FBG , 2h postprandial blood glucose 2hPG and fasting insulin FINS levels, with fewer adverse events The increased incidence of IR and its vital role as a major and common cause of numerous metabolic diseases have created an urgent need to gain insight into the etiology and pathogenesis of IR, as well as to explore better early diagnostic methods and therapeutic strategies for it.

The diagnosis of insulin resistance is currently inconclusive, while it is important to detect IR early and predict individual response to treatment. In addition to the few simple indices of IR calculated from biochemical or anthropometric variables currently in use, emerging biomarkers may now be the way forward, but this still needs to be supported by clinical data.

Different ranges and criteria are also needed for the diagnosis and monitoring of different metabolic diseases. As mentioned above, IR is a central mechanism in many metabolic diseases. Since this is the case, IR should be considered as a therapeutic target for patients with a combination of multiple metabolic diseases so that multiple diseases can be treated simultaneously with the same treatment approach, thereby reducing healthcare expenditures.

Although there is no universally accepted theory to explain the mechanisms that cause IR. Nevertheless, there is growing evidence linking ectopic lipid accumulation, ER stress, plasma concentration of inflammatory cytokines, oxidative stress, abnormalities in insulin signaling, and other factors to IR.

In recent years, the exploration of the molecular mechanisms of IR has also led to the emergence of new therapeutic concepts beyond metformin and TZD. Regardless lifestyle modification remains the most basic and least costly intervention.

Normative criteria need to be developed for different metabolic diseases considering IR as a focus. FL and HJ provided the idea of the manuscript. XZ, XA, and CY contributed equally to this manuscript. XZ, XA, WS, and CY drafted the manuscript and searched the relevant literature.

XZ and XA drafted the figures, and all authors approved the final version of the manuscript. All authors agree to be accountable for all aspects of work ensuring integrity and accuracy. All authors contributed to the article and approved the submitted version.

This work was supported by Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine. No: ZYYCXTD-D The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

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

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Bugianesi E, McCullough AJ, Marchesini G. Insulin resistance: a metabolic pathway to chronic liver disease.

Hepatology 42 5 — doi: PubMed Abstract CrossRef Full Text Google Scholar. Sharma VR, Matta ST, Haymond MW, Chung ST. Measuring insulin resistance in humans. Horm Res Paediatr 93 — Muniyappa R, Madan R, Varghese RT. Assessing insulin sensitivity and resistance in humans. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, editors.

South Dartmouth MA: MDText. com; Inc. Copyright © ; MDText. Google Scholar. Gar C, Rottenkolber M, Prehn C, Adamski J, Seissler J, Lechner A. Serum and plasma amino acids as markers of prediabetes; insulin resistance; and incident diabetes. Crit Rev Clin Lab Sci 55 1 — Park SE, Park CY, Sweeney G.

Biomarkers of insulin sensitivity and insulin resistance: Past; present and future. Crit Rev Clin Lab Sci 52 4 — Milburn MV, Lawton KA. Application of metabolomics to diagnosis of insulin resistance.

Annu Rev Med — Yang R, Hu Y, Lee CH, Liu Y, Diaz-Canestro C, Fong CHY, et al. PM20D1 is a circulating biomarker closely associated with obesity; insulin resistance and metabolic syndrome. Eur J Endocrinol 2 — Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 20 2 Chooi YC, Ding C, Magkos F.

The epidemiology of obesity. Metabolism — Steenblock C, Schwarz PEH, Ludwig B, Linkermann A, Zimmet P, Kulebyakin K, et al. COVID and metabolic disease: mechanisms and clinical management.

Lancet Diabetes Endocrinol 9 11 — Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al.

The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J Hepatol 71 4 — Lonardo A, Nascimbeni F, Mantovani A, Targher G.

Hypertension; diabetes; atherosclerosis and NASH: Cause or consequence? Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF diabetes atlas: Global; regional and country-level diabetes prevalence estimates for and projections for Diabetes Res Clin Pract Kaul K, Apostolopoulou M, Roden M.

Insulin resistance in type 1 diabetes mellitus. Metabolism 64 12 — Nadeau KJ, Regensteiner JG, Bauer TA, Brown MS, Dorosz JL, Hull A, et al. Insulin resistance in adolescents with type 1 diabetes and its relationship to cardiovascular function.

J Clin Endocrinol Metab 95 2 — Cree-Green M, Newcomer BR, Brown MS, Baumgartner AD, Bergman B, Drew B, et al. Delayed skeletal muscle mitochondrial ADP recovery in youth with type 1 diabetes relates to muscle insulin resistance. Diabetes 64 2 — Schauer IE, Snell-Bergeon JK, Bergman BC, Maahs DM, Kretowski A, Eckel RH, et al.

Insulin resistance; defective insulin-mediated fatty acid suppression; and coronary artery calcification in subjects with and without type 1 diabetes: The CACTI study.

Diabetes 60 1 — Donga E, Dekkers OM, Corssmit EP, Romijn JA. Insulin resistance in patients with type 1 diabetes assessed by glucose clamp studies: systematic review and meta-analysis. Eur J Endocrinol 1 —9.

Liu HY, Cao SY, Hong T, Han J, Liu Z, Cao W. Insulin is a stronger inducer of insulin resistance than hyperglycemia in mice with type 1 diabetes mellitus T1DM. J Biol Chem 40 — Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes.

Cell Metab 29 5 — Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46 1 :3— Kahn SE, Hull RL, Utzschneider KM.

Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature —6. Rattarasarn C. Dysregulated lipid storage and its relationship with insulin resistance and cardiovascular risk factors in non-obese asian patients with type 2 diabetes. Adipocyte 7 2 — Pop A, Clenciu D, Anghel M, Radu S, Socea B, Mota E, et al.

Insulin resistance is associated with all chronic complications in type 1 diabetes. J Diabetes. Pan Y, Zhong S, Zhou K, Tian Z, Chen F, Liu Z, et al. Association between diabetes complications and the triglyceride-glucose index in hospitalized patients with type 2 diabetes.

J Diabetes Res Wang S, Shi J, Peng Y, Fang Q, Mu Q, Gu W, et al. Stronger association of triglyceride glucose index than the HOMA-IR with arterial stiffness in patients with type 2 diabetes: a real-world single-centre study. Cardiovasc Diabetol 20 1 Jia G, Whaley-Connell A, Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease.

Diabetologia 61 1 —8. Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy.

Nat Rev Endocrinol 12 3 — Svensson M, Eriksson JW. Insulin resistance in diabetic nephropathy—cause or consequence? Diabetes Metab Res Rev 22 5 — Godsland IF.

Insulin resistance and hyperinsulinaemia in the development and progression of cancer. Clin Sci Lond. Hernandez AV, Pasupuleti V, Benites-Zapata VA, Thota P, Deshpande A, Perez-Lopez FR.

Insulin resistance and endometrial cancer risk: A systematic review and meta-analysis. Eur J Cancer.

Yin DT, He H, Yu K, Xie J, Lei M, Ma R, et al. The association between thyroid cancer and insulin resistance; metabolic syndrome and its components: A systematic review and meta-analysis.

Int J Surg , — CrossRef Full Text Google Scholar. Kim NH, Chang Y, Lee SR, Ryu S, Kim HJ. Glycemic status; insulin resistance; and risk of pancreatic cancer mortality in individuals with and without diabetes.

Am J Gastroenterol 11 —8. Pan K, Chlebowski RT, Mortimer JE, Gunter MJ, Rohan T, Vitolins MZ, et al.

Insulin resistance and breast cancer incidence and mortality in postmenopausal women in the women's health initiative. Cancer 16 — Chiefari E, Mirabelli M, La Vignera S, Tanyolaç S, Foti DP, Aversa A, et al. Insulin resistance and cancer: In search for a causal link. Int J Mol Sci 22 Barber TM, Kyrou I, Randeva HS, Weickert MO.

Mechanisms of insulin resistance at the crossroad of obesity with associated metabolic abnormalities and cognitive dysfunction.

Int J Mol Sci 22 2. Mu N, Zhu Y, Wang Y, Zhang H, Xue F. Insulin resistance: a significant risk factor of endometrial cancer. Gynecol Oncol 3 —7. Arcidiacono B, Iiritano S, Nocera A, Possidente K, Nevolo MT, Ventura V, et al.

Insulin resistance and cancer risk: an overview of the pathogenetic mechanisms. Exp Diabetes Res Inoue M, Tsugane S. Insulin resistance and cancer: epidemiological evidence. Endocr Relat Cancer 19 5 :F1—8. Kong Y, Hsieh CH, Alonso LC. Front Endocrinol Lausanne Ramos-Lopez O, Riezu-Boj JI, Milagro FI, Martinez JA.

DNA methylation signatures at endoplasmic reticulum stress genes are associated with adiposity and insulin resistance. Mol Genet Metab 1 —8. Kwa M, Plottel CS, Blaser MJ, Adams S. The intestinal microbiome and estrogen receptor-positive female breast cancer.

J Natl Cancer Inst 8. Adeva-Andany MM, Martínez-Rodríguez J, González-Lucán M, Fernández-Fernández C, Castro-Quintela E. Insulin resistance is a cardiovascular risk factor in humans. Diabetes Metab Syndr 13 2 — Adeva-Andany MM, Fernández-Fernández C, Carneiro-Freire N, Castro-Quintela E, Pedre-Piñeiro A, Seco-Filgueira M.

Insulin resistance underlies the elevated cardiovascular risk associated with kidney disease and glomerular hyperfiltration. Rev Cardiovasc Med 21 1 — Saely CH, Aczel S, Marte T, Langer P, Hoefle G, Drexel H. The metabolic syndrome; insulin resistance; and cardiovascular risk in diabetic and nondiabetic patients.

J Clin Endocrinol Metab 90 10 — Zhang X, Li J, Zheng S, Luo Q, Zhou C, Wang C. Fasting insulin; insulin resistance; and risk of cardiovascular or all-cause mortality in non-diabetic adults: a meta-analysis.

Biosci Rep 37 5. Eddy D, Schlessinger L, Kahn R, Peskin B, Schiebinger R. Relationship of insulin resistance and related metabolic variables to coronary artery disease: a mathematical analysis.

Diabetes Care 32 2 —6. Novo G, Manno G, Russo R, Buccheri D, Dell'Oglio S, Morreale P, et al. Impact of insulin resistance on cardiac and vascular function. Int J Cardiol —9. Wang M, Li Y, Li S, Lv J.

Endothelial dysfunction and diabetic cardiomyopathy. Front Endocrinol Nakamura M, Sadoshima J. Cardiomyopathy in obesity, insulin resistance and diabetes. J Physiol 14 — Qi Y, Xu Z, Zhu Q, Thomas C, Kumar R, Feng H, et al. Myocardial loss of IRS1 and IRS2 causes heart failure and is controlled by p38α MAPK during insulin resistance.

Diabetes 62 11 — Razani B, Chakravarthy MV, Semenkovich CF. Insulin resistance and atherosclerosis. Endocrinol Metab Clin North Am 37 3 — Impaired nutrition contributes to hyperlipidemia and insulin resistance causing hyperglycemia.

This condition alters cellular metabolism and intracellular signaling that negatively impact cells. In the cardiomyocyte, this damage can be summarized into three actions: 1 alteration in insulin signaling. All these effects induce cellular events including: 1 gene expression modifications, 2 hyperglycemia and dyslipidemia, 3 activation of oxidative stress and inflammatory response, 4 endothelial dysfunction, and 5 ectopic lipid accumulation, which, favored by obesity, perpetuates the metabolic deregulation.

Overall, insulin resistance contributes to generate CVD via two independent pathways: 1 atheroma plaque formation and 2 ventricular hypertrophy and diastolic abnormality.

Both effects lead to heart failure. Future research is needed to understand the precise mechanism between insulin resistance and its progression to heart failure with a focus on new therapy development.

Steinberger J, Daniels SR, American Heart Association Atherosclerosis H, Obesity in the Young C, American Heart Association Diabetes C. Obesity, insulin resistance, diabetes, and cardiovascular risk in children: an American Heart Association scientific statement from the Atherosclerosis, Hypertension, and Obesity in the Young Committee Council on Cardiovascular Disease in the Young and the Diabetes Committee Council on Nutrition, Physical Activity, and Metabolism.

Article PubMed Google Scholar. Steinberger J, Moorehead C, Katch V, Rocchini AP. Relationship between insulin resistance and abnormal lipid profile in obese adolescents.

J Pediatr. Article PubMed CAS Google Scholar. Ferreira AP, Oliveira CE, Franca NM. Metabolic syndrome and risk factors for cardiovascular disease in obese children: the relationship with insulin resistance HOMA-IR.

Jornal de pediatria. Reaven G. Insulin resistance and coronary heart disease in nondiabetic individuals. Arterioscler Thromb Vasc Biol.

Wilcox G. Insulin and insulin resistance. Clin Biochem Rev. PubMed PubMed Central Google Scholar. Gast KB, Tjeerdema N, Stijnen T, Smit JW, Dekkers OM.

Insulin resistance and risk of incident cardiovascular events in adults without diabetes: meta-analysis. PLoS ONE. Article PubMed PubMed Central CAS Google Scholar. Bornfeldt KE, Tabas I. Insulin resistance, hyperglycemia, and atherosclerosis.

Cell Metab. Davidson JA, Parkin CG. Is hyperglycemia a causal factor in cardiovascular disease? Does proving this relationship really matter? Diabetes Care. Article PubMed PubMed Central Google Scholar. Laakso M, Kuusisto J.

Insulin resistance and hyperglycaemia in cardiovascular disease development. Nat Rev Endocrinol. Janus A, Szahidewicz-Krupska E, Mazur G, Doroszko A. Insulin resistance and endothelial dysfunction constitute a common therapeutic target in cardiometabolic disorders.

Mediators Inflamm. Scott PH, Brunn GJ, Kohn AD, Roth RA, Lawrence JC Jr. Evidence of insulin-stimulated phosphorylation and activation of the mammalian target of rapamycin mediated by a protein kinase B signaling pathway.

Proc Natl Acad Sci USA. Bogan JS. Regulation of glucose transporter translocation in health and diabetes. Annu Rev Biochem. Zimmer HG. Regulation of and intervention into the oxidative pentose phosphate pathway and adenine nucleotide metabolism in the heart.

Mol Cell Biochem. Choi SM, Tucker DF, Gross DN, Easton RM, DiPilato LM, Dean AS, Monks BR, Birnbaum MJ. Insulin regulates adipocyte lipolysis via an Akt-independent signaling pathway. Mol Cell Biol. Duncan RE, Ahmadian M, Jaworski K, Sarkadi-Nagy E, Sul HS. Regulation of lipolysis in adipocytes. Annu Rev Nutr.

Czech MP, Tencerova M, Pedersen DJ, Aouadi M. Insulin signalling mechanisms for triacylglycerol storage. Shulman GI. Cellular mechanisms of insulin resistance. J Clin Investig. Hojlund K. Metabolism and insulin signaling in common metabolic disorders and inherited insulin resistance.

Dan Med J. PubMed Google Scholar. Kahn BB, Flier JS. Obesity and insulin resistance. Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, Raptis SA. Insulin effects in muscle and adipose tissue. Diabetes Res Clin Pract. Reaven GM. Pathophysiology of insulin resistance in human disease.

Physiol Rev. Wu G, Meininger CJ. Nitric oxide and vascular insulin resistance. BioFactors Oxford, England. Article CAS Google Scholar. Wang CC, Gurevich I, Draznin B. Insulin affects vascular smooth muscle cell phenotype and migration via distinct signaling pathways.

Berg J, Tymoczko J, Stryer L: Food intake and starvation induce metabolic changes. In: Biochemistry. Catalano PM. Obesity, insulin resistance and pregnancy outcome. Reproduction Cambridge, England. Bonora E. Insulin resistance as an independent risk factor for cardiovascular disease: clinical assessment and therapy approaches.

Av Diabetol. Google Scholar. Goodwin PJ, Ennis M, Bahl M, Fantus IG, Pritchard KI, Trudeau ME, Koo J, Hood N. High insulin levels in newly diagnosed breast cancer patients reflect underlying insulin resistance and are associated with components of the insulin resistance syndrome.

Breast Cancer Res Treat. Seriolo B, Ferrone C, Cutolo M. Longterm anti-tumor necrosis factor-alpha treatment in patients with refractory rheumatoid arthritis: relationship between insulin resistance and disease activity. J Rheumatol. PubMed CAS Google Scholar.

Williams T, Mortada R, Porter S. Diagnosis and treatment of polycystic ovary syndrome. Am Fam Physician. Lallukka S, Yki-Jarvinen H. Non-alcoholic fatty liver disease and risk of type 2 diabetes.

Best Pract Res Clin Endocrinol Metab. Rader DJ. Effect of insulin resistance, dyslipidemia, and intra-abdominal adiposity on the development of cardiovascular disease and diabetes mellitus.

Am J Med. Wende AR, Abel ED. Lipotoxicity in the heart. Biochem Biophys Acta. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Wang CC, Goalstone ML, Draznin B. Molecular mechanisms of insulin resistance that impact cardiovascular biology. Moller DE, Kaufman KD. Metabolic syndrome: a clinical and molecular perspective.

Annu Rev Med. Matthaei S, Stumvoll M, Kellerer M, Haring HU. Pathophysiology and pharmacological treatment of insulin resistance. Endocr Rev. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links.

The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux. Tamemoto H, Kadowaki T, Tobe K, Yagi T, Sakura H, Hayakawa T, Terauchi Y, Ueki K, Kaburagi Y, Satoh S, et al. Insulin resistance and growth retardation in mice lacking insulin receptor substrate Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, Zhang Y, Bernal D, Pons S, Shulman GI, et al.

Disruption of IRS-2 causes type 2 diabetes in mice. Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw EB 3rd, Kaestner KH, Bartolomei MS, Shulman GI, Birnbaum MJ.

Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 PKB beta. Saini V.

Molecular mechanisms of insulin resistance in type 2 diabetes mellitus. World J Diabetes. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, Slezak LA, Andersen DK, Hundal RS, Rothman DL, et al. Effects of free fatty acids on glucose transport and IRSassociated phosphatidylinositol 3-kinase activity.

Sinha R, Dufour S, Petersen KF, LeBon V, Enoksson S, Ma YZ, Savoye M, Rothman DL, Shulman GI, Caprio S. Assessment of skeletal muscle triglyceride content by 1 H nuclear magnetic resonance spectroscopy in lean and obese adolescents: relationships to insulin sensitivity, total body fat, and central adiposity.

Unger RH, Orci L. Lipotoxic diseases of nonadipose tissues in obesity. Int J Obes Related Metab Dis. Dong B, Qi D, Yang L, Huang Y, Xiao X, Tai N, Wen L, Wong FS.

TLR4 regulates cardiac lipid accumulation and diabetic heart disease in the nonobese diabetic mouse model of type 1 diabetes.

Am J Physiol Heart Circ Physiol. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue.

Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance.

Draznin B. Molecular mechanisms of insulin resistance: serine phosphorylation of insulin receptor substrate-1 and increased expression of p85 alpha—the two sides of a coin. Tremblay F, Krebs M, Dombrowski L, Brehm A, Bernroider E, Roth E, Nowotny P, Waldhausl W, Marette A, Roden M.

Overactivation of S6 kinase 1 as a cause of human insulin resistance during increased amino acid availability. Chiang GG, Abraham RT. Phosphorylation of mammalian target of rapamycin mTOR at ser is mediated by p70S6 kinase.

J Biol Chem. Gao Z, Zhang X, Zuberi A, Hwang D, Quon MJ, Lefevre M, Ye J. Inhibition of insulin sensitivity by free fatty acids requires activation of multiple serine kinases in 3T3-L1 adipocytes. Mol Endocrinol. Aroor AR, Mandavia CH, Sowers JR.

Insulin resistance and heart failure: molecular mechanisms. Heart Fail Clin. Flegal KM, Graubard BI, Williamson DF, Gail MH. Excess deaths associated with underweight, overweight, and obesity. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA.

The hormone resistin links obesity to diabetes. Liu L, Feng J, Zhang G, Yuan X, Li F, Yang T, Hao S, Huang D, Hsue C, Lou Q. Visceral adipose tissue is more strongly associated with insulin resistance than subcutaneous adipose tissue in Chinese subjects with pre-diabetes.

Curr Med Res Opin. Palmer BF, Clegg DJ. The sexual dimorphism of obesity. Mol Cell Endocrinol. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease.

N Engl J Med. Lalia AZ, Dasari S, Johnson ML, Robinson MM, Konopka AR, Distelmaier K, Port JD, Glavin MT, Esponda RR, Nair KS, et al. Predictors of whole-body insulin sensitivity across ages and adiposity in adult humans. J Clin Endocrinol Metab.

Gonzalez N, Moreno-Villegas Z, Gonzalez-Bris A, Egido J, Lorenzo O. Regulation of visceral and epicardial adipose tissue for preventing cardiovascular injuries associated to obesity and diabetes.

Cardiovasc Diabetol. Kim JI, Huh JY, Sohn JH, Choe SS, Lee YS, Lim CY, Jo A, Park SB, Han W, Kim JB. Lipid-overloaded enlarged adipocytes provoke insulin resistance independent of inflammation.

Alman AC, Smith SR, Eckel RH, Hokanson JE, Burkhardt BR, Sudini PR, Wu Y, Schauer IE, Pereira RI, Snell-Bergeon JK. The ratio of pericardial to subcutaneous adipose tissues is associated with insulin resistance.

Obesity Silver Spring, Md. Fitzgibbons TP, Czech MP. Epicardial and perivascular adipose tissues and their influence on cardiovascular disease: basic mechanisms and clinical associations.

J Am Heart Assoc. Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol. Iacobellis G, Ribaudo MC, Zappaterreno A, Iannucci CV, Leonetti F. Relation between epicardial adipose tissue and left ventricular mass.

Am J Cardiol. Rijzewijk LJ, van der Meer RW, Smit JW, Diamant M, Bax JJ, Hammer S, Romijn JA, de Roos A, Lamb HJ. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus.

J Am Coll Cardiol. Nyman K, Granér M, Pentikäinen MO, Lundbom J, Hakkarainen A, Sirén R, Nieminen MS, Taskinen M-R, Lundbom N, Lauerma K. Cardiac steatosis and left ventricular function in men with metabolic syndrome.

J Cardiovasc Magn Reson. Abel ED, Litwin SE, Sweeney G. Cardiac remodeling in obesity. Bonora E, Kiechl S, Willeit J, Oberhollenzer F, Egger G, Targher G, Alberiche M, Bonadonna RC, Muggeo M. Prevalence of insulin resistance in metabolic disorders: the Bruneck Study.

Insulin sensitivity and atherosclerosis. The Insulin Resistance Atherosclerosis Study IRAS Investigators. Tenenbaum A, Adler Y, Boyko V, Tenenbaum H, Fisman EZ, Tanne D, Lapidot M, Schwammenthal E, Feinberg MS, Matas Z, et al.

Insulin resistance is associated with increased risk of major cardiovascular events in patients with preexisting coronary artery disease. Am Heart J. Eddy D, Schlessinger L, Kahn R, Peskin B, Schiebinger R. Relationship of insulin resistance and related metabolic variables to coronary artery disease: a mathematical analysis.

Savaiano DA, Story JA. Cardiovascular disease and fiber: is insulin resistance the missing link? Nutr Rev. Kong C, Elatrozy T, Anyaoku V, Robinson S, Richmond W, Elkeles RS. Insulin resistance, cardiovascular risk factors and ultrasonically measured early arterial disease in normotensive Type 2 diabetic subjects.

Diabetes Metab Res Rev. Ginsberg HN. Insulin resistance and cardiovascular disease. Bloomgarden ZT. Insulin resistance, dyslipidemia, and cardiovascular disease. Kozakova M, Natali A, Dekker J, Beck-Nielsen H, Laakso M, Nilsson P, Balkau B, Ferrannini E.

Insulin sensitivity and carotid intima-media thickness: relationship between insulin sensitivity and cardiovascular risk study. Min J, Weitian Z, Peng C, Yan P, Bo Z, Yan W, Yun B, Xukai W. Correlation between insulin-induced estrogen receptor methylation and atherosclerosis.

Chanda D, Luiken JJ, Glatz JF. Signaling pathways involved in cardiac energy metabolism. FEBS Lett. Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D, Orci L, Unger RH.

Lipotoxic heart disease in obese rats: implications for human obesity. Ramírez E, Picatoste B, González-Bris A, Oteo M, Cruz F, Caro-Vadillo A, Egido J, Tuñón J, Morcillo MA, Lorenzo Ó.

Sitagliptin improved glucose assimilation in detriment of fatty-acid utilization in experimental type-II diabetes: role of GLP-1 isoforms in Glut4 receptor trafficking. Goldberg IJ. Clinical review diabetic dyslipidemia: causes and consequences.

Sparks JD, Sparks CE, Adeli K. Selective hepatic insulin resistance, VLDL overproduction, and hypertriglyceridemia. Zimmet P, Alberti KG, Shaw J. Global and societal implications of the diabetes epidemic.

Austin MA, Hokanson JE, Edwards KL. Hypertriglyceridemia as a cardiovascular risk factor. Hokanson JE. Hypertriglyceridemia and risk of coronary heart disease.

Curr Cardiol Rep. Sung KC, Park HY, Kim MJ, Reaven G. Metabolic markers associated with insulin resistance predict type 2 diabetes in Koreans with normal blood pressure or prehypertension.

Ginsberg HN, Zhang YL, Hernandez-Ono A. Metabolic syndrome: focus on dyslipidemia. Yadav R, Hama S, Liu Y, Siahmansur T, Schofield J, Syed AA, France M, Pemberton P, Adam S, Ho JH, et al. Effect of Roux-en-Y bariatric surgery on lipoproteins, insulin resistance, and systemic and vascular inflammation in obesity and diabetes.

Front Immunol. de Luca C, Olefsky JM. Inflammation and insulin resistance. den Boer MA, Voshol PJ, Kuipers F, Romijn JA, Havekes LM. Hepatic glucose production is more sensitive to insulin-mediated inhibition than hepatic VLDL-triglyceride production.

Am J Physiol Endocrinol Metab. Semenkovich CF. Insulin resistance and atherosclerosis. Lewis GF, Steiner G. Acute effects of insulin in the control of VLDL production in humans. Implications for the insulin-resistant state.

Haas ME, Attie AD, Biddinger SB. The regulation of ApoB metabolism by insulin. Trends Endocrinol Metab.

Verges B. Pathophysiology of diabetic dyslipidaemia: where are we? Pont F, Duvillard L, Florentin E, Gambert P, Verges B. Early kinetic abnormalities of apoB-containing lipoproteins in insulin-resistant women with abdominal obesity.

Hoogeveen RC, Gaubatz JW, Sun W, Dodge RC, Crosby JR, Jiang J, Couper D, Virani SS, Kathiresan S, Boerwinkle E, et al. Small dense low-density lipoprotein-cholesterol concentrations predict risk for coronary heart disease: the Atherosclerosis Risk in Communities ARIC study.

Packard CJ. Triacylglycerol-rich lipoproteins and the generation of small, dense low-density lipoprotein. Biochem Soc Trans. Sandhofer A, Kaser S, Ritsch A, Laimer M, Engl J, Paulweber B, Patsch JR, Ebenbichler CF.

Cholesteryl ester transfer protein in metabolic syndrome. Rashid S, Watanabe T, Sakaue T, Lewis GF. Mechanisms of HDL lowering in insulin resistant, hypertriglyceridemic states: the combined effect of HDL triglyceride enrichment and elevated hepatic lipase activity.

Clin Biochem. von Bibra H, Saha S, Hapfelmeier A, Muller G, Schwarz PEH. Kim MK, Ahn CW, Kang S, Nam JS, Kim KR, Park JS. Relationship between the triglyceride glucose index and coronary artery calcification in Korean adults. Mazidi M, Kengne AP, Katsiki N, Mikhailidis DP, Banach M. J Diabetes Complications.

Jorge-Galarza E, Posadas-Romero C, Torres-Tamayo M, Medina-Urrutia AX, Rodas-Diaz MA, Posadas-Sanchez R, Vargas-Alarcon G, Gonzalez-Salazar MD, Cardoso-Saldana GC, Juarez-Rojas JG. Insulin resistance in adipose tissue but not in liver is associated with aortic valve calcification.

Dis Markers. Zhou MS, Schulman IH, Zeng Q. Link between the renin—angiotensin system and insulin resistance: implications for cardiovascular disease. Vasc Med. Zhou MS, Schulman IH, Raij L. Nitric oxide, angiotensin II, and hypertension. Semin Nephrol. Landsberg L. Insulin resistance and hypertension.

Clin Exp Hypertens. Briet M, Schiffrin EL. Aldosterone: effects on the kidney and cardiovascular system. Nat Rev Nephrol. Oana F, Takeda H, Hayakawa K, Matsuzawa A, Akahane S, Isaji M, Akahane M.

Goossens GH. The renin—angiotensin system in the pathophysiology of type 2 diabetes. Obesity Facts. Schulman IH, Zhou MS. Vascular insulin resistance: a potential link between cardiovascular and metabolic diseases.

Curr Hypertens Rep. Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Vascular inflammation, insulin resistance, and endothelial dysfunction in salt-sensitive hypertension: role of nuclear factor kappa B activation.

J Hypertens. Andreozzi F, Laratta E, Sciacqua A, Perticone F, Sesti G. Angiotensin II impairs the insulin signaling pathway promoting production of nitric oxide by inducing phosphorylation of insulin receptor substrate-1 on Ser and Ser in human umbilical vein endothelial cells.

Circ Res. Wei Y, Whaley-Connell AT, Chen K, Habibi J, Uptergrove GM, Clark SE, Stump CS, Ferrario CM, Sowers JR. NADPH oxidase contributes to vascular inflammation, insulin resistance, and remodeling in the transgenic mRen2 rat. Matsuura K, Hagiwara N.

The pleiotropic effects of ARB in vascular endothelial progenitor cells. Curr Vasc Pharmacol. Group NS, McMurray JJ, Holman RR, Haffner SM, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, et al.

Effect of valsartan on the incidence of diabetes and cardiovascular events. Article Google Scholar. Perlstein TS, Henry RR, Mather KJ, Rickels MR, Abate NI, Grundy SM, Mai Y, Albu JB, Marks JB, Pool JL, et al. Effect of angiotensin receptor blockade on insulin sensitivity and endothelial function in abdominally obese hypertensive patients with impaired fasting glucose.

Clin Sci Lond. Kim JA, Montagnani M, Koh KK, Quon MJ. Reciprocal relationships between insulin resistance and endothelial dysfunction: molecular and pathophysiological mechanisms. Tousoulis D, Simopoulou C, Papageorgiou N, Oikonomou E, Hatzis G, Siasos G, Tsiamis E, Stefanadis C.

Endothelial dysfunction in conduit arteries and in microcirculation. Novel therapeutic approaches. Pharmacol Ther. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Westergren HU, Svedlund S, Momo RA, Blomster JI, Wahlander K, Rehnstrom E, Greasley PJ, Fritsche-Danielson R, Oscarsson J, Gan LM.

Insulin resistance, endothelial function, angiogenic factors and clinical outcome in non-diabetic patients with chest pain without myocardial perfusion defects. Dinesh Shah A, Langenberg C, Rapsomaniki E, Denaxas S, Pujades-Rodriguez M, Gale CP, Deanfield J, Smeeth L, Timmis A, Hemingway H.

Type 2 diabetes and incidence of a wide range of cardiovascular diseases: a cohort study in 1. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: have all risk factors the same strength?

Ciccone MM, Cortese F, Gesualdo M, Donvito I, Carbonara S, De Pergola G. Endocr Metab Immune Disord Drug Targets. Selvin E, Marinopoulos S, Berkenblit G, Rami T, Brancati FL, Powe NR, Golden SH.

Meta-analysis: glycosylated hemoglobin and cardiovascular disease in diabetes mellitus. Ann Intern Med. Meyer ML, Gotman NM, Soliman EZ, Whitsel EA, Arens R, Cai J, Daviglus ML, Denes P, Gonzalez HM, Moreiras J, et al.

Paneni F, Volpe M, Luscher TF, Cosentino F. Ceriello A. Vasc Pharmacol. Fiorentino TV, Prioletta A, Zuo P, Folli F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases.

Curr Pharm Des. Pistrosch F, Natali A, Hanefeld M. Is hyperglycemia a cardiovascular risk factor? Giacco F, Brownlee M. Oxidative stress and diabetic complications. Nowotny K, Jung T, Hohn A, Weber D, Grune T.

Advanced glycation end products and oxidative stress in type 2 diabetes mellitus. Yan SF, Ramasamy R, Schmidt AM. The RAGE axis: a fundamental mechanism signaling danger to the vulnerable vasculature.

Sonnenblick EH, Stam AC Jr. Cardiac muscle: activation and contraction. Annu Rev Physiol. Johansen L, Quistorff B. Int J Sports Med. Duffield R, Dawson B, Goodman C.

Energy system contribution to m and m track running events. J Sci Med Sport. Kassiotis C, Rajabi M, Taegtmeyer H. Metabolic reserve of the heart: the forgotten link between contraction and coronary flow.

Prog Cardiovasc Dis. Kota SK, Kota SK, Jammula S, Panda S, Modi KD. Effect of diabetes on alteration of metabolism in cardiac myocytes: therapeutic implications. Diabetes Technol Ther. Stanley WC, Recchia FA, Lopaschuk GD.

Myocardial substrate metabolism in the normal and failing heart. Carley AN, Severson DL. Fatty acid metabolism is enhanced in type 2 diabetic hearts. Brandt JM, Djouadi F, Kelly DP. Fatty acids activate transcription of the muscle carnitine palmitoyltransferase I gene in cardiac myocytes via the peroxisome proliferator-activated receptor alpha.

Goodwin GW, Taegtmeyer H. Improved energy homeostasis of the heart in the metabolic state of exercise. Opie LH. Cardiac metabolism—emergence, decline, and resurgence. Part II.

Cardiovasc Res. Henning SL, Wambolt RB, Schonekess BO, Lopaschuk GD, Allard MF. Contribution of glycogen to aerobic myocardial glucose utilization. Wu G, Fang YZ, Yang S, Lupton JR, Turner ND.

Glutathione metabolism and its implications for health. The Journal of nutrition. Shao D, Tian R. Glucose transporters in cardiac metabolism and hypertrophy.

Comp Physiol. Malfitano C, de Souza Junior AL, Carbonaro M, Bolsoni-Lopes A, Figueroa D, de Souza LE, Silva KA, Consolim-Colombo F, Curi R, Irigoyen MC. Glucose and fatty acid metabolism in infarcted heart from streptozotocin-induced diabetic rats after 2 weeks of tissue remodeling.

Kolwicz SC Jr, Purohit S, Tian R. Cardiac metabolism and its interactions with contraction, growth, and survival of cardiomyocytes. Wright JJ, Kim J, Buchanan J, Boudina S, Sena S, Bakirtzi K, Ilkun O, Theobald HA, Cooksey RC, Kandror KV, et al.

Mechanisms for increased myocardial fatty acid utilization following short-term high-fat feeding. Su X, Abumrad NA. Cellular fatty acid uptake: a pathway under construction. Ajith TA, Jayakumar TG. Peroxisome proliferator-activated receptors in cardiac energy metabolism and cardiovascular disease.

Clin Exp Pharmacol Physiol. Oakes ND, Thalen P, Aasum E, Edgley A, Larsen T, Furler SM, Ljung B, Severson D. Cardiac metabolism in mice: tracer method developments and in vivo application revealing profound metabolic inflexibility in diabetes.

Lipid metabolism and signaling in cardiac lipotoxicity. Goldberg IJ, Trent CM, Schulze PC. Lipid metabolism and toxicity in the heart. Lipoapoptosis: its mechanism and its diseases. Park TS, Hu Y, Noh HL, Drosatos K, Okajima K, Buchanan J, Tuinei J, Homma S, Jiang XC, Abel ED, et al.

Ceramide is a cardiotoxin in lipotoxic cardiomyopathy. J Lipid Res. Liu Y, Neumann D, Glatz JF, Luiken JJ. Molecular mechanism of lipid-induced cardiac insulin resistance and contractile dysfunction. Prostaglandins Leukot Essent Fatty Acids. Article PubMed Central PubMed Google Scholar.

Feuvray D, Idell-Wenger JA, Neely JR. Effects of ischemia on rat myocardial function and metabolism in diabetes. Fricovsky ES, Suarez J, Ihm SH, Scott BT, Suarez-Ramirez JA, Banerjee I, Torres-Gonzalez M, Wang H, Ellrott I, Maya-Ramos L, et al.

Excess protein O -GlcNAcylation and the progression of diabetic cardiomyopathy. Am J Physiol Regul Integr Comp Physiol. Hwang YC, Kaneko M, Bakr S, Liao H, Lu Y, Lewis ER, Yan S, Ii S, Itakura M, Rui L, et al.

Central role for aldose reductase pathway in myocardial ischemic injury. FASEB J. Zuurbier CJ, Eerbeek O, Goedhart PT, Struys EA, Verhoeven NM, Jakobs C, Ince C. Inhibition of the pentose phosphate pathway decreases ischemia—reperfusion-induced creatine kinase release in the heart.

Salabei JK, Lorkiewicz PK, Mehra P, Gibb AA, Haberzettl P, Hong KU, Wei X, Zhang X, Li Q, Wysoczynski M, et al. Type 2 Diabetes Dysregulates Glucose Metabolism in Cardiac Progenitor Cells. Keller U, Lustenberger M, Stauffacher W. van der Vusse GJ, van Bilsen M, Glatz JF. Cardiac fatty acid uptake and transport in health and disease.

Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, Koves T, Gardell SJ, Kruger M, Hoppel CL, et al. The failing heart relies on ketone bodies as a fuel. Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P, Ng CP, Huang Y, Haldar S, Verdin E.

Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell metabolism. Article PubMed CAS PubMed Central Google Scholar. Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte GR, Tran D, Perez G, Gutierrez-Casado E, Koike S, Knotts TA, et al.

A ketogenic diet extends longevity and healthspan in adult mice. Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini DM. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing.

Kosinski C, Jornayvaz FR: Effects of Ketogenic Diets on Cardiovascular Risk Factors: Evidence from Animal and Human Studies.

Nutrients , 9 5. Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ. Comparison of the atkins, ornish, weight watchers, and zone diets for weight loss and heart disease risk reduction: a randomized trial. Kim JA, Wei Y, Sowers JR.

Role of mitochondrial dysfunction in insulin resistance. Jeong EM, Chung J, Liu H, Go Y, Gladstein S, Farzaneh-Far A, Lewandowski ED, Dudley SC Jr. Role of mitochondrial oxidative stress in glucose tolerance, insulin resistance, and cardiac diastolic dysfunction.

Mei Y, Thompson MD, Cohen RA, Tong X. Endoplasmic reticulum stress and related pathological processes. J Pharm Biomed Anal. Taddeo EP, Laker RC, Breen DS, Akhtar YN, Kenwood BM, Liao JA, Zhang M, Fazakerley DJ, Tomsig JL, Harris TE, et al. Opening of the mitochondrial permeability transition pore links mitochondrial dysfunction to insulin resistance in skeletal muscle.

Mol Metab. Mandavia CH, Aroor AR, Demarco VG, Sowers JR. Molecular and metabolic mechanisms of cardiac dysfunction in diabetes. Life Sci. Download references. VO, SN, OE, CA and FZ conducted a review of the literature and contributed to conception and design and wrote the first draft the review; CS contributed to conception and design of the article and critically reviewed the drafts of the manuscript.

All authors read and approved the final manuscript. This study was supported by Fondo Nacional de Desarrollo Científico y Tecnológico FONDECYT , Lions Medical Research Foundation Australia , and Diabetes Australia. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile. Faculty of Pharmacy, Department of Clinical Biochemistry and Immunology, University of Concepcion, Concepción, Chile.

Department of Obstetrics and Gynecology, Ochsner Baptist Hospital, New Orleans, Louisiana, USA. You can also search for this author in PubMed Google Scholar. Correspondence to Carlos Salomon. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.

Reprints and permissions.

14 Natural Ways to Improve Your Insulin Sensitivity

A relatively safe and well accepted approach in the prevention and treatment of IR is via lifestyle interventions. Nutritional intervention is an important first step that emphasizes a low-calorie and low-fat diet that stimulates excessive insulin demands. In addition, increased physical activity is recommended to help increase energy expenditures and improve muscle insulin sensitivity, this two approach represent the fundamental treatment for IR.

In this review, the mechanism of insulin action and IR are first described to promote the development of new therapeutic strategies.

Further, the direct and indirect effects of insulin on target tissues are discussed to better understand the pivotal role of tissue crosstalk in systemic insulin action. Lastly, diseases associated with IR are discussed and summarized. Many methods and multiple surrogate markers have been developed to calculate the IR.

We then summarize the current measurements and potential biomarkers of IR to facilitate the clinical diagnosis. Finally, we provide the general approaches including lifestyle intervention, specific pharmacologic interventions and clinical trials to reduce IR.

Insulin is an endocrine peptide hormone with 51 amino acids and composed of an α and a β chain linked together as a dimer by two disulfide bridges 18 along with a third intrachain disulfide bridge in the α chain.

Accumulation of reports have demonstrated that IR is a complex metabolic disorder with integrated pathophysiology. The exact causes of IR has not been fully determined, 36 , 37 , 38 but ongoing research seeks to better understand how IR develops. Here, we focus on the underlying mechanism of IR including direct defective of insulin signaling, epidemiological factors, interorgan metabolic crosstalk, metabolic mediators, genetic mutation, epigenetic dysregulation, non-coding RNAs, and gut microbiota dysbiosis.

As has been mentioned, the proper modulators acting on different steps of the signaling pathway ensure appropriate biological responses to insulin in different tissues. Thus, the diverse defect in signal transduction contributes to IR.

Insulin exerts its biological effects by binding to its cell-surface receptors, therby activating specific adapter proteins, such us the insulin receptor substrate IRS proteins principally IRS1 and IRS2 , Src-homology 2 SH2 and protein-tyrosine phosphatase 1B PTP1B , eventually promoting downstream insulin signaling involving glucose homeostasis.

Most individuals that are obese or diabetic exhibit decreased surface INSR content and INSR kinase IRK activity. Second, decreased expression or serine phosphorylation of IRS proteins 44 , 45 can reduce their binding to PI3K, thereby down-regulating PI3K activation and inducing apparent IR.

It is generally accepted that diverse downstream targets of Akt activation lead to different distal signaling in target tissues response to insulin. Different investigations have indicated that premenopausal women exhibit many less metabolic disorders than men, including lower incidence of IR, although this effect diminishes severely when women reach the postmenopausal situation.

Concomitantly, clinical and experimental observations 70 , 71 have revealed that endogenous estrogens can protect against IR primarily through ER-α activation in multiple tissues, including in the brain, liver, skeletal muscle, and adipose tissue, in addition to pancreatic β cells.

Further, female hormone estrogens are determinants that mediate body adiposity levels and body fat distribution in addition to glucose metabolism and insulin sensitivity.

Specifically, insulin sensitivity and capacities for insulin responses in women is significantly higher than men. Male homozygous for the polymorphism of PPP1R3A gene that involved in glycogen synthase activity are significantly younger at diagnosis than female.

Thus, additional studies are required to understand mechanisms underlying sex differences and IR development. South Asian children exhibit greater IR compared with white European children, while girls are more insulin resistant than boys, with sex and ethnicity differences related to insulin sensitivity and body composition.

Despite the above objective factors, some modifiable lifestyle factors including diet, exercise, smoking, sleep and stress are also considered to contribute to IR.

Further, circadian clocks disruption might also be an important factor to IR development via various factors including clock gene mutations, disturbed sleep cycles, shift work and jet lag. Different investigations suggest that vitamin D supplementation might reduce IR in some people due to increasing insulin receptor genes transcription and anti-inflammatory properties, 95 while some researchers found that Vitamin D has no effect on IR.

Both experimental animals and clinical studies have shown that many hormones can induce IR including glucocorticoids GCs , 97 cortisol, 98 growth hormone, 99 and human placental lactogen, which may decrease the insulin-suppressive effects on glucose production and reduce the insulin-stimulated glucose uptake.

Several other clinical medications including anti-adrenergic such as salbutamol, salmeterol, and formoterol , HIV protease inhibitors, , atypical antipsychotics and some exogenous insulin that may improve IR because of the disordered insulin signaling. All together, there may have synergistic effects of different risk factors on insulin resistance, scientific researchers should cooperate with medical experts to reduce the chances of becoming insulin resistant.

As discribed above, insulin signaling calibrates glucose homeostasis by limiting hepatic glucose output via decreased gluconeogenesis and glycogenolysis activities.

These processes consequently increase the glucose uptake rates in muscle and adipose tissues. In addition, insulin profoundly affects lipid metabolism by increasing lipid synthesis in liver and fat cells Fig. Despite stimulated glucose uptake, insulin rapidly reduces hepatic glucose output and hepatic glucose production HGP by activating glycogen synthesis, and suppressing glycogenolysis and gluconeogenesis in liver.

Insulin induces SREBP-1c maturation via a proteolytic mechanism started in the endoplasmic reticulum ER , wherein hepatic IR is highly associated with hepatic steatosis.

Accordingly, restoration of nuclear SREBP-1c expression in liver-specific Chrebp defective mice normalized expression of some lipogenic genes, while not affecting glycolytic genes expressing. In contrast, ChREBP overexpression alone failed to promote the expression of lipogenic genes in the livers of mice lacking active SREBPs.

Together, these data demonstrate that SREBP-1c mediates the induction of insulin lipogenic genes, but that SREBP-1c and ChREBP are both necessary for harmonious induction of glycolytic and lipogenic genes. Altogether, these above pathways and components can be used to clarify the popular pathophysiology of hepatic IR.

The lipid metabolisms including increased de novo lipogenesis and attenuation of lipolysis in the adipose tissue largely coordinate with glucose homeostasis response to insulin stimulation. De novo lipogenesis regulation in adipose is similar to that in livers, wherein adipose-ChREBP is a major determinant of adipose tissue fatty acid production and systemic insulin sensitivity, that is induced by GLUT4-mediated glucose uptake, and genetically ablating ChREBP impairs insulin sensitivity in adipose tissue In addition, lipogenic gene FASN and DGAT mRNA expression in adipose tissue have been shown to correlate strongly and positively with insulin sensitivity, which were may reduced by larger adipocytes in adipose tissue of obese individuals.

The lipogenesis stimulation of insulin is also reduced in larger, more insulin-resistant cells. Insulin suppression of lipolysis includes the hydrolytic cleavage of triglycerides, resulting in the generation of fatty acids and glycerol. The best understood effectors for this process are PDE3B and ABHD15 that operated by the suppression of cAMP to attenuate pro-lipolytic PKA signaling toward adipose triglyceride lipase ATGL , hormone-sensitive lipase HSL , and perilipin PLIN.

Further, inhibition of PDE3B inhibits insulin-induced glucose uptake and antilipolysis. Insulin stimulated protein synthesis is mediated by activation of the protein kinases Akt and mTOR specifically mTORC1 and mTORC2 in numerous insulin-responsive cell types, such as hepatocytes, adipocytes, and myocytes.

Inhibition of mTOR by rapamycin obviously impairs insulin-activated protein synthesis. Amino acids metabolic substrates enhance insulin sensitivity and responsiveness of the protein synthesis system by increasing mTOR activity and inhibiting protein degradation in liver, muscle, and heart tissues.

These processes, in turn, promote protein synthesis and antagonize protein degradation. Adiponectin is the most abundant protein secreted by adipose tissue and exhibits potent anti-inflammatory properties. Moreover, targeted disruption of AdipoR1 results in halted adiponectin-induced AMPK activation, increased endogenous glucose production and increased IR.

Similarly, AdipoR2 deletion results in decreased PPAR-α signaling pathway activity and IR. In addition, chemerin is a chemokine highly expressed in liver and white adipose tissue that regulates the expression of adipocyte genes involved in glucose and lipid homeostasis like IRS-1 tyrosine phosphorylation activity, GLUT4, fatty acid synthase and adiponectin.

Thus, chemerin may increase insulin sensitivity in adipose tissue. Leptin is a cytokine encoded by ob gene and produced by the adipocytes. In summary, adipose tissue is a central node for distinct adipokines and bioactive mediators in IR pathophysiology. Consequently, identifying the effects of new adipokines will help in the development of new therapeutic strategies for obesity-induced diseases.

The specific insulin actions in adipose tissue include activation of glucose uptake and triglyceride synthesis, suppression of triglyceride hydrolysis and free fatty acids FFA and glycerol release into the blood circulation. Once the adipose tissue expandability exceeded limit under overnutrition, excess lipids and toxic lipid metabolites FFA, diacylglycerol, ceramide accumulated in non-adipose tissues, thus leading to lipid-induced toxicity lipotoxicity and developed IR in liver and muscle.

This process would in turn induce increased intracellular citrate levels, thereby inhibiting glucosephosphate G6P accumulation. Increased G6P levels then result in decreased hexokinase activity, increased glucose accumulation, and reduced glucose uptake. Other studies have demonstrated the relevance of the glucose-fatty acid cycle to lipid-induced IR.

For example, lipid infusions combined with heparin can be used to activate lipoprotein lipase, thereby increasing plasma concentrations of fatty acids. Further, these infusions promote muscle lipid accumulation and effectively induce IR.

Consistent with the above studies, elevated plasma fatty acid concentrations can result in increased intracellular diacylglycerol DAG levels, leading to the activation of protein kinase C isoform PKC-θ and PKC-ε isoforms in skeletal muscles and liver respectively.

Since diacylglycerol acyltransferase 1 DGAT1 can increase the conversion of DAG into triacylglycerol TAG , DGAT1 overexpression could decrease DAG levels and improve insulin sensitivity partially attenuating the fat-induced activation of DAG-responsive PKCs.

Taken together, these studies strongly support that DAG as a key intermediate of TAG synthesis from fatty acids has central modulation and potential therapeutic values in IR. Ceramide is another specific lipid metabolite that increases in concentration, along with DAG, in association with IR in obese mice.

Thus, ectopic lipid metabolite concentrations e. Consequently, concerted efforts to decrease lipid components in these organs are the most efficacious therapeutic targets for treating IR and metabolic diseases.

Some human genetic studies indicated that different genomic loci were associated with fasting insulin levels, higher triglyceride and lower HDL cholesterol levels, , which are different hallmarks of IR. The peroxisome proliferator-activated receptor gamma PPARγ variant Pro12Ala was one of the first genetic variants identified that is involved in fatty acid and energy metabolism and that is associated with a low risk of developing T2DM.

Nevertheless, additional studies are needed to assess the functional relationships between the genetic variants and IR, that are also influenced by various lifestyle and environmental factors. Recent studies have suggested that epigenetic modifications such as DNA methylation DNAm and histone post-translational modifications PTM are implicated in the development of systemic IR.

Global and site-specific DNA methylation is generally mediated by DNA methyltransferases DNMTs. These processes mainly occur in the context of CG dinucleotides CpGs and promoter region, while also involving covalent addition or removal of methyl groups as a means to repress or stimulate transcription, respectively.

For example, increased INS promoter methylation levels and INS mRNA suppression were observed under over-nutrition conditions and obese T2DM patients.

Another study demonstrated that increased IGFBP2 DNA methylation levels were are associated with lower mRNA expression levels in Visceral Adipose tissue VAT of abdominal obesity.

Moreover, the first global genome-wide epigenetic analysis in VAT from IR and insulin-sensitive IS morbidly obese patients identified a novel IR-related gene, the zinc finger protein ZNF exhibited the highest DNA methylation difference, and its methylation levels is lower in IR patient than in IS patient, consistent with increased transcription levels, such studies provide potential epigenetic biomarkers related to IR in addition to novel treatment targets for the prevention and treatment of metabolic disorders.

For example, peroxisome proliferator-activated receptor-α and -γ PPAR-α and PPAR-γ, respectively are encoded by PPARA and PPARG , respectively, and they are the two primary nuclear peroxisome proliferator-activated receptors involved in lipid metabolism. Higher PPARA and PPARG methylation levels were observed in association with obesity, consistent with decreased PPAR-α and PPAR-γ protein expression levels, that lead to dyslipidemia and IR.

SLC19A1, a gene encoding a membrane folate carrier, was reduced in obese WAT and induced global DNA hypermethylation of chemokine C-C motif chemokine ligand 2 CCL2 that is a key factor in WAT inflammation, resulting in increased CCL2 protein secretion and the development of IR in obese. In addition, several genes methylation involved in hypoxia stress and endoplasmic reticulum stress were regulated in obesity related metabolic diseases.

Recent epigenetic genome-wide analysis identified low HIF3A methylation levels upregulates HIF3A expression in adipose tissue, thereby leading to adipose tissue dysfunction and adiposity.

Ramos-Lopez et al. Specifically, increased insulin concentrations and HOMA-IR index were accompanied by lower ERO1LB and NFE2L2 methylation levels. The histone modification effect on gene expression mainly includes histones methylation and acetylation. Histone methylation could either activate gene transcription H3K4, H3K36, and H3K79 or silence gene expression H3K9 and H3K27 , which depends on the modification site.

Histone acetylation increases the accessibility and gene expression of various transcription factors by reducing the positive charge and histone affinity for DNA.

Increasing evidence indicates , that IGFR, InsR, IRS1, Akt, GLUT4, and PPAR are more deacetylated in association with IR than in normal physiological conditions. In contrast, IRS2, FoxO, JNK, and AMPK are usually acetylated in association with IR.

Castellano-Castillo, D. Further, global proteomic analyses have revealed 15 histone modifications that are differentially abundant in hepatic IR.

MicroRNAs miRNAs are small ncRNAs nucleotides incorporated into Argonaute Ago protein to form miRISCs, which can inhibit the expression of partially or completely complementary target mRMAs. Several miRNAs are involved in β cell differentiation and mature β cell functioning.

For example, islet-specific miR overexpression represses glucose-stimulated insulin secretion GSIS and insulin gene transcription, that is then reversed upon miR inhibition. Thus, these markers may improve disease prediction and prevention in individuals at high risk for T2DM.

Furthermore, pdx1, neurogenin-3 ngn3 , and a transcriptional factor essential for insulin transcription MafA are essential transcription factors for β-cell differentiation.

Thus, miRa2 can directly modulate insulin expression through foxA2 and then pdx1. miR expression is induced by the cellular redox regulator thioredoxin-interacting protein TXNIP that then represses MafA, thereby inhibiting insulin production. Numerous studies suggest that miRNAs have pivotal roles in glucose and lipid metabolism.

miR was first reported to directly regulate GLUT4 expression in adipocytes. In addition, the anti-diabetic drug metformin can up-regulate miRp expression to suppress G6Pase and inhibit hepatic gluconeogenesis.

The balance of low-density lipoprotein LDL and high-density lipoprotein HDL molecules that are synthesized in hepatocytes is critical for lipid homeostasis.

Many miRNAs have been identified as critical regulators of HDL and LDL biogenesis. For example, miR, miR, miR, and miRa repress expression of the ATP-binding cassette transporter ABCA1 that mediates hepatic HDL generation.

In addition, miRc targets the gene encoding microsomal triglyceride transfer protein MTP that is required for the lipidation of newly synthesized APOB in the liver for LD lipoprotein production.

miRc overexpression reduces the assembly and secretion of these APOB-containing lipoproteins, resulting in decreased plasma LDL levels. miRa and miR repress LDLR expression and inhibition of these miRNAs results in enhanced LDLR expression and clearance of circulating LDL.

Further, miR and miRd target the LDLR chaperonin PCSK9 and IDOL in addition to the rate-limiting enzyme in cholesterol biosynthesis, HMGCR. Chronic inflammation in insulin-reactive tissues is one of the most important causes of IR and increasing evidence suggests that miRNAs has a pivotal role in the inflammatory process.

Obesity inhibited miR expression in adipose tissue macrophages ATMs , and miR was shown to target Delta-like-4 DLL4 , a Notch1 ligand is associated with ATM inflammation. Conversely, Wang et al.

discovered that miRp is significantly upregulated in Natural killer NK cells-derived exosomes from lean mice, which directly targets SKI family transcriptional corepressor 1 SKOR1 , subsequently downregulated the expression levels of pro-inflammatory cytokine factors including IL-1β, IL-6, and TNF-α levels and attenuated IR.

Therefore, it might be that metabolism-regulating miRNAs play a vital role in the dynamics of metabolic homeostasis. Long non-coding RNAs lncRNAs are non-coding transcripts more than nucleotides, and the subcellular localization of lncRNAs determines their function.

LncRNAs located in the nucleus could affect chromosomal biology or interact with transcription factors to regulate gene transcription; lncRNAs located in cytosol could modulate mRNA stability and translational efficiency by acting as sponges for miRNAs or direct pairing with mRNA.

Recent advances have shown that lncRNAs play crucial roles in the pathologys of IR and diabetes. Glucose and lipid metabolism disorders are the primary causes for the pathophysiological development of IR. The lncRNA SRA promotes insulin-stimulated glucose uptake by co-activating PPARγ, leading to increased phosphorylation of the downstream targets Akt and FOXO1 in adipocytes.

These processes are closely related to the genes PGC1a and CPT1b that reverse FFA-induced lipid accumulation and improve IR. In addition the insulin target tissues, transcriptome profiling and different studies have identified several β-cell specific lncRNAs that contribute to obesity-mediated β-cell dysfunction and apoptosis.

LncRNA MALAT1 downregulation may lead to pancreatic β-cell dysfunction and T2DM development by direct interaction and regulation of polypyrimidine bundle binding protein 1 PTBP1. Further, lncRNA-p overexpression can decrease the β cell apoptosis ratio and partially reverse the glucotoxicity effects on GSIS function.

Contrary to conventional linear RNA, circRNAs are noncoding RNAs that generated from precursor mRNAs by back-splicing circularization, which is derived from exonic circRNAs, intronic circRNAs, exonic-intronic circRNAs and ntergenic circRNAs.

Recent studies have suggested that newly identified circRNAs are novel factors in the initiation and development of IR. CircHIPK3 is one of the most abundant circRNAs in β-cells and regulates hyperglycemia and IR by sequestering miRp and miRp, thereby increasing mRNA expression of key β-cell genes e.

Similar to the miRNAs and lncRNAs, several circRNAs also contribute to the the regulation of glucose and lipid homeostasis. Deep sequencing analysis of adipose circRNA revealed that circArhgap is highly upregulated during differentiation of human white adipocytes.

Thus, circRNAs likely serve as important regulators of adipocyte differentiation and lipid metabolism. Another circRNA deep sequencing analysis of sera from patients with metabolic syndrome MetS identified the presence of a novel circRNA, circRNF, involved in MetS progression.

AMPK is a critical factor in energy homeostasis including glycolysis, lipolysis, and fatty acid oxidation FAO. CircACC1 is a circRNA derived from the human acetyl-CoA carboxylase 1 ACC1 gene and directly binds to the β and γ subunits of AMPK, facilitating its activity, and promoting glycolysis and fatty acid β-oxidation during metabolic stress.

circMAP3K4 is another potentially important circRNA involved in glucose metabolism that is highly expressed in the placentas of patients with gestational diabetes mellitus GDM and the IR model.

Nevertheless, the exact roles and regulatory mechanisms of circRNAs in IR require additional clarity. The microbes living in the human gut are key contributors to host metabolism and immune function through mediating the interaction between the host and environment, or releasing metabolites and cytokines.

Different factors influencing these alterations of gut microbiome composition have been explored including diet, exercise, circadian disruption, antibiotics treatments, and genetics. The gut microbial communities of the groups significantly diverged over time, with participants on animal diets experiencing proliferation of bile-tolerant microorganisms e.

For example, microbiome genome-wide association studies mGWAS have identified that variants of different genes for example, VDR , LCT , NOD2 , FUT2 , and APOA5 that are associated with distinct gut microbiome compositions. Growing evidence in the last two decades has suggested that gut microbial dysbiosis contributes to increased risks of metabolic defects like obesity, IR, and diabetes.

LPS circulation then contributes to the chronic inflammation of liver and adipose tissue that is associated with the development of IR, in addition to other conditions associated with metabolic syndromes. As we all know, IR is a state in which higher than normal concentrations of insulin are needed for a normal response, leading directly to hyperinsulinaemia and impaired glucose tolerance.

Non-alcoholic fatty liver disease NAFLD is one of the most common liver diseases worldwide. Adipose tissue is a physiologic reservoir of fatty acids, when the storage capacity is exceeded, the accumulation of heterotopic lipids leads to lipotoxicity, thereby promoting low-grade inflammation and IR in the liver.

Lipotoxic injury appears to occur in response to excessive levels of serum free fatty acids FFAs in hepatocytes. At present, the molecular mechanism of insulin in PCOS has been well described. Such modifications then activate NF-κB that is involved in the expression of proinflammatory mediators such as TNF and IL-6, , and that induces key steroidogenic molecules, like CYP11A1, CYP17A1 and StAR, leading to further aggravation of hyperandrogenemia.

Cardiovascular diseases CVDs are the leading causes of death globally. The World Health Organization estimates that Moreover, over 23 million people are estimated to die from CVDs each year by However, the most common types of CVDs include high blood pressure, coronary artery disease CAD , stroke, cerebrovascular disease and rheumatic heart disease RHD.

Identifying new therapies to reduce IR may contribute to the reduced prevalence of CVDs. Insulin primarily enters the brain via selective, saturable transport across the blood-brain barrier BBB , , Peripherally produced insulin can also be actively transported into the brain via an endocytic-exocytic mechanism.

Current researches have demonstrated that the mechanisms of systemic IR and brain-specific IR have close links with AD pathogenesis. Pioglitazone acts similarly as Rosiglitazone by reducing tau and Aβ deposits in the hippocampus, and improving neuronal plasticity and learning in AD.

Moreover, overlapping pathological features exist for diabetes, IR, and AD. Chronic kidney disease CKD involves a gradual loss of kidney function and inability to filter blood , and is a major risk factor for end-stage kidney failure ESKF and CVDs.

Numerous recent epidemiological studies have suggested that IR increases the risks for different cancers including colon, liver, pancreas, breast, endometrium, thyroid and gastric cancer. Further, a growing body of evidence suggests that increased insulin, in addition to IGF1 and IGF2 levels critically influence tumor initiation and progression in IR patients.

As we all know, IR is related to several metabolic abnormalities including obesity, glucose tolerance, dyslipidemia, type 2 diabetes and other metabolic syndrome.

Actually, IR precedes the occurrence of T2DM, so how to increase the accurate assessment of insulin sensitivity is very important to predict the risk and evaluate the management of impaired insulin sensitivity and metabolic syndrome in research and clinical practice. HOMA2 updated HOMA model which took account of variations in hepatic and peripheral glucose resistance , homeostatic Model Assessment for IR HOMA-IR , the oral glucose insulin sensitivity index OGSI , fasting Insulin FINS , and fasting plasma glucose FPG based on fasting glucose and insulin levels , , , , are widely utilized IR measurements in clinical research.

Other indices based on fasting insulin include the glucose to insulin ratio GIR , the quantitative insulin sensitivity check index QUICKI , , , triglycerides McAuley Index alone or in accordance with HDL cholesterol HDL-C , whole-body insulin sensitivity index WBISI , Matsuda Index to evaluate whole body physiological insulin sensitivity by the above methods.

Indeed, the early symptoms of IR in different individuals are not obvious, and the related symptoms are very complex, combining with screening indicators may provide more precise diagnosis for IR in the general population.

No medications exist currently that are specifically approved to treat IR, but IR management 91 , , is possible through lifestyle changes like dietary, increased exercise, and disease prevention in addition to alternative medications Fig. Among these treatments, lifestyle changes should be the main focus for IR treatment, with nutritional intervention to decrease calories, avoidance of carbohydrates, and focusing on aliments with low glycemic index including vegetables, fruits, whole-grain products, nuts, lean meats or beans to provide higher fiber, vitamins, healthy fats and protein are particularly helpful for people trying to improve insulin sensitivity.

Table 1. Metformin is a first-line medication and the most widely-prescribed insulin-sensitizing agent in T2DM and PCOS patients. For example, 1 Glucagon-like peptide 1 GLP1 is an intestinal hormone that can enhance insulin secretion in a glucose-dependent manner by activating the GLP-1 receptor GLP-1R that is highly expressed on islet β cells.

are now world-wide therapy of T2DM since and could improve insulin sensitivity. In clinical research, scientists and physicians have explored different strategies to prevent and treat diabetes mellitus and IR.

gov to reduce IR and summarized them mainly include: 1 Diet intervention, such as Low-fat vegetarian Food, high-protein food, calorie restriction, vitamin D supplementation to reduce the IR in human obesity.

We present some clinical trials of IR intervention in Table 2. Over the past years, our knowledge of the pathogenesis of IR and T2DM has improved, the development of new treatments of IR and metabolic syndrome have gained certain success, while the complexity of IR and the presence of multiple feedback loops make a challenge to the specific intervention.

In recent years, accumulating preclinical studies on the intervention of IR have been reported, which have important reference significance for the development of new drugs.

We present the related studies on IR reported in recent years in Table 3 , including animal models, treatment methods and results.

Pre-clinical IR intervention mainly includes drug intervention, probiotic therapy and exercise supplement. Drug therapy to improve IR is the main research direction at present.

Researchers found that Valdecoxib VAL can inhibit inflammation and endoplasmic reticulum ER stress through AMPK-regulated HSPB1 pathway, thus improving skeletal muscle IR under hyperlipidemia.

The researchers found that the mixed nasal administration of GLP-1 receptor agonist and L-form of peneracin can effectively alleviate the cognitive dysfunction of SAMP8 mice. Natividad et al. Regular exercise is an alternative intervention measure to maintain the blood sugar level in the normal range and reduce the risk factors.

Hsu and colleagues found that exercise combined with probiotics intervention can have a positive effect on blood sugar and increase insulin sensitivity in mice. The above results show that drug intervention, probiotic supplementation and intensive exercise can improve IR but more clinical data are still needed.

Overall, the increased incidence of IR and the key roles of IR plays in many diseases, urgently require a better understanding of IR pathogenesis in addition to how IR interacts with genetics and different environments.

A deeper understanding of IR can be achieved with a more systematic approach involving large-scale omics to study the molecular landscape is of major importance in addition to exploring new intervention strategies to prevent abnormal IR syndrome.

Banting, F. The internal secretion of the pancreas. Indian J. CAS PubMed Google Scholar. SANGER, F. The amino-acid sequence in the glycyl chain of insulin. The identification of lower peptides from partial hydrolysates. Biochem J. Article CAS PubMed PubMed Central Google Scholar.

The investigation of peptides from enzymic hydrolysates. Kung, Y. Total synthesis of crystalline bovine insulin. Goeddel, D. et al. Expression in Escherichia coli of chemically synthesized genes for human insulin.

USA 76 , — Vecchio, I. The discovery of insulin: an important milestone in the history of medicine. Front Endocrinol. Article Google Scholar. Cheatham, B. Insulin action and the insulin signaling network. Root, H. Insulin resistance and bronze diabetes.

Laakso, M. Insulin resistance and hyperglycaemia in cardiovascular disease development. Article CAS PubMed Google Scholar. Bugianesi, E. Insulin resistance in nonalcoholic fatty liver disease. Saklayen, M. The global epidemic of the metabolic syndrome.

Diamanti-Kandarakis, E. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications. Stenvers, D. Circadian clocks and insulin resistance.

Article PubMed CAS Google Scholar. Freeman AM, Pennings N. Insulin Resistance. In: StatPearls Internet. Treasure Island FL : StatPearls Publishing. PMID: American Diabetes Association. Prevention or delay of type 2 diabetes: standards of medical care in diabetes Diabetes Care 44 , S34—S39 Pharmacologic approaches to glycemic treatment: Standards of Medical Care in Diabetes Diabetes Care 44 , S—S Weiss, M.

Insulin biosynthesis, secretion, structure, and structure-activity relationships. In: Feingold KR, Anawalt B, Boyce A, et al. South Dartmouth MA : MDText. com, Inc. Sanger, F. Chemistry of insulin. Science , — Katsoyannis, P.

Synthesis of insulin. Lee, J. The insulin receptor: structure, function, and signaling. Pessin, J. Signaling pathways in insulin action: molecular targets of insulin resistance. Invest , — Haeusler, R. Biochemical and cellular properties of insulin receptor signalling. Cell Biol. White, M.

Mechanisms of insulin action. In Atlas of diabetes pp. Springer, Boston, MA Newgard, C. Organizing glucose disposal: emerging roles of the glycogen targeting subunits of protein phosphatase Diabetes 49 , — Beurel, E.

Glycogen synthase kinase-3 GSK3 : regulation, actions, and diseases. Article CAS Google Scholar. Dong, X. Inactivation of hepatic Foxo1 by insulin signaling is required for adaptive nutrient homeostasis and endocrine growth regulation.

Cell Metab. Puigserver, P. Insulin-regulated hepatic gluconeogenesis through FOXO1—PGC-1α interaction. Nature , — Vander Haar, E. Garami, A. Cell 11 , — Laplante, M. mTORC1 activates SREBP-1c and uncouples lipogenesis from gluconeogenesis. USA , — Han, Y. Post-translational regulation of lipogenesis via AMPK-dependent phosphorylation of insulin-induced gene.

Calejman, C. mTORC2-AKT signaling to ATP-citrate lyase drives brown adipogenesis and de novo lipogenesis. Xia, W. Loss of ABHD15 impairs the anti-lipolytic action of insulin by altering PDE3B stability and contributes to insulin resistance. Cell Rep.

James, D. The aetiology and molecular landscape of insulin resistance. Tam, C. Defining insulin resistance from hyperinsulinemic-euglycemic clamps. Diabetes care 35 , — Samuel, V. Mechanisms for insulin resistance: common threads and missing links.

Cell , — Ye, J. Mechanisms of insulin resistance in obesity. Front Med. Article PubMed PubMed Central Google Scholar. Yaribeygi, H.

Insulin resistance: Review of the underlying molecular mechanisms. Cell Physiol. De Meyts, P. The insulin receptor: a prototype for dimeric, allosteric membrane receptors? Trends Biochem Sci. Caro, J. Insulin receptor kinase in human skeletal muscle from obese subjects with and without noninsulin dependent diabetes.

Invest 79 , — Fröjdö, S. Alterations of insulin signaling in type 2 diabetes: a review of the current evidence from humans. Biochim Biophys. Acta , 83—92 Fisher, S.

Michael, M. Loss of insulin signaling in hepatocytes leads to severe insulin resistance and progressive hepatic dysfunction. Cell 6 , 87—97 Davis, R.

The c-Jun NH2-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser Article PubMed Google Scholar.

Carvalho-Filho, M. Diabetes 54 , — Taniguchi, C. Critical nodes in signalling pathways: insights into insulin action.

Brachmann, S. Phosphoinositide 3-kinase catalytic subunit deletion and regulatory subunit deletion have opposite effects on insulin sensitivity in mice. Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp70 S6 kinase, DNA synthesis, and glucose transporter translocation.

CAS PubMed PubMed Central Google Scholar. Czech, M. Signaling mechanisms that regulate glucose transport. Luo, J. Loss of class IA PI3K signaling in muscle leads to impaired muscle growth, insulin response, and hyperlipidemia.

Cong, L. Physiological role of Akt in insulin-stimulated translocation of GLUT4 in transfected rat adipose cells. Xia, J. Targeted induction of ceramide degradation leads to improved systemic metabolism and reduced hepatic steatosis.

Le Marchand-Brustel, Y. Insulin receptor tyrosine kinase is defective in skeletal muscle of insulin-resistant obese mice. Brozinick, J. Defective signaling through Akt-2 and-3 but not Akt-1 in insulin-resistant human skeletal muscle: potential role in insulin resistance.

It furthers the University's objective of excellence in research, scholarship, and education by publishing worldwide. Sign In or Create an Account. Endocrine Society Journals. Advanced Search. Search Menu.

Article Navigation. Close mobile search navigation Article Navigation. Volume Article Contents Methods and Materials. Journal Article. Diurnal Variation in Insulin Sensitivity of Glucose Metabolism Is Associated With Diurnal Variations in Whole-Body and Cellular Fatty Acid Metabolism in Metabolically Normal Women.

Jun Yoshino , Jun Yoshino. Oxford Academic. Paloma Almeda-Valdes. Bruce W. Adewole L. Shin-ichiro Imai. Bettina Mittendorfer. Samuel Klein. PDF Split View Views. Cite Cite Jun Yoshino, Paloma Almeda-Valdes, Bruce W. Select Format Select format. ris Mendeley, Papers, Zotero. enw EndNote. bibtex BibTex.

txt Medlars, RefWorks Download citation. Permissions Icon Permissions. Figure 1. Open in new tab Download slide. Figure 2. and P. contributed equally to this work. adipose tissue triglyceride lipase. glyceraldehydephosphate dehydrogenase. Google Scholar Crossref. Search ADS. Van Cauter. Google Scholar PubMed.

OpenURL Placeholder Text. Issue Section:. Download all slides. Supplementary data. jc - pdf file. Views 11, More metrics information. Total Views 11, Email alerts Article activity alert. Advance article alerts. New issue alert. Receive exclusive offers and updates from Oxford Academic.

More on this topic Soluble CD Is Associated With CD mRNA Expression in Adipose Tissue and With Insulin Sensitivity in Steady-State Condition but Not in Response to Calorie Restriction. Threshold for Effects of Vitamin D Deficiency on Glucose Metabolism in Obese Female African-American Adolescents.

Hepatic Insulin Resistance in Antipsychotic Naive Schizophrenic Patients: Stable Isotope Studies of Glucose Metabolism. Plasma Levels and Adipose Tissue Messenger Ribonucleic Acid Expression of Retinol-Binding Protein 4 Are Reduced during Calorie Restriction in Obese Subjects but Are Not Related to Diet-Induced Changes in Insulin Sensitivity.

Related articles in PubMed Chaenomeles sinensis Thouin Koehne fruit polyphenols alleviate high-fat diet-induced obesity and liver steatosis by improving lipid metabolism in mice.

Glucocorticoid-induced adrenal insufficiency after therapy with intravenous methylprednisolone in patients with moderate-to-severe and active Graves' orbitopathy: assessment with a low-dose corticotropin test.

Prospecting for rare earth element hyper accumulators in the Paris Herbarium using X-ray fluorescence spectroscopy reveals new distributional and taxon discoveries. Treatment Options for Tennis Elbow - An Umbrella Review.

Citing articles via Web of Science Latest Most Read Most Cited Magnesium Depletion Score and Metabolic Syndrome in US Adults: Analysis of NHANES Clinical and Histopathological Risk Factors for Radioactive Iodine Refractory Follicular and Oncocytic Thyroid Carcinoma.

Non-Operative, Active Surveillance of Larger Malignant and Suspicious Thyroid Nodules. Carotid Intima-Media Thickness in Surgically or Conservatively Managed Patients With Primary Hyperparathyroidism. Differential diagnosis of post pancreatitis diabetes mellitus based on pancreatic and gut hormone characteristics.

More from Oxford Academic. Clinical Medicine. Endocrinology and Diabetes. Medicine and Health. It has been shown that inhibition of PTP1B, a main negative regulator of insulin receptor signaling, can improve glucose homeostasis and insulin signaling In the insulin receptor signaling cascade, protein tyrosine kinase amplifies the insulin signaling response, and phosphatase is necessary to regulate the rate and duration of the reaction IR occurs in a variety of tissues, including skeletal muscle, liver, kidney and adipose tissue, and its mechanisms are specific.

Among the target organs of insulin, bone, as an endocrine organ, can regulate energy homeostasis by altering insulin sensitivity, dietary behavior, and adipocytes There seems to be a bilateral relationship between bone and IR that binds them together in a biological partnership Among them, skeletal muscle estrogen receptor α plays a crucial role in maintaining systemic glucose homeostasis and insulin sensitivity It has been repeatedly demonstrated that skeletal muscle tissue plays an important role in the maintenance of systemic glucose homeostasis and overall metabolic health.

In addition, the crosstalk between muscle factors and adipokines leads to negative feedback, which in turn aggravates muscle reduction obesity and IR In the kidney, the effector cells of insulin are podocytes in which nucleotide-binding oligomerization domain 2 NOD2 is highly expressed.

NOD2 is a major member of the NOD receptor family and is involved in the innate immune response. It induces podocyte IR by activating the inflammatory response In terms of hepatic IR, IRA, one of the isoforms of the insulin receptor, whose expression in the liver of mice on a high-fat diet increase hepatic glucose uptake, decrease lipid accumulation, and reduce or at least delay the development of fatty liver and NASH.

This suggests that a gene therapy approach to hepatic IRA expression could act as a facilitator of glucose uptake in IR states — Insulin acts by binding to the INSR and activating downstream signaling pathways which have been extensively studied.

Although where the defect occurs in the insulin signaling pathway remains a matter of doubt, many key insulin signaling pathway components have been identified. IR is caused by defects in one or more of these signaling components Environment, such as diet and exercise, and genetics, as well as the interaction between the two, play a major role in the development of IR and metabolic disease.

Exercise and dietary habits may directly or indirectly drive changes in the host internal microenvironment. Current research suggests that extracellular influences such as inflammation, hypoxic environments, lipotoxicity or immune abnormalities can trigger intracellular stress in key metabolic target tissues, which impairs the normal metabolic function of insulin in these cells thereby causing the progression of whole-body IR Obesity characterized by a chronic, low-grade inflammatory state is closely associated with IR.

The mechanisms of inflammation leading to IR mainly include inflammatory factors acting on the insulin signaling system to interfere with INSR signal transduction. TNF-α and IL-1β are additional macrophage-derived pro-inflammatory mediators that directly affect insulin sensitivity , TNF-α stimulates insulin-resistant adipose tissue through IRS protein interference by abnormal signals on phosphorylated serine residues of IRS1 In addition, TNF-α could affect insulin signaling through serine phosphorylation and kinase pathway defects 99 , CRP is another marker of inflammation associated with IR and metabolic diseases and is a widely used clinical biomarker.

CRP binds to leptin, blocks leptin signaling and modulates its central action and hypothalamic signaling, thereby directly interfering with energy homeostasis, insulin sensitivity and glucose homeostasis , The above pro-inflammatory cytokines exert their effects by stimulating major intracellular inflammatory pathways, and the activation of these pathways also promotes increased expression of the inflammatory factors involved in IR.

Toll-like receptor TLR , especially TLR4, participates in IR-related inflammation by increasing the gene expression of IKKβ, NF-κB transcription factors, and pro-inflammatory mediators in adipose tissue macrophages — IKK is an enzyme complex that activates the NF-κB transcription factor It has also been shown that NF-κB receptor activator RANKL is a potent stimulator of NF-κB and that systemic or hepatic blockade of RANKL signaling leads to significant improvements in hepatic insulin sensitivity and prevents the development of diabetes And JNK signaling in adipocytes leads to an increase in circulating concentrations of hepatic factor fibroblast growth factor 21 FGF21 , which regulates systemic metabolism In the pathogenesis of IR and metabolic diseases, immune cells play a crucial role.

Adipose tissue contains most types of immune cells, which under conditions of obesity contribute to a complex network of inflammation and IR with activation and infiltration of pro-inflammatory immune cells in adipose tissue, including macrophages, neutrophils, eosinophils, mast cells, NK cells, MAIT cells, CD4 T cells, CD8 T cells, regulatory T cells and B cells, as well as high levels of pro-inflammatory molecules Among them, adipose tissue macrophages can be divided into M1 phenotype pro-inflammatory macrophages and M2 phenotype anti-inflammatory macrophages , representing the two extremes of macrophage polarization.

M1 macrophages are highly antimicrobial and antigen-presenting, producing pro-inflammatory cytokines, such as TNF-α, and reactive oxygen species ROS that worsen inflammation, mast cells, neutrophils and dendritic cells directly or indirectly exacerbate IR In contrast, M2 macrophages help maintain insulin sensitivity in lean adipose tissue, as well as eosinophils and innate lymphocytes appear to have a protective effect on glucose homeostasis and insulin sensitivity — Crosstalk between M1-M2 macrophage polarization plays an important role in IR through the shift from M1 to M2 phenotype and activation of transcription factors , Dysregulation of visceral adipose tissue macrophage ATM response to microenvironmental changes underlies the development of abnormal local and systemic inflammation and IR In the obese state, enhanced macrophage infiltration and secretion of various inflammatory cytokines in white adipose tissue activate JNK and NF-κB, causing local and systemic IR , Macrophages can alter their phenotype in response to changes in the microenvironment and macrophage differentiation.

In the past, more attention has been paid to the regulation of insulin sensitivity by innate immune cells, particularly macrophage mediated, which have been mentioned before. Cells of the adaptive immune system, B lymphocytes and T lymphocytes, and their respective subsets, are also thought to be important regulators of glucose homeostasis and play an important role in the immunopathogenesis of autoimmune diabetes , , Impaired through an adaptive immune response, IR can also be driven by inflammation and dysregulation of the gut microbiota, as in pathogen-induced periodontitis In addition, the intestinal immune system is an important regulator of glucose homeostasis and obesity-related IR in turn affects intestinal permeability and thus systemic IR Another essential part of the immune defense system is the complement system.

It plays an important role in activating innate and adaptive immune responses, promoting apoptosis, and eliminating damaged endogenous cells.

Patients with obesity exhibit activation of the complement system in their adipose tissue, which is connected to changes in glucose metabolism and subclinical inflammation Adipose tissue hypoxia is causally related to obesity-induced IR, especially in high-fat diet HFD fed and early obese patients, as adipocyte respiration becomes uncoupled, resulting in a state of increased oxygen consumption and relative adipocyte hypoxia Clinically, obstructive sleep apnea OSA , characterized by intermittent hypoxia IH , is a widely prevalent respiratory disorder with a particularly high prevalence in obese patients and is associated with IR and metabolic diseases such as hypertension, cardiovascular risk and NAFLD , Not only in obese individuals, but an animal study found that IH cause acute IR in lean or healthy mice, which is related to reduced glucose utilization in oxidized muscle fibers.

As the glucose infusion rate decreased, hypoxia induced systemic IRA The key regulators of oxygen homeostasis in response to hypoxia are the hypoxia-inducible factors HIFs , a family of transcription factors activated by hypoxia.

Adipocyte hypoxia could trigger HIF-1α induction causing adipose tissue inflammation and IR , HIFmediated activation of NOX4 transcription and the consequent increase in H2O2 led to intermittent hypoxia-induced pancreatic β-cell dysfunction In hypoxic adipocytes, HIF-1α activates the NLRP3 inflammasome pathway and stimulates IR by upregulating the expression of pla2g In obesity-induced intestinal hypoxia, HIF-2α increases the production of ceramide, to promote the expression of the key enzyme sialidase 3 encoding Neu3, which leads to the development of IR in obese mice induced by a high-fat diet While in skeletal muscle, hypoxia is a stimulus stimulating GLUT4 translocation via activation of AMPK, causing defects of glucose transport and this may counteract IR Insulin regulates lipid metabolism through the typical insulin signaling cascade, while metabolites can also directly regulate insulin sensitivity by modulating components of the insulin signaling pathway Lipids have multiple roles as signaling molecules, metabolic substrates and cell membrane components, and can also alter proteins that affect insulin sensitivity Lipotoxicity is when the storage capacity of adipose tissue is overloaded due to obesity, overnutrition, etc.

High concentrations of lipids and lipid derivatives cause deleterious effects on cells through mechanisms including oxidative stress, endoplasmic reticulum ER stress, c-Jun NH2-terminal kinase JNK -induced toxicity, and BH3-pure protein-induced mitochondrial and lysosomal dysfunction , Numerous studies have reported that Adipose tissue dysfunction and lipotoxicity play a role in metabolic disorders and IR , This is associated with a chronic elevation of free fatty acids FFA, also called non-esterified fatty acids in plasma due to adipose tissue dysfunction Adipose malnutrition or adipose tissue dysfunction can lead to pathologically elevated FFAs.

Chronically elevated FFAs appear to cause adipocyte production of inflammatory factors, decreased insulin biosynthesis, glucose-stimulated insulin secretion, and glucose sensitivity in β-cells. The ER stress pathway is a key mediator of inflammation induced by serum excess FFA and IR in various cell types, and PERK and IKKβ are key signaling components The obesity-induced increase in adipocyte volume and tissue mass will lead to inflammation, additional disturbances in adipose tissue function, and ultimately adipose tissue fibrosis Adipose tissue macrophages are an abundant immune component of hypertrophy, which plays a key role in diet-induced T2DM and IR In renal ectopic lipid accumulation, lipotoxicity promotes podocyte injury, tubular injury, thylakoid proliferation, endothelial cell activation and macrophage-derived foam cell formation, which contribute to the development of renal IR and other renal diseases, especially diabetic nephropathy In skeletal muscle, sustained nutrient overload of L6 myotubes leads to lipotoxicity that promotes activation of the IKKβ-NFkB pathway in muscle cells, inducing increased cellular ROS and impaired insulin action in the myotubes Saturated fatty acids are known to increase the production of lipotoxic products such as ceramide and diacylglycerol, which disrupt islet beta-cell function, vascular reactivity and mitochondrial metabolism, and also play a key role in the induction of muscle IR — Similarly, defective fatty acid oxidation FAO and consequent lipotoxicity in cardiac cells induce a range of pathological responses, including oxidative stress, DNA damage, inflammation and insulin resistance.

The obesity-mediated atrial fibrillation and structural remodeling can be attenuated by promoting FAO, activating AMPK signaling and attenuating atrial lipotoxicity through levocarnitine LCA Lysophosphatidic acid LPA is an effective, biologically active lipid. After binding to G protein-coupled receptors, it can profoundly affect cell signal transduction and function.

Metabolic and inflammatory disorders, including obesity and IR, are associated with modifications in LPA signaling as well as the production and function of autocrine motility factors Additionally, it has been discovered that the anti-adipogenic transcription factor GATA-3 is a possible molecular target that affects adipogenesis.

Those with obesity and IR exhibit increased GATA-3 expression when compared to insulin-sensitive individuals with BMI matches While lifestyle interventions such as physical activity have been confirmed to have a positive effect on insulin sensitivity in skeletal muscle, affecting lipid metabolism Ceramides are a family of lipid molecules consisting of sphingosine and a fatty acid.

The synthesis of de novo ceramides depends on the availability of free fatty acids, especially palmitate, whose over-intake may lead to an excessive accumulation of ceramides In addition to their function in lipid bilayers, these molecules are also thought to be biologically active agents involved in a variety of intracellular pathways, such as free radical production, release of inflammatory cytokines, apoptotic processes, and regulation of gene expression.

Ceramides are metabolic products that accumulate in individuals suffering from obesity or dyslipidemia and alter cellular processes in response to fuel overload ceramides accumulation over time modulates signaling and metabolic pathways that drive lipotoxicity and IR, causing tissue dysfunction Numerous studies have been conducted in recent years to confirm the critical role played by ceramides in glucose homeostasis and insulin signaling These evidence are particularly strong in skeletal muscle, while the data in liver and WA are somewhat more equivocal , Ceramides are synthesized by ceramide synthase CerS through N-acylation.

To date, six mammalian CerS have been identified CerS that show different affinities for the fatty acid acyl-CoA chain length used for sphingomyelin N-acylation. CerS6 is specific for C14 and C16 acyl chain lengths, and CerS6 levels are significantly increased in obese adipose tissue , In addition, ceramide may cause IR by accumulating in mitochondria and causing mitochondrial reactive oxygen species ROS or by promoting the secretion of pro-inflammatory factors Another lipid metabolite closely associated with IR is DAG, whose accumulation in skeletal muscle, adipocytes and liver is thought to promote IR by altering cellular signaling at its specific location, due to increased serum FFA levels The DAG hypothesis of IR is that the interference of activated PKC, especially the novel PKC isoforms including δ, ϵ, ν, and θ, with insulin signaling is due to the accumulation of DAG in insulin-sensitive tissues , In particular, 1,2-DAG, which derives from esterification and accumulates mainly in the membranes, is clearly associated with PKC activation, and these isoforms then phosphorylate IRS1 serine with the result that decrease PI3K activation , It is worth noting that the role of intracellular ceramide and DAG in IR is controversial and that defects in these components are unlikely to be the sole cause of IR.

It is true that not all studies have confirmed a role for the DAG-PKC-insulin receptor pathway in IR; for example, some studies have shown that PKCϵ deficiency in the liver has no effect on systemic insulin sensitivity in mice , and there are also experiments in which acute knockout of PKCϵ in the liver protects rats from IR Therefore, more in-depth studies on proximal insulin signaling with DAG and ceramide are still needed.

Organelles, including the endoplasmic reticulum ER , mitochondria and endoplasmata, contribute to a range of cellular functions through their unique local environment and molecular composition. Organelles can actively communicate and cooperate with each other through vesicle trafficking pathways and membrane contact points MCSs to maintain cellular homeostasis, which facilitates the exchange of metabolites and other information required for normal cellular physiology Imbalances in organelle interactions may lead to various pathological processes, such as imbalances in cellular energy metabolism Recent studies have shown that mitochondria could interact with various organelles , which are essential for energy metabolism and cell survival, and increasing evidence shows that mitochondrial dysfunction in skeletal muscle and mitochondrial overactivation may induce IR The production of mitochondrial ROS is thought to adjust skeletal muscle insulin sensitivity.

Mitochondrial quality control mechanisms are regulated by PGC-1α, which may affect age-related mitochondrial dysfunction and insulin sensitivity The continuous processes that occur in the skeletal muscle after excessive intake of a high-fat diet include the accumulation of cytosolic fatty acids, increased production of ROS, mutation, and aging.

The ensuing mitochondrial dysfunction could lead to decreased β-oxidation, respiratory function, and increased glycolipid toxicity. Together, these events induce IR in the skeletal muscle The physical contact site between the mitochondria and endoplasmic reticulum ER is called the mitochondrial-associated membrane MAM.

The imbalance of MAMs significantly leads to IR. ER stress may be the main mechanism by which MAM induces IR in the brain, especially in the hypothalamus , Exosome-like vesicles ELVs are the smallest type of extracellular vesicles released from cells that play a role in cell crosstalk because they regulate insulin signaling and β-cell quality, and released ELVs leading to IR or β-cell apoptosis PTEN is not only a tumor suppressor gene but also a metabolic regulator.

Under physiological and T2D conditions, PTEN also has a negative regulatory function in insulin signaling through its inhibition in the PI3K pathway , PTEN reduces the level of phosphatidylinositol-3, 4, 5-phosphate PIP3. This leads to impaired insulin signaling and promotion of IR in the pathogenesis of T2D.

The function of PTEN in regulating insulin signaling in different organs has been identified. The role of PTEN in the regulation of insulin action in many cell types has been elucidated through mouse models of lacking PTEN in metabolic organs and in vitro cell culture , Interventions targeting PTEN regulatory signaling may therefore be a promising target aimed at reversing insulin resistance.

In addition to its effects on skeleton, Vit D has significant effects on pancreatic β-cells function and metabolic syndrome including blood pressure, abdominal obesity, glucose metabolism associated with it, as calcitriol functions as a chemical messenger by interacting with calcium flux-regulating receptors on beta cells As the results of a meta-analysis showed, there was an inverse relationship between serum Vit D concentration and metabolic syndrome risk in the general adult population in cross-sectional studies Vitro studies showed that Vit D could regulate lipid and glucose metabolism in adipose tissue, skeletal muscle and liver, and pancreatic insulin secretion Minerals are essential micronutrients for the human body.

Deficiencies in certain micronutrients due to differences in diet composition may lead to imbalances in glucose homeostasis and IR Magnesium is a cofactor required for glucose access to cells and carbohydrate metabolism, and it has the function of regulating the electrical activity of pancreatic beta cells and insulin secretion Mechanistically explained, magnesium is a cofactor in the downstream action of the insulin cascade.

Low magnesium ion levels lead to defective tyrosine kinase activity, blocking intracellular insulin action and altered cellular glucose transport, thus promoting IR On the other hand, magnesium deficiency inhibits cellular defenses against oxidative damage and triggers chronic systemic inflammation that enhances IR.

As demonstrated in a longitudinal study, magnesium intake was also inversely associated with high-sensitivity CRP, IL-6 and fibrinogen levels, as well as HOMA-IR There is evidence suggesting that magnesium supplementation attenuates IR in patients with hypomagnesemia-associated IR Also, animal studies have shown that dietary magnesium supplementation to increase plasma magnesium concentrations reduces blood glucose levels, improves mitochondrial function, and reduces oxidative stress in diabetic mice However, new intervention studies are still needed to clarify the role of nutrients in the prevention of this metabolic disorder, as well as to standardize the type, dose, and timing of magnesium supplementation.

Zinc is an essential micronutrient for metabolism, which plays a particularly critical role in the islets. Diabetes affects zinc homeostasis, and disturbances in zinc homeostasis have been associated with diabetes and IR Because zinc is an essential component of insulin, it regulates islet cell secretion and promotes its binding to hepatocyte membranes while maintaining phosphorylation and dephosphorylation levels of the receptor.

Zinc influx mediated by Slc39a5, a zinc exporter in pancreatic β-cells, plays a role in insulin processing and secretion by inducing Glut2 expression through Sirt1-mediated activation of Pgc-1α In addition, zinc acts as a pro-antioxidant to reduce the formation of ROS, which is particularly beneficial in aging and IR Mineral deficiencies are directly or indirectly associated with oxidative stress, which ultimately leads to IR or diabetes The brain is also an insulin-sensitive organ with a large number of insulin receptors distributed , The action of insulin in the brain produces a variety of behavioral and metabolic effects that influence eating behavior, peripheral metabolism, and cognitive performance Disturbances in the role of insulin in the brain reveal a possible link between metabolism and cognitive health.

The hypothalamus plays a fundamental role in the survival and control of physiological processes necessary for vital physical functions, including various endocrine functions. Injecting insulin via intranasal administration leads to an increase and subsequent decrease in plasma insulin, affecting peripheral metabolism, and a decrease in BOLD signaling and cerebral blood flow in the hypothalamus is observed , It appears that the effects of central insulin may have a biphasic effect on peripheral insulin sensitivity Insulin signaling has been shown to affect the molecular cascade of hippocampal plasticity, learning, and memory Furthermore, the insulin-responsive glucose transporter GluT4 has a key part in hippocampal memory processes, and reduced activation of this transporter may underlie IR-induced cognitive deficits Autophagy is a self-degrading process that is conserved in all eukaryotic cells and plays a crucial role in balancing energy sources during critical periods of development and in response to nutritional stress.

Autophagy also promotes cellular senescence and cell surface antigen presentation, prevents genomic instability and necrosis, and it is an important mechanism for a variety of physiological processes, such as cellular homeostasis, senescence, immunity, oxidation, differentiation, and cell death and survival Recent studies have shown that autophagy is an important regulator of organelle function and insulin signaling, and that loss of autophagy is a key component of defective insulin action in obesity, which may be specifically related to ER function It has been found that autophagy deficiency and its resulting mitochondrial dysfunction increase fibroblast growth factor 21 Fgf21 expression through the induction of Atf4.

The induction of Fgf21 promotes protection against diet-induced obesity and IR In addition, exercise induces autophagy through the regulator BCL2, which may contribute to beneficial metabolic effects and improve IR in muscle In addition to the aforementioned influences such as metabolites and cytokines, the trillion bacterial colonized gut microbiota can also contribute to IR , Patients with metabolic syndrome showed increased insulin sensitivity after six weeks of infusion of gut microbiota from lean individuals.

Levels of gut microbiota producing butyrate, which has been shown to prevent and treat diet-induced insulin resistance in mice by promoting energy expenditure and inducing mitochondrial function, were also increased , Dietary reasons for obesity may promote IR both through mechanisms independent of the gut microbiota and through mechanisms dependent on the bacterial community Intestinal dysbiosis is associated with the transfer of bacterial lipopolysaccharide LPS into the systemic circulation and its induction of metabolic endotoxemia, leading to a chronic subclinical inflammatory process and the development of IR through activation of toll-like receptor 4 TLR4 — In addition to the LPA mentioned above, branched-chain amino acids BCAAs are another harmful gut microbially regulated metabolite whose levels are increased in the serum metabolome of IR individuals.

Prevotella copri has been shown in mice experiments to induce IR, exacerbate glucose intolerance and increase circulating levels of BCAAs Moreover, gut microbiota-derived short-chain fatty acids SCFA may improve IR and prevent T2DM by reducing the secretion of pro-inflammatory cytokines and chemokines and decreasing local macrophage infiltration, as well as increasing the lipid storage capacity of white adipose tissue , , Taken together, targeting gut microbes may have the potential to reduce IR and decrease the incidence of related metabolic diseases.

This lifestyle triggers several mechanisms such as the development of IR that aggravate metabolic stress. Next, the contribution of non-pharmacological therapies, including exercise and diet, to the alleviation of IR will be elaborated.

Exercise is well known to improve metabolic disease by improving obesity and enhancing insulin sensitivity. A meta-analysis determined the effectiveness of a structured exercise intervention program for IR in T2DM, and the evidence highlights that regular exercise improves glycemic control and therefore can be recommended for reducing IR with a moderate level of evidence As we know, physical exercise increases the oxidative capacity and biogenesis of mitochondrial substrates in skeletal muscle.

It was shown that treadmill training modulates the increase in mitochondrial substrate oxidation in liver and skeletal muscle induced by a high-energy diet in mice, disconnecting it from pyruvate and acetyl CoA-driven lipid synthesis.

This may help prevent the long-term deleterious effects of excessive nutritional intake on liver mitochondrial function and insulin sensitivity, thereby preventing the development of metabolic diseases such as fatty liver and NAFLD As described in the mechanism section, intermittent hypoxia leads to disturbances in the gut microbiota-circulating exosome pathway, disrupting adipocyte homeostasis and leading to metabolic dysfunction manifested as IR, whereas experiments have shown that such changes can be attenuated by physical activity, as regular non-strenuous activity will lead to substantial improvements in the gut microbiota-exosome pathway In addition, available data suggest that aerobic exercise can lead to increased insulin sensitivity and enhanced glucose metabolism through a variety of different molecular mechanisms, including upregulation of insulin transporters on cell membranes of insulin-dependent cells, reduction of adipokines, normalization of redox status, improvement of β-cell function, regulation of IRS-1 phosphorylation, reduction of ceramide plasma levels, and induction of angiogenesis, which may lead to a reduced incidence of diabetic complications, as well as other metabolic effects , Other forms of exercise, such as yoga, have also been shown to improve IR.

Several meta-analyses have shown that yoga is a safe and effective intervention to reduce waist circumference and systolic blood pressure in patients with metabolic syndrome, particularly in improving cardio-metabolic health , Some traditional Chinese health exercises, such as qigong and tai chi, have also been shown to have a measurable effect on weight, waist circumference, leg strength, increase HDL cholesterol, and result in significant improvements in IR , As mentioned above, high-fat diets and the obesity they induce are a major cause of IR.

Conversely, weight loss, when necessary, and dietary interventions such as intermittent fasting programs that reduce carbohydrates in the diet can significantly improve glycemic and insulin responses. The Mediterranean diet is characterized by a wide range of cardio-protective nutrients, with beneficial effects on several outcomes related to metabolic health, and significant beneficial changes in metabolic risk factors, including HOMA-IR index — There are also RCT studies reporting that a high-protein diet is more effective in controlling IR and glycemic variability compared to a Mediterranean diet, which may be related to the satiety and increased metabolic rate associated with a high-protein, low-sugar diet In terms of dietary composition, a key dietary strategy for treating IR and improving glycemic control is to consume foods and meals that reduce the glucose fluctuations known to induce oxidative stress and beta cell damage The contribution of high-fat diets to obesity and IR is well known.

However, a single-minded approach to weight loss by replacing fat intake with carbohydrates is counterproductive because it could exacerbate IR. Researchers suggest that calorie restriction for weight loss and rationing of the macronutrient composition of the diet is important.

The possible mechanism for this is that calcium and vitamin D in supplemental dairy products may facilitate lipolysis and optimize glucose metabolism Carbohydrates are the main macro-nutrient influencing the glycemic response, especially after a meal.

In recent years, some researchers have proposed that consumption of carbohydrates rich in dietary fiber and low glycemic index, such as whole grains, is beneficial in improving insulin sensitivity and metabolic flexibility, independent of gut hormones , A recent meta-analysis reported that increasing daily fiber intake by 15 or 35 grams compared to a low-fiber diet reduced homeostatic model assessment of insulin resistance HOMA-IR , leading to improvements in glycemic control, lipids, weight, and inflammation, as well as a reduction in premature mortality Not only is the amount of carbohydrate intake important, but the timing of major carbohydrate intake during the day is also a determining factor in the increase in glucose and insulin after meals and the improvement or otherwise of IR The results of some randomized controlled trial RCT studies suggest that it is advisable to consume at least half of the carbohydrates at lunch and to avoid consuming large amounts of carbohydrates at breakfast or dinner in order to control blood glucose spikes, which may be related to diurnal variations in insulin sensitivity — Results of another study showed that 10 hours of restrictive eating improved quality of life by reducing body weight and improving blood glucose, insulin sensitivity and related metabolic disorders Other dietary strategies have been shown to prevent high-fat diet-induced IR, such as the intake of flavonoid-rich natural products, like flavonoids, which upregulate the expression of related genes through cell surface G protein-coupled estrogen receptors Although lifestyle modification and weight loss are highly recommended to improve IR and its associated metabolic disorders, they have limited effectiveness, slow onset of action, and low feasibility.

Pharmacological treatments to increase insulin sensitivity will be described next. Currently, the main drugs that can effectively improve IR are anti-hyperglycemic drugs, including metformin, thiazolidinediones TZD , sodium glucose cotransporter SGLT -2 inhibitors SGLT2i , etc.

Metformin, the most commonly used insulin-sensitizing agent, has been a guideline-recommended first-line treatment for T2DM for decades and has recently found new applications in the prevention and treatment of various diseases, including metabolic disorders and cardiovascular diseases Metformin improves IR by modulating metabolic mechanisms and mitochondrial biogenesis through altering microRNAs levels by AMPK-dependent or AMPK-independent mechanisms TZDs, such as pioglitazone, are potent insulin sensitizers targeting PPARγ and PI3K, regulating the transcription of nuclear transcription factors, stimulating mainly white adipose tissue remodeling, and regulating lipid flux for insulin sensitization and beta cell protection , SGLT2i is a relatively new class of glucose-lowering drug that not only lowers blood glucose by inhibiting renal glucose reuptake, leading to increased urinary glucose excretion and lower blood glucose, but also improves insulin sensitivity in patients with T2DM by reducing body weight or glucose toxicity , And in a randomized, double-blind, placebo-controlled clinical trial, it was shown that 8 weeks of treatment with SGLT2i empagliflozin restored insulin sensitivity in the hypothalamus of patients with prediabetes Glucose-lowering drugs have also shown good, stacked effects in patients who do not have good response with one drug alone.

For example, the addition of rosiglitazone to metformin can be clinically important in improving glycemic control, insulin sensitivity and beta-cell function The addition of sitagliptin or metformin to pioglitazone monotherapy also leads to faster and better improvement in IR and inflammatory status parameters Other therapies, as well as some new drugs in clinical trials, such as anti-inflammatory drugs, drugs that target hepatic lipid and energy metabolism, renin-angiotensin-aldosterone system blockers, vitamin D, antioxidants, probiotics and fecal transplants, have also shown improvement in IR Among them, selected clinical trials in the last decade have been listed in Table 2.

As mentioned previously, low-grade chronic inflammation is associated with IR and metabolic disturbances. For example, in in vitro and in vivo mouse models of diet-induced hyperinsulinemia, low-dose naltrexone attenuates hyperinsulinemia-induced proinflammatory cytokine release and restores insulin sensitivity However, it is worth noting that corticosteroids can cause IR and hyperglycemia due to their metabolic effects, and statins also increase the risk of IR, although they can reduce circulating inflammatory markers TCM plays an equally critical role in the treatment of many acute and chronic diseases, especially its adeptness in restoring the dynamic balance of the body in systemic diseases.

Its main therapeutic measures include herbal medicine, acupuncture and Tui Na. Several classical herbal formulations have been widely used in the clinical treatment of T2DM and various other metabolic disorders. For example, GegenQinlian decoction improves IR in fat, liver and muscle tissue through a variety of compounds, targets, pathways and mechanisms Yi Qi Zeng Min Tang has been shown to improve IR in high-fat fed Sprague-Dawley rats without increasing body weight Because it reduced the expression of PI3K p85 mRNA and IRS1 protein, Fu Fang Zhen Zhu Tiao Zhi formula similarly improved IR in vitro and in rats with metabolic syndrome Gui Zhi Fu Ling Wan, Dingkun Pill and Liuwei Dihuang Pills are herbal formulas widely used in the treatment of gynecological disorders and have the effect of harmonizing Qi and blood or dispelling blood stasis in Chinese medical theory.

In addition, the efficacy of acupuncture in improving IR is equally impressive, as a recent meta-analysis showed that acupuncture improved HOMA-IR and ISI as well as fasting blood glucose FBG , 2h postprandial blood glucose 2hPG and fasting insulin FINS levels, with fewer adverse events The increased incidence of IR and its vital role as a major and common cause of numerous metabolic diseases have created an urgent need to gain insight into the etiology and pathogenesis of IR, as well as to explore better early diagnostic methods and therapeutic strategies for it.

The diagnosis of insulin resistance is currently inconclusive, while it is important to detect IR early and predict individual response to treatment. In addition to the few simple indices of IR calculated from biochemical or anthropometric variables currently in use, emerging biomarkers may now be the way forward, but this still needs to be supported by clinical data.

Different ranges and criteria are also needed for the diagnosis and monitoring of different metabolic diseases. As mentioned above, IR is a central mechanism in many metabolic diseases. Since this is the case, IR should be considered as a therapeutic target for patients with a combination of multiple metabolic diseases so that multiple diseases can be treated simultaneously with the same treatment approach, thereby reducing healthcare expenditures.

Although there is no universally accepted theory to explain the mechanisms that cause IR. Nevertheless, there is growing evidence linking ectopic lipid accumulation, ER stress, plasma concentration of inflammatory cytokines, oxidative stress, abnormalities in insulin signaling, and other factors to IR.

In recent years, the exploration of the molecular mechanisms of IR has also led to the emergence of new therapeutic concepts beyond metformin and TZD.

Regardless lifestyle modification remains the most basic and least costly intervention. Normative criteria need to be developed for different metabolic diseases considering IR as a focus. FL and HJ provided the idea of the manuscript. XZ, XA, and CY contributed equally to this manuscript.

XZ, XA, WS, and CY drafted the manuscript and searched the relevant literature. XZ and XA drafted the figures, and all authors approved the final version of the manuscript. All authors agree to be accountable for all aspects of work ensuring integrity and accuracy.

All authors contributed to the article and approved the submitted version. This work was supported by Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine. No: ZYYCXTD-D The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

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

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Bugianesi E, McCullough AJ, Marchesini G. Insulin resistance: a metabolic pathway to chronic liver disease.

Hepatology 42 5 — doi: PubMed Abstract CrossRef Full Text Google Scholar. Sharma VR, Matta ST, Haymond MW, Chung ST. Measuring insulin resistance in humans. Horm Res Paediatr 93 — Muniyappa R, Madan R, Varghese RT.

Assessing insulin sensitivity and resistance in humans. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, editors. South Dartmouth MA: MDText.

com; Inc. Copyright © ; MDText. Google Scholar. Gar C, Rottenkolber M, Prehn C, Adamski J, Seissler J, Lechner A. Serum and plasma amino acids as markers of prediabetes; insulin resistance; and incident diabetes. Crit Rev Clin Lab Sci 55 1 — Park SE, Park CY, Sweeney G.

Biomarkers of insulin sensitivity and insulin resistance: Past; present and future. Crit Rev Clin Lab Sci 52 4 — Milburn MV, Lawton KA.

Application of metabolomics to diagnosis of insulin resistance. Annu Rev Med — Yang R, Hu Y, Lee CH, Liu Y, Diaz-Canestro C, Fong CHY, et al. PM20D1 is a circulating biomarker closely associated with obesity; insulin resistance and metabolic syndrome.

Eur J Endocrinol 2 — Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep 20 2 Chooi YC, Ding C, Magkos F.

The epidemiology of obesity. Metabolism — Steenblock C, Schwarz PEH, Ludwig B, Linkermann A, Zimmet P, Kulebyakin K, et al. COVID and metabolic disease: mechanisms and clinical management.

Lancet Diabetes Endocrinol 9 11 — Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J Hepatol 71 4 — Lonardo A, Nascimbeni F, Mantovani A, Targher G. Hypertension; diabetes; atherosclerosis and NASH: Cause or consequence?

Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF diabetes atlas: Global; regional and country-level diabetes prevalence estimates for and projections for Diabetes Res Clin Pract Kaul K, Apostolopoulou M, Roden M. Insulin resistance in type 1 diabetes mellitus.

Metabolism 64 12 — Nadeau KJ, Regensteiner JG, Bauer TA, Brown MS, Dorosz JL, Hull A, et al. Insulin resistance in adolescents with type 1 diabetes and its relationship to cardiovascular function.

J Clin Endocrinol Metab 95 2 — Cree-Green M, Newcomer BR, Brown MS, Baumgartner AD, Bergman B, Drew B, et al. Delayed skeletal muscle mitochondrial ADP recovery in youth with type 1 diabetes relates to muscle insulin resistance. Diabetes 64 2 — Schauer IE, Snell-Bergeon JK, Bergman BC, Maahs DM, Kretowski A, Eckel RH, et al.

Insulin resistance; defective insulin-mediated fatty acid suppression; and coronary artery calcification in subjects with and without type 1 diabetes: The CACTI study. Diabetes 60 1 — Donga E, Dekkers OM, Corssmit EP, Romijn JA.

Insulin resistance in patients with type 1 diabetes assessed by glucose clamp studies: systematic review and meta-analysis. Eur J Endocrinol 1 —9. Liu HY, Cao SY, Hong T, Han J, Liu Z, Cao W.

Insulin is a stronger inducer of insulin resistance than hyperglycemia in mice with type 1 diabetes mellitus T1DM. J Biol Chem 40 — Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metab 29 5 — Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes.

Diabetologia 46 1 :3— Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature —6. Rattarasarn C. Dysregulated lipid storage and its relationship with insulin resistance and cardiovascular risk factors in non-obese asian patients with type 2 diabetes.

Adipocyte 7 2 — Pop A, Clenciu D, Anghel M, Radu S, Socea B, Mota E, et al. Insulin resistance is associated with all chronic complications in type 1 diabetes. J Diabetes. Pan Y, Zhong S, Zhou K, Tian Z, Chen F, Liu Z, et al. Association between diabetes complications and the triglyceride-glucose index in hospitalized patients with type 2 diabetes.

J Diabetes Res Wang S, Shi J, Peng Y, Fang Q, Mu Q, Gu W, et al. Stronger association of triglyceride glucose index than the HOMA-IR with arterial stiffness in patients with type 2 diabetes: a real-world single-centre study.

Cardiovasc Diabetol 20 1 Jia G, Whaley-Connell A, Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia 61 1 —8. Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy.

Nat Rev Endocrinol 12 3 — Svensson M, Eriksson JW. Insulin resistance in diabetic nephropathy—cause or consequence? Diabetes Metab Res Rev 22 5 — Godsland IF.

Insulin resistance and hyperinsulinaemia in the development and progression of cancer. Clin Sci Lond. Hernandez AV, Pasupuleti V, Benites-Zapata VA, Thota P, Deshpande A, Perez-Lopez FR. Insulin resistance and endometrial cancer risk: A systematic review and meta-analysis.

Eur J Cancer. Yin DT, He H, Yu K, Xie J, Lei M, Ma R, et al. The association between thyroid cancer and insulin resistance; metabolic syndrome and its components: A systematic review and meta-analysis. Int J Surg , —

Metabolism and insulin sensitivity

Metabolism and insulin sensitivity -

Many different supplements can help increase insulin sensitivity, including vitamin C , probiotics , and magnesium. That said, many other supplements, such as zinc, folate, and vitamin D, do not appear to have this effect, according to research As with all supplements, there is a risk they may interact with any current medication you may be taking.

Insulin is an important hormone that has many roles in the body. When your insulin sensitivity is low, it puts pressure on your pancreas to increase insulin production to clear sugar from your blood.

Low insulin sensitivity is also called insulin resistance. Insulin sensitivity describes how your cells respond to insulin. Symptoms develop when your cells are resistant to insulin. Insulin resistance can result in chronically high blood sugar levels, which are thought to increase your risk of many diseases, including diabetes and heart disease.

Insulin resistance is bad for your health, but having increased insulin sensitivity is good. It means your cells are responding to insulin in a healthier way, which reduces your chance of developing diabetes.

Consider trying some of the suggestions in this article to help increase your insulin sensitivity and lower your risk of disease but be sure to talk with a healthcare professional first before making changes, especially adding supplements to your treatment regimen.

Read this article in Spanish. Our experts continually monitor the health and wellness space, and we update our articles when new information becomes available. VIEW ALL HISTORY.

This article is based on scientific evidence, written by experts and fact checked by experts. Our team of licensed nutritionists and dietitians strive to be objective, unbiased, honest and to present both sides of the argument.

This article contains scientific references. The numbers in the parentheses 1, 2, 3 are clickable links to peer-reviewed scientific papers. Has taking insulin led to weight gain for you? Learn why this happens, plus how you can manage your weight once you've started insulin treatment.

When it comes to managing diabetes, adding the right superfoods to your diet is key. Try these simple, delicious recipes for breakfast, lunch, and…. A Quiz for Teens Are You a Workaholic? How Well Do You Sleep? Health Conditions Discover Plan Connect. Nutrition Evidence Based Top Natural Ways to Improve Your Insulin Sensitivity.

Medically reviewed by Kelly Wood, MD — By Ryan Raman, MS, RD — Updated on October 30, Get more sleep. Exercise more. Explore our top resources. Reduce stress. Diabetes Metab Res Rev. Ginsberg HN. Insulin resistance and cardiovascular disease. Bloomgarden ZT. Insulin resistance, dyslipidemia, and cardiovascular disease.

Kozakova M, Natali A, Dekker J, Beck-Nielsen H, Laakso M, Nilsson P, Balkau B, Ferrannini E. Insulin sensitivity and carotid intima-media thickness: relationship between insulin sensitivity and cardiovascular risk study.

Min J, Weitian Z, Peng C, Yan P, Bo Z, Yan W, Yun B, Xukai W. Correlation between insulin-induced estrogen receptor methylation and atherosclerosis. Chanda D, Luiken JJ, Glatz JF. Signaling pathways involved in cardiac energy metabolism. FEBS Lett. Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D, Orci L, Unger RH.

Lipotoxic heart disease in obese rats: implications for human obesity. Ramírez E, Picatoste B, González-Bris A, Oteo M, Cruz F, Caro-Vadillo A, Egido J, Tuñón J, Morcillo MA, Lorenzo Ó. Sitagliptin improved glucose assimilation in detriment of fatty-acid utilization in experimental type-II diabetes: role of GLP-1 isoforms in Glut4 receptor trafficking.

Goldberg IJ. Clinical review diabetic dyslipidemia: causes and consequences. Sparks JD, Sparks CE, Adeli K. Selective hepatic insulin resistance, VLDL overproduction, and hypertriglyceridemia.

Zimmet P, Alberti KG, Shaw J. Global and societal implications of the diabetes epidemic. Austin MA, Hokanson JE, Edwards KL. Hypertriglyceridemia as a cardiovascular risk factor.

Hokanson JE. Hypertriglyceridemia and risk of coronary heart disease. Curr Cardiol Rep. Sung KC, Park HY, Kim MJ, Reaven G. Metabolic markers associated with insulin resistance predict type 2 diabetes in Koreans with normal blood pressure or prehypertension. Ginsberg HN, Zhang YL, Hernandez-Ono A.

Metabolic syndrome: focus on dyslipidemia. Yadav R, Hama S, Liu Y, Siahmansur T, Schofield J, Syed AA, France M, Pemberton P, Adam S, Ho JH, et al. Effect of Roux-en-Y bariatric surgery on lipoproteins, insulin resistance, and systemic and vascular inflammation in obesity and diabetes.

Front Immunol. de Luca C, Olefsky JM. Inflammation and insulin resistance. den Boer MA, Voshol PJ, Kuipers F, Romijn JA, Havekes LM. Hepatic glucose production is more sensitive to insulin-mediated inhibition than hepatic VLDL-triglyceride production.

Am J Physiol Endocrinol Metab. Semenkovich CF. Insulin resistance and atherosclerosis. Lewis GF, Steiner G. Acute effects of insulin in the control of VLDL production in humans.

Implications for the insulin-resistant state. Haas ME, Attie AD, Biddinger SB. The regulation of ApoB metabolism by insulin. Trends Endocrinol Metab.

Verges B. Pathophysiology of diabetic dyslipidaemia: where are we? Pont F, Duvillard L, Florentin E, Gambert P, Verges B. Early kinetic abnormalities of apoB-containing lipoproteins in insulin-resistant women with abdominal obesity.

Hoogeveen RC, Gaubatz JW, Sun W, Dodge RC, Crosby JR, Jiang J, Couper D, Virani SS, Kathiresan S, Boerwinkle E, et al. Small dense low-density lipoprotein-cholesterol concentrations predict risk for coronary heart disease: the Atherosclerosis Risk in Communities ARIC study.

Packard CJ. Triacylglycerol-rich lipoproteins and the generation of small, dense low-density lipoprotein. Biochem Soc Trans.

Sandhofer A, Kaser S, Ritsch A, Laimer M, Engl J, Paulweber B, Patsch JR, Ebenbichler CF. Cholesteryl ester transfer protein in metabolic syndrome. Rashid S, Watanabe T, Sakaue T, Lewis GF. Mechanisms of HDL lowering in insulin resistant, hypertriglyceridemic states: the combined effect of HDL triglyceride enrichment and elevated hepatic lipase activity.

Clin Biochem. von Bibra H, Saha S, Hapfelmeier A, Muller G, Schwarz PEH. Kim MK, Ahn CW, Kang S, Nam JS, Kim KR, Park JS. Relationship between the triglyceride glucose index and coronary artery calcification in Korean adults.

Mazidi M, Kengne AP, Katsiki N, Mikhailidis DP, Banach M. J Diabetes Complications. Jorge-Galarza E, Posadas-Romero C, Torres-Tamayo M, Medina-Urrutia AX, Rodas-Diaz MA, Posadas-Sanchez R, Vargas-Alarcon G, Gonzalez-Salazar MD, Cardoso-Saldana GC, Juarez-Rojas JG.

Insulin resistance in adipose tissue but not in liver is associated with aortic valve calcification. Dis Markers. Zhou MS, Schulman IH, Zeng Q. Link between the renin—angiotensin system and insulin resistance: implications for cardiovascular disease. Vasc Med.

Zhou MS, Schulman IH, Raij L. Nitric oxide, angiotensin II, and hypertension. Semin Nephrol. Landsberg L. Insulin resistance and hypertension. Clin Exp Hypertens. Briet M, Schiffrin EL. Aldosterone: effects on the kidney and cardiovascular system.

Nat Rev Nephrol. Oana F, Takeda H, Hayakawa K, Matsuzawa A, Akahane S, Isaji M, Akahane M. Goossens GH. The renin—angiotensin system in the pathophysiology of type 2 diabetes. Obesity Facts.

Schulman IH, Zhou MS. Vascular insulin resistance: a potential link between cardiovascular and metabolic diseases. Curr Hypertens Rep. Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Vascular inflammation, insulin resistance, and endothelial dysfunction in salt-sensitive hypertension: role of nuclear factor kappa B activation.

J Hypertens. Andreozzi F, Laratta E, Sciacqua A, Perticone F, Sesti G. Angiotensin II impairs the insulin signaling pathway promoting production of nitric oxide by inducing phosphorylation of insulin receptor substrate-1 on Ser and Ser in human umbilical vein endothelial cells.

Circ Res. Wei Y, Whaley-Connell AT, Chen K, Habibi J, Uptergrove GM, Clark SE, Stump CS, Ferrario CM, Sowers JR. NADPH oxidase contributes to vascular inflammation, insulin resistance, and remodeling in the transgenic mRen2 rat.

Matsuura K, Hagiwara N. The pleiotropic effects of ARB in vascular endothelial progenitor cells. Curr Vasc Pharmacol. Group NS, McMurray JJ, Holman RR, Haffner SM, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, et al.

Effect of valsartan on the incidence of diabetes and cardiovascular events. Article Google Scholar. Perlstein TS, Henry RR, Mather KJ, Rickels MR, Abate NI, Grundy SM, Mai Y, Albu JB, Marks JB, Pool JL, et al. Effect of angiotensin receptor blockade on insulin sensitivity and endothelial function in abdominally obese hypertensive patients with impaired fasting glucose.

Clin Sci Lond. Kim JA, Montagnani M, Koh KK, Quon MJ. Reciprocal relationships between insulin resistance and endothelial dysfunction: molecular and pathophysiological mechanisms.

Tousoulis D, Simopoulou C, Papageorgiou N, Oikonomou E, Hatzis G, Siasos G, Tsiamis E, Stefanadis C. Endothelial dysfunction in conduit arteries and in microcirculation. Novel therapeutic approaches. Pharmacol Ther. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Westergren HU, Svedlund S, Momo RA, Blomster JI, Wahlander K, Rehnstrom E, Greasley PJ, Fritsche-Danielson R, Oscarsson J, Gan LM.

Insulin resistance, endothelial function, angiogenic factors and clinical outcome in non-diabetic patients with chest pain without myocardial perfusion defects.

Dinesh Shah A, Langenberg C, Rapsomaniki E, Denaxas S, Pujades-Rodriguez M, Gale CP, Deanfield J, Smeeth L, Timmis A, Hemingway H. Type 2 diabetes and incidence of a wide range of cardiovascular diseases: a cohort study in 1.

Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: have all risk factors the same strength? Ciccone MM, Cortese F, Gesualdo M, Donvito I, Carbonara S, De Pergola G.

Endocr Metab Immune Disord Drug Targets. Selvin E, Marinopoulos S, Berkenblit G, Rami T, Brancati FL, Powe NR, Golden SH. Meta-analysis: glycosylated hemoglobin and cardiovascular disease in diabetes mellitus. Ann Intern Med. Meyer ML, Gotman NM, Soliman EZ, Whitsel EA, Arens R, Cai J, Daviglus ML, Denes P, Gonzalez HM, Moreiras J, et al.

Paneni F, Volpe M, Luscher TF, Cosentino F. Ceriello A. Vasc Pharmacol. Fiorentino TV, Prioletta A, Zuo P, Folli F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr Pharm Des. Pistrosch F, Natali A, Hanefeld M. Is hyperglycemia a cardiovascular risk factor?

Giacco F, Brownlee M. Oxidative stress and diabetic complications. Nowotny K, Jung T, Hohn A, Weber D, Grune T. Advanced glycation end products and oxidative stress in type 2 diabetes mellitus.

Yan SF, Ramasamy R, Schmidt AM. The RAGE axis: a fundamental mechanism signaling danger to the vulnerable vasculature. Sonnenblick EH, Stam AC Jr. Cardiac muscle: activation and contraction. Annu Rev Physiol. Johansen L, Quistorff B. Int J Sports Med. Duffield R, Dawson B, Goodman C. Energy system contribution to m and m track running events.

J Sci Med Sport. Kassiotis C, Rajabi M, Taegtmeyer H. Metabolic reserve of the heart: the forgotten link between contraction and coronary flow. Prog Cardiovasc Dis. Kota SK, Kota SK, Jammula S, Panda S, Modi KD.

Effect of diabetes on alteration of metabolism in cardiac myocytes: therapeutic implications. Diabetes Technol Ther. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabolism in the normal and failing heart.

Carley AN, Severson DL. Fatty acid metabolism is enhanced in type 2 diabetic hearts. Brandt JM, Djouadi F, Kelly DP. Fatty acids activate transcription of the muscle carnitine palmitoyltransferase I gene in cardiac myocytes via the peroxisome proliferator-activated receptor alpha.

Goodwin GW, Taegtmeyer H. Improved energy homeostasis of the heart in the metabolic state of exercise. Opie LH. Cardiac metabolism—emergence, decline, and resurgence. Part II. Cardiovasc Res. Henning SL, Wambolt RB, Schonekess BO, Lopaschuk GD, Allard MF.

Contribution of glycogen to aerobic myocardial glucose utilization. Wu G, Fang YZ, Yang S, Lupton JR, Turner ND. Glutathione metabolism and its implications for health.

The Journal of nutrition. Shao D, Tian R. Glucose transporters in cardiac metabolism and hypertrophy. Comp Physiol. Malfitano C, de Souza Junior AL, Carbonaro M, Bolsoni-Lopes A, Figueroa D, de Souza LE, Silva KA, Consolim-Colombo F, Curi R, Irigoyen MC.

Glucose and fatty acid metabolism in infarcted heart from streptozotocin-induced diabetic rats after 2 weeks of tissue remodeling. Kolwicz SC Jr, Purohit S, Tian R. Cardiac metabolism and its interactions with contraction, growth, and survival of cardiomyocytes. Wright JJ, Kim J, Buchanan J, Boudina S, Sena S, Bakirtzi K, Ilkun O, Theobald HA, Cooksey RC, Kandror KV, et al.

Mechanisms for increased myocardial fatty acid utilization following short-term high-fat feeding. Su X, Abumrad NA. Cellular fatty acid uptake: a pathway under construction. Ajith TA, Jayakumar TG. Peroxisome proliferator-activated receptors in cardiac energy metabolism and cardiovascular disease.

Clin Exp Pharmacol Physiol. Oakes ND, Thalen P, Aasum E, Edgley A, Larsen T, Furler SM, Ljung B, Severson D.

Cardiac metabolism in mice: tracer method developments and in vivo application revealing profound metabolic inflexibility in diabetes. Lipid metabolism and signaling in cardiac lipotoxicity. Goldberg IJ, Trent CM, Schulze PC. Lipid metabolism and toxicity in the heart. Lipoapoptosis: its mechanism and its diseases.

Park TS, Hu Y, Noh HL, Drosatos K, Okajima K, Buchanan J, Tuinei J, Homma S, Jiang XC, Abel ED, et al. Ceramide is a cardiotoxin in lipotoxic cardiomyopathy. J Lipid Res. Liu Y, Neumann D, Glatz JF, Luiken JJ.

Molecular mechanism of lipid-induced cardiac insulin resistance and contractile dysfunction. Prostaglandins Leukot Essent Fatty Acids. Article PubMed Central PubMed Google Scholar. Feuvray D, Idell-Wenger JA, Neely JR.

Effects of ischemia on rat myocardial function and metabolism in diabetes. Fricovsky ES, Suarez J, Ihm SH, Scott BT, Suarez-Ramirez JA, Banerjee I, Torres-Gonzalez M, Wang H, Ellrott I, Maya-Ramos L, et al. Excess protein O -GlcNAcylation and the progression of diabetic cardiomyopathy.

Am J Physiol Regul Integr Comp Physiol. Hwang YC, Kaneko M, Bakr S, Liao H, Lu Y, Lewis ER, Yan S, Ii S, Itakura M, Rui L, et al. Central role for aldose reductase pathway in myocardial ischemic injury.

FASEB J. Zuurbier CJ, Eerbeek O, Goedhart PT, Struys EA, Verhoeven NM, Jakobs C, Ince C. Inhibition of the pentose phosphate pathway decreases ischemia—reperfusion-induced creatine kinase release in the heart. Salabei JK, Lorkiewicz PK, Mehra P, Gibb AA, Haberzettl P, Hong KU, Wei X, Zhang X, Li Q, Wysoczynski M, et al.

Type 2 Diabetes Dysregulates Glucose Metabolism in Cardiac Progenitor Cells. Keller U, Lustenberger M, Stauffacher W. van der Vusse GJ, van Bilsen M, Glatz JF.

Cardiac fatty acid uptake and transport in health and disease. Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, Koves T, Gardell SJ, Kruger M, Hoppel CL, et al. The failing heart relies on ketone bodies as a fuel.

Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P, Ng CP, Huang Y, Haldar S, Verdin E. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell metabolism. Article PubMed CAS PubMed Central Google Scholar.

Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte GR, Tran D, Perez G, Gutierrez-Casado E, Koike S, Knotts TA, et al. A ketogenic diet extends longevity and healthspan in adult mice. Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini DM. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing.

Kosinski C, Jornayvaz FR: Effects of Ketogenic Diets on Cardiovascular Risk Factors: Evidence from Animal and Human Studies. Nutrients , 9 5. Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ.

Comparison of the atkins, ornish, weight watchers, and zone diets for weight loss and heart disease risk reduction: a randomized trial.

Kim JA, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin resistance. Jeong EM, Chung J, Liu H, Go Y, Gladstein S, Farzaneh-Far A, Lewandowski ED, Dudley SC Jr. Role of mitochondrial oxidative stress in glucose tolerance, insulin resistance, and cardiac diastolic dysfunction.

Mei Y, Thompson MD, Cohen RA, Tong X. Endoplasmic reticulum stress and related pathological processes. J Pharm Biomed Anal. Taddeo EP, Laker RC, Breen DS, Akhtar YN, Kenwood BM, Liao JA, Zhang M, Fazakerley DJ, Tomsig JL, Harris TE, et al.

Opening of the mitochondrial permeability transition pore links mitochondrial dysfunction to insulin resistance in skeletal muscle. Mol Metab. Mandavia CH, Aroor AR, Demarco VG, Sowers JR. Molecular and metabolic mechanisms of cardiac dysfunction in diabetes.

Life Sci. Download references. VO, SN, OE, CA and FZ conducted a review of the literature and contributed to conception and design and wrote the first draft the review; CS contributed to conception and design of the article and critically reviewed the drafts of the manuscript.

All authors read and approved the final manuscript. This study was supported by Fondo Nacional de Desarrollo Científico y Tecnológico FONDECYT , Lions Medical Research Foundation Australia , and Diabetes Australia.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile.

Faculty of Pharmacy, Department of Clinical Biochemistry and Immunology, University of Concepcion, Concepción, Chile. Department of Obstetrics and Gynecology, Ochsner Baptist Hospital, New Orleans, Louisiana, USA. You can also search for this author in PubMed Google Scholar.

Correspondence to Carlos Salomon. Open Access This article is distributed under the terms of the Creative Commons Attribution 4. Reprints and permissions. Ormazabal, V. et al. Association between insulin resistance and the development of cardiovascular disease.

Cardiovasc Diabetol 17 , Download citation. Received : 22 May Accepted : 20 August Published : 31 August Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative.

Skip to main content. Search all BMC articles Search. Download PDF. Review Open access Published: 31 August Association between insulin resistance and the development of cardiovascular disease Valeska Ormazabal 1 , Soumyalekshmi Nair 2 , Omar Elfeky 2 , Claudio Aguayo 3 , Carlos Salomon ORCID: orcid.

Zuñiga 3 Show authors Cardiovascular Diabetology volume 17 , Article number: Cite this article k Accesses Citations Altmetric Metrics details. Abstract For many years, cardiovascular disease CVD has been the leading cause of death around the world.

Background The pathological processes and risk factors associated with CVD begin as early as during childhood [ 1 ]. Insulin signaling Insulin is a potent anabolic hormone that exerts a variety of effects on many types of cells.

Full size image. Insulin resistance Insulin resistance is defined as an experimental or clinical condition in which insulin exerts a biological effect lower than expected. Cellular mechanisms of insulin resistance Insulin works on multiple processes, essentially providing an integrated set of signals that allows the correct balance between nutrient supply and demand [ 33 ].

Insulin resistance and cardiovascular disease Elevated levels of LDL, smoking, elevated blood pressure and type 1 and type 2 diabetes, are well known risk factors for CVD, however, insulin resistance, hyperglycaemia and inflammation can also lead to and predict adverse cardiovascular events.

Insulin resistance and dyslipidemia The dyslipidemia induced by insulin resistance and type 2 diabetes diabetic dyslipidemia [ 82 ] is characterized by the lipid triad: 1 high levels of plasma triglycerides, 2 low levels of HDL, and 3 the appearance of small dense low-density lipoproteins sdLDL , as well as an excessive postprandial lipemia [ 35 , 82 , 83 , 84 ].

Insulin resistance and lipoproteins profile alterations VLDL, very low-density lipoprotein, is assembled and produced in the liver, which depends on the availability of substrates and is tightly regulated by insulin [ 91 ]. Insulin resistance and endothelial dysfunction The integrity of the functional endothelium is a fundamental vascular health element.

Chronic hyperglycemia in cardiovascular disease The increased CVD risk in patients with type 2 diabetes has been known for many years [ ]. Insulin resistance and changes in the cardiac metabolism The thickest layer of the heart wall is the myocardium, composed of cardiac muscle cells, thus, the knowledge provided by skeletal muscle cell physiology helps explain the cardiac metabolic function [ ].

Instead, energy is stored in cardiac muscle cells in three forms: 1. Conclusions Insulin essentially provides an integrated set of signals allowing the balance between nutrient demand and availability. References Steinberger J, Daniels SR, American Heart Association Atherosclerosis H, Obesity in the Young C, American Heart Association Diabetes C.

Article PubMed Google Scholar Steinberger J, Moorehead C, Katch V, Rocchini AP. Article PubMed CAS Google Scholar Ferreira AP, Oliveira CE, Franca NM. Article PubMed Google Scholar Reaven G. Article PubMed CAS Google Scholar Wilcox G.

PubMed PubMed Central Google Scholar Gast KB, Tjeerdema N, Stijnen T, Smit JW, Dekkers OM. Article PubMed PubMed Central CAS Google Scholar Bornfeldt KE, Tabas I. Article PubMed PubMed Central CAS Google Scholar Davidson JA, Parkin CG.

Article PubMed PubMed Central Google Scholar Laakso M, Kuusisto J. Article PubMed CAS Google Scholar Janus A, Szahidewicz-Krupska E, Mazur G, Doroszko A. Article PubMed PubMed Central CAS Google Scholar Scott PH, Brunn GJ, Kohn AD, Roth RA, Lawrence JC Jr. Article PubMed CAS Google Scholar Bogan JS.

Article PubMed CAS Google Scholar Zimmer HG. Article PubMed Google Scholar Choi SM, Tucker DF, Gross DN, Easton RM, DiPilato LM, Dean AS, Monks BR, Birnbaum MJ.

Article PubMed PubMed Central CAS Google Scholar Duncan RE, Ahmadian M, Jaworski K, Sarkadi-Nagy E, Sul HS. Article PubMed PubMed Central CAS Google Scholar Czech MP, Tencerova M, Pedersen DJ, Aouadi M.

Article PubMed PubMed Central CAS Google Scholar Shulman GI. Article PubMed CAS Google Scholar Hojlund K. PubMed Google Scholar Kahn BB, Flier JS. Article PubMed CAS Google Scholar Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, Raptis SA. Article PubMed CAS Google Scholar Reaven GM. Article PubMed CAS Google Scholar Wu G, Meininger CJ.

Article CAS Google Scholar Wang CC, Gurevich I, Draznin B. Article PubMed CAS Google Scholar Berg J, Tymoczko J, Stryer L: Food intake and starvation induce metabolic changes.

Article CAS Google Scholar Bonora E. Google Scholar Goodwin PJ, Ennis M, Bahl M, Fantus IG, Pritchard KI, Trudeau ME, Koo J, Hood N. Article PubMed CAS Google Scholar Seriolo B, Ferrone C, Cutolo M.

A recent meta-analysis reported that increasing daily fiber intake by 15 or 35 grams compared to a low-fiber diet reduced homeostatic model assessment of insulin resistance HOMA-IR , leading to improvements in glycemic control, lipids, weight, and inflammation, as well as a reduction in premature mortality Not only is the amount of carbohydrate intake important, but the timing of major carbohydrate intake during the day is also a determining factor in the increase in glucose and insulin after meals and the improvement or otherwise of IR The results of some randomized controlled trial RCT studies suggest that it is advisable to consume at least half of the carbohydrates at lunch and to avoid consuming large amounts of carbohydrates at breakfast or dinner in order to control blood glucose spikes, which may be related to diurnal variations in insulin sensitivity — Results of another study showed that 10 hours of restrictive eating improved quality of life by reducing body weight and improving blood glucose, insulin sensitivity and related metabolic disorders Other dietary strategies have been shown to prevent high-fat diet-induced IR, such as the intake of flavonoid-rich natural products, like flavonoids, which upregulate the expression of related genes through cell surface G protein-coupled estrogen receptors Although lifestyle modification and weight loss are highly recommended to improve IR and its associated metabolic disorders, they have limited effectiveness, slow onset of action, and low feasibility.

Pharmacological treatments to increase insulin sensitivity will be described next. Currently, the main drugs that can effectively improve IR are anti-hyperglycemic drugs, including metformin, thiazolidinediones TZD , sodium glucose cotransporter SGLT -2 inhibitors SGLT2i , etc.

Metformin, the most commonly used insulin-sensitizing agent, has been a guideline-recommended first-line treatment for T2DM for decades and has recently found new applications in the prevention and treatment of various diseases, including metabolic disorders and cardiovascular diseases Metformin improves IR by modulating metabolic mechanisms and mitochondrial biogenesis through altering microRNAs levels by AMPK-dependent or AMPK-independent mechanisms TZDs, such as pioglitazone, are potent insulin sensitizers targeting PPARγ and PI3K, regulating the transcription of nuclear transcription factors, stimulating mainly white adipose tissue remodeling, and regulating lipid flux for insulin sensitization and beta cell protection , SGLT2i is a relatively new class of glucose-lowering drug that not only lowers blood glucose by inhibiting renal glucose reuptake, leading to increased urinary glucose excretion and lower blood glucose, but also improves insulin sensitivity in patients with T2DM by reducing body weight or glucose toxicity , And in a randomized, double-blind, placebo-controlled clinical trial, it was shown that 8 weeks of treatment with SGLT2i empagliflozin restored insulin sensitivity in the hypothalamus of patients with prediabetes Glucose-lowering drugs have also shown good, stacked effects in patients who do not have good response with one drug alone.

For example, the addition of rosiglitazone to metformin can be clinically important in improving glycemic control, insulin sensitivity and beta-cell function The addition of sitagliptin or metformin to pioglitazone monotherapy also leads to faster and better improvement in IR and inflammatory status parameters Other therapies, as well as some new drugs in clinical trials, such as anti-inflammatory drugs, drugs that target hepatic lipid and energy metabolism, renin-angiotensin-aldosterone system blockers, vitamin D, antioxidants, probiotics and fecal transplants, have also shown improvement in IR Among them, selected clinical trials in the last decade have been listed in Table 2.

As mentioned previously, low-grade chronic inflammation is associated with IR and metabolic disturbances. For example, in in vitro and in vivo mouse models of diet-induced hyperinsulinemia, low-dose naltrexone attenuates hyperinsulinemia-induced proinflammatory cytokine release and restores insulin sensitivity However, it is worth noting that corticosteroids can cause IR and hyperglycemia due to their metabolic effects, and statins also increase the risk of IR, although they can reduce circulating inflammatory markers TCM plays an equally critical role in the treatment of many acute and chronic diseases, especially its adeptness in restoring the dynamic balance of the body in systemic diseases.

Its main therapeutic measures include herbal medicine, acupuncture and Tui Na. Several classical herbal formulations have been widely used in the clinical treatment of T2DM and various other metabolic disorders.

For example, GegenQinlian decoction improves IR in fat, liver and muscle tissue through a variety of compounds, targets, pathways and mechanisms Yi Qi Zeng Min Tang has been shown to improve IR in high-fat fed Sprague-Dawley rats without increasing body weight Because it reduced the expression of PI3K p85 mRNA and IRS1 protein, Fu Fang Zhen Zhu Tiao Zhi formula similarly improved IR in vitro and in rats with metabolic syndrome Gui Zhi Fu Ling Wan, Dingkun Pill and Liuwei Dihuang Pills are herbal formulas widely used in the treatment of gynecological disorders and have the effect of harmonizing Qi and blood or dispelling blood stasis in Chinese medical theory.

In addition, the efficacy of acupuncture in improving IR is equally impressive, as a recent meta-analysis showed that acupuncture improved HOMA-IR and ISI as well as fasting blood glucose FBG , 2h postprandial blood glucose 2hPG and fasting insulin FINS levels, with fewer adverse events The increased incidence of IR and its vital role as a major and common cause of numerous metabolic diseases have created an urgent need to gain insight into the etiology and pathogenesis of IR, as well as to explore better early diagnostic methods and therapeutic strategies for it.

The diagnosis of insulin resistance is currently inconclusive, while it is important to detect IR early and predict individual response to treatment. In addition to the few simple indices of IR calculated from biochemical or anthropometric variables currently in use, emerging biomarkers may now be the way forward, but this still needs to be supported by clinical data.

Different ranges and criteria are also needed for the diagnosis and monitoring of different metabolic diseases.

As mentioned above, IR is a central mechanism in many metabolic diseases. Since this is the case, IR should be considered as a therapeutic target for patients with a combination of multiple metabolic diseases so that multiple diseases can be treated simultaneously with the same treatment approach, thereby reducing healthcare expenditures.

Although there is no universally accepted theory to explain the mechanisms that cause IR. Nevertheless, there is growing evidence linking ectopic lipid accumulation, ER stress, plasma concentration of inflammatory cytokines, oxidative stress, abnormalities in insulin signaling, and other factors to IR.

In recent years, the exploration of the molecular mechanisms of IR has also led to the emergence of new therapeutic concepts beyond metformin and TZD.

Regardless lifestyle modification remains the most basic and least costly intervention. Normative criteria need to be developed for different metabolic diseases considering IR as a focus. FL and HJ provided the idea of the manuscript. XZ, XA, and CY contributed equally to this manuscript.

XZ, XA, WS, and CY drafted the manuscript and searched the relevant literature. XZ and XA drafted the figures, and all authors approved the final version of the manuscript.

All authors agree to be accountable for all aspects of work ensuring integrity and accuracy. All authors contributed to the article and approved the submitted version.

This work was supported by Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine.

No: ZYYCXTD-D The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

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

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Bugianesi E, McCullough AJ, Marchesini G.

Insulin resistance: a metabolic pathway to chronic liver disease. Hepatology 42 5 — doi: PubMed Abstract CrossRef Full Text Google Scholar. Sharma VR, Matta ST, Haymond MW, Chung ST. Measuring insulin resistance in humans.

Horm Res Paediatr 93 — Muniyappa R, Madan R, Varghese RT. Assessing insulin sensitivity and resistance in humans. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, editors. South Dartmouth MA: MDText. com; Inc.

Copyright © ; MDText. Google Scholar. Gar C, Rottenkolber M, Prehn C, Adamski J, Seissler J, Lechner A. Serum and plasma amino acids as markers of prediabetes; insulin resistance; and incident diabetes.

Crit Rev Clin Lab Sci 55 1 — Park SE, Park CY, Sweeney G. Biomarkers of insulin sensitivity and insulin resistance: Past; present and future. Crit Rev Clin Lab Sci 52 4 — Milburn MV, Lawton KA.

Application of metabolomics to diagnosis of insulin resistance. Annu Rev Med — Yang R, Hu Y, Lee CH, Liu Y, Diaz-Canestro C, Fong CHY, et al. PM20D1 is a circulating biomarker closely associated with obesity; insulin resistance and metabolic syndrome. Eur J Endocrinol 2 — Saklayen MG.

The global epidemic of the metabolic syndrome. Curr Hypertens Rep 20 2 Chooi YC, Ding C, Magkos F. The epidemiology of obesity.

Metabolism — Steenblock C, Schwarz PEH, Ludwig B, Linkermann A, Zimmet P, Kulebyakin K, et al. COVID and metabolic disease: mechanisms and clinical management. Lancet Diabetes Endocrinol 9 11 — Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al.

The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J Hepatol 71 4 — Lonardo A, Nascimbeni F, Mantovani A, Targher G. Hypertension; diabetes; atherosclerosis and NASH: Cause or consequence? Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al.

IDF diabetes atlas: Global; regional and country-level diabetes prevalence estimates for and projections for Diabetes Res Clin Pract Kaul K, Apostolopoulou M, Roden M.

Insulin resistance in type 1 diabetes mellitus. Metabolism 64 12 — Nadeau KJ, Regensteiner JG, Bauer TA, Brown MS, Dorosz JL, Hull A, et al. Insulin resistance in adolescents with type 1 diabetes and its relationship to cardiovascular function.

J Clin Endocrinol Metab 95 2 — Cree-Green M, Newcomer BR, Brown MS, Baumgartner AD, Bergman B, Drew B, et al. Delayed skeletal muscle mitochondrial ADP recovery in youth with type 1 diabetes relates to muscle insulin resistance. Diabetes 64 2 — Schauer IE, Snell-Bergeon JK, Bergman BC, Maahs DM, Kretowski A, Eckel RH, et al.

Insulin resistance; defective insulin-mediated fatty acid suppression; and coronary artery calcification in subjects with and without type 1 diabetes: The CACTI study. Diabetes 60 1 — Donga E, Dekkers OM, Corssmit EP, Romijn JA.

Insulin resistance in patients with type 1 diabetes assessed by glucose clamp studies: systematic review and meta-analysis. Eur J Endocrinol 1 —9. Liu HY, Cao SY, Hong T, Han J, Liu Z, Cao W. Insulin is a stronger inducer of insulin resistance than hyperglycemia in mice with type 1 diabetes mellitus T1DM.

J Biol Chem 40 — Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metab 29 5 — Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46 1 :3— Kahn SE, Hull RL, Utzschneider KM.

Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature —6. Rattarasarn C. Dysregulated lipid storage and its relationship with insulin resistance and cardiovascular risk factors in non-obese asian patients with type 2 diabetes.

Adipocyte 7 2 — Pop A, Clenciu D, Anghel M, Radu S, Socea B, Mota E, et al. Insulin resistance is associated with all chronic complications in type 1 diabetes. J Diabetes. Pan Y, Zhong S, Zhou K, Tian Z, Chen F, Liu Z, et al.

Association between diabetes complications and the triglyceride-glucose index in hospitalized patients with type 2 diabetes. J Diabetes Res Wang S, Shi J, Peng Y, Fang Q, Mu Q, Gu W, et al.

Stronger association of triglyceride glucose index than the HOMA-IR with arterial stiffness in patients with type 2 diabetes: a real-world single-centre study. Cardiovasc Diabetol 20 1 Jia G, Whaley-Connell A, Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease.

Diabetologia 61 1 —8. Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Nat Rev Endocrinol 12 3 — Svensson M, Eriksson JW. Insulin resistance in diabetic nephropathy—cause or consequence?

Diabetes Metab Res Rev 22 5 — Godsland IF. Insulin resistance and hyperinsulinaemia in the development and progression of cancer. Clin Sci Lond. Hernandez AV, Pasupuleti V, Benites-Zapata VA, Thota P, Deshpande A, Perez-Lopez FR.

Insulin resistance and endometrial cancer risk: A systematic review and meta-analysis. Eur J Cancer. Yin DT, He H, Yu K, Xie J, Lei M, Ma R, et al. The association between thyroid cancer and insulin resistance; metabolic syndrome and its components: A systematic review and meta-analysis.

Int J Surg , — CrossRef Full Text Google Scholar. Kim NH, Chang Y, Lee SR, Ryu S, Kim HJ. Glycemic status; insulin resistance; and risk of pancreatic cancer mortality in individuals with and without diabetes. Am J Gastroenterol 11 —8.

Pan K, Chlebowski RT, Mortimer JE, Gunter MJ, Rohan T, Vitolins MZ, et al. Insulin resistance and breast cancer incidence and mortality in postmenopausal women in the women's health initiative. Cancer 16 — Chiefari E, Mirabelli M, La Vignera S, Tanyolaç S, Foti DP, Aversa A, et al.

Insulin resistance and cancer: In search for a causal link. Int J Mol Sci 22 Barber TM, Kyrou I, Randeva HS, Weickert MO. Mechanisms of insulin resistance at the crossroad of obesity with associated metabolic abnormalities and cognitive dysfunction.

Int J Mol Sci 22 2. Mu N, Zhu Y, Wang Y, Zhang H, Xue F. Insulin resistance: a significant risk factor of endometrial cancer. Gynecol Oncol 3 —7. Arcidiacono B, Iiritano S, Nocera A, Possidente K, Nevolo MT, Ventura V, et al. Insulin resistance and cancer risk: an overview of the pathogenetic mechanisms.

Exp Diabetes Res Inoue M, Tsugane S. Insulin resistance and cancer: epidemiological evidence. Endocr Relat Cancer 19 5 :F1—8. Kong Y, Hsieh CH, Alonso LC. Front Endocrinol Lausanne Ramos-Lopez O, Riezu-Boj JI, Milagro FI, Martinez JA.

DNA methylation signatures at endoplasmic reticulum stress genes are associated with adiposity and insulin resistance. Mol Genet Metab 1 —8. Kwa M, Plottel CS, Blaser MJ, Adams S. The intestinal microbiome and estrogen receptor-positive female breast cancer.

J Natl Cancer Inst 8. Adeva-Andany MM, Martínez-Rodríguez J, González-Lucán M, Fernández-Fernández C, Castro-Quintela E. Insulin resistance is a cardiovascular risk factor in humans. Diabetes Metab Syndr 13 2 — Adeva-Andany MM, Fernández-Fernández C, Carneiro-Freire N, Castro-Quintela E, Pedre-Piñeiro A, Seco-Filgueira M.

Insulin resistance underlies the elevated cardiovascular risk associated with kidney disease and glomerular hyperfiltration. Rev Cardiovasc Med 21 1 — Saely CH, Aczel S, Marte T, Langer P, Hoefle G, Drexel H.

The metabolic syndrome; insulin resistance; and cardiovascular risk in diabetic and nondiabetic patients. J Clin Endocrinol Metab 90 10 — Zhang X, Li J, Zheng S, Luo Q, Zhou C, Wang C. Fasting insulin; insulin resistance; and risk of cardiovascular or all-cause mortality in non-diabetic adults: a meta-analysis.

Biosci Rep 37 5. Eddy D, Schlessinger L, Kahn R, Peskin B, Schiebinger R. Relationship of insulin resistance and related metabolic variables to coronary artery disease: a mathematical analysis.

Diabetes Care 32 2 —6. Novo G, Manno G, Russo R, Buccheri D, Dell'Oglio S, Morreale P, et al. Impact of insulin resistance on cardiac and vascular function.

Int J Cardiol —9. Wang M, Li Y, Li S, Lv J. Endothelial dysfunction and diabetic cardiomyopathy. Front Endocrinol Nakamura M, Sadoshima J. Cardiomyopathy in obesity, insulin resistance and diabetes. J Physiol 14 — Qi Y, Xu Z, Zhu Q, Thomas C, Kumar R, Feng H, et al. Myocardial loss of IRS1 and IRS2 causes heart failure and is controlled by p38α MAPK during insulin resistance.

Diabetes 62 11 — Razani B, Chakravarthy MV, Semenkovich CF. Insulin resistance and atherosclerosis. Endocrinol Metab Clin North Am 37 3 — Fernández-Real JM, Ricart W. Insulin resistance and chronic cardiovascular inflammatory syndrome. Endocr Rev 24 3 — Kim SH, Reaven G.

Sex differences in insulin resistance and cardiovascular disease risk. J Clin Endocrinol Metab 98 11 :E— Robins SJ, Rubins HB, Faas FH, Schaefer EJ, Elam MB, Anderson JW, et al. Insulin resistance and cardiovascular events with low HDL cholesterol: the veterans affairs HDL intervention trial VA-HIT.

Diabetes Care 26 5 —7. Abdul-Ghani MA, Jayyousi A, DeFronzo RA, Asaad N, Al-Suwaidi J. Insulin resistance the link between T2DM and CVD: Basic mechanisms and clinical implications. Curr Vasc Pharmacol 17 2 — Muzurović E, Mikhailidis DP, Mantzoros C. Non-alcoholic fatty liver disease; insulin resistance; metabolic syndrome and their association with vascular risk.

Metabolism Valenti L, Bugianesi E, Pajvani U, Targher G. Nonalcoholic fatty liver disease: cause or consequence of type 2 diabetes?

Liver Int 36 11 — Dongiovanni P, Stender S, Pietrelli A, Mancina RM, Cespiati A, Petta S, et al. Causal relationship of hepatic fat with liver damage and insulin resistance in nonalcoholic fatty liver.

J Intern Med 4 — PubMed Abstract Google Scholar. Watt MJ, Miotto PM, De Nardo W, Montgomery MK. The Liver as an Endocrine Organ-Linking NAFLD and Insulin Resistance. Endocr Rev Oct 1;40 5 Titchenell PMLazar MABirnbaum MJ.

Unraveling the Regulation of Hepatic Metabolism by Insulin. Trends Endocrinol Metab 28 7 — Huang JF, Tsai PC, Yeh ML, Huang CF, Huang CI, Hsieh MH, et al.

Risk stratification of non-alcoholic fatty liver disease across body mass index in a community basis. J Formos Med Assoc 1 Pt 1 — Enooku K, Kondo M, Fujiwara N, Sasako T, Shibahara J, Kado A, et al. Hepatic IRS1 and ß-catenin expression is associated with histological progression and overt diabetes emergence in NAFLD patients.

J Gastroenterol 53 12 — Bugianesi E, Gastaldelli A, Vanni E, Gambino R, Cassader M, Baldi S, et al. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: sites and mechanisms. Diabetologia 48 4 — Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, Contos MJ, Sterling RK, et al.

Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology 5 — Mantovani A, Byrne CD, Bonora E, Targher G. Nonalcoholic fatty liver disease and risk of incident type 2 diabetes: A meta-analysis.

Diabetes Care 41 2 — Shipovskaya AA, Dudanova OP, Kurbatova IV. The clinical significance of insulin resistance in non-diabetic patients with early forms of non-alcoholic fatty liver disease.

Ter Arkh. Alemzadeh R, Kichler J, Calhoun M. Spectrum of metabolic dysfunction in relationship with hyperandrogenemia in obese adolescent girls with polycystic ovary syndrome.

Eur J Endocrinol 6 —9. Macut D, Bjekić-Macut J, Rahelić D, Doknić M. Insulin and the polycystic ovary syndrome. Diabetes Res Clin Pract — Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications.

Endocr Rev 33 6 — Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, et al. Diagnosis and treatment of polycystic ovary syndrome: an endocrine society clinical practice guideline.

J Clin Endocrinol Metab 98 12 — Dumesic DA, Oberfield SE, Stener-Victorin E, Marshall JC, Laven JS, Legro RS. Scientific statement on the diagnostic criteria; epidemiology; pathophysiology; and molecular genetics of polycystic ovary syndrome.

Endocr Rev 36 5 — He FF, Li YM. Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a review. J Ovarian Res 13 1 Falcone T, Finegood DT, Fantus IG, Morris D. Androgen response to endogenous insulin secretion during the frequently sampled intravenous glucose tolerance test in normal and hyperandrogenic women.

J Clin Endocrinol Metab 71 6 —7. Vrbikova J, Hill M, Bendlova B, Grimmichova T, Dvorakova K, Vondra K, et al. Incretin levels in polycystic ovary syndrome. Eur J Endocrinol 2 —7. Stepto NK, Cassar S, Joham AE, Hutchison SK, Harrison CL, Goldstein RF, et al. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic-hyperinsulaemic clamp.

Hum Reprod 28 3 — Rosenfield RL, Ehrmann DA. The pathogenesis of polycystic ovary syndrome PCOS : The hypothesis of PCOS as functional ovarian hyperandrogenism revisited. Endocr Rev 37 5 — Clarembeau F, Bale G, Lanthier N. Cirrhosis and insulin resistance: current knowledge; pathophysiological mechanisms; complications and potential treatments.

Fu YH, Liu WJ, Lee CL, Wang JS. Associations of insulin resistance and insulin secretion with bone mineral density and osteoporosis in a general population.

Kobayashi H, Tokudome G, Hara Y, Sugano N, Endo S, Suetsugu Y, et al. Insulin resistance is a risk factor for the progression of chronic kidney disease. Clin Nephrol. Cree MG, Wolfe RR. Postburn trauma insulin resistance and fat metabolism.

Am J Physiol Endocrinol Metab 1 :E1—9. Nagpal M, De D, Handa S, Pal A, Sachdeva N. Insulin resistance and metabolic syndrome in young men with acne. JAMA Dermatol 4 — Hsu CS, Wang PC, Chen JH, Su WC, Tseng TC, Chen HD, et al. Increasing insulin resistance is associated with increased severity and prevalence of gastro-oesophageal reflux disease.

Aliment Pharmacol Ther 34 8 — Carmelli D, Cardon LR, Fabsitz R. Clustering of hypertension; diabetes; and obesity in adult male twins: same genes or same environments? Am J Hum Genet 55 3 — Lin HF, Boden-Albala B, Juo SH, Park N, Rundek T, Sacco RL. Heritabilities of the metabolic syndrome and its components in the northern manhattan family study.

Diabetologia 48 10 — Wan ZL, Huang K, Xu B, Hu SQ, Wang S, Chu YC, et al. Diabetes-associated mutations in human insulin: crystal structure and photo-cross-linking studies of a-chain variant insulin wakayama. Biochemistry 44 13 — Tager H, Given B, Baldwin D, Mako M, Markese J, Rubenstein A, et al.

A structurally abnormal insulin causing human diabetes. Nature —5. Taylor SI, Kadowaki T, Kadowaki H, Accili D, Cama A, McKeon C. Mutations in insulin-receptor gene in insulin-resistant patients. Diabetes Care 13 3 — Verdecchia F, Akcan N, Dastamani A, Morgan K, Semple RK, Shah P.

Unusual glycemic presentations in a child with a novel heterozygous intragenic INSR deletion. Horm Res Paediatr 93 6 — Brown AE, Walker M. Genetics of insulin resistance and the metabolic syndrome. Curr Cardiol Rep 18 8 Mercado MM, McLenithan JC, Silver KD, Shuldiner AR.

Genetics of insulin resistance. Curr Diabetes Rep 2 1 — Al-Beltagi M, Bediwy AS, Saeed NK. Insulin-resistance in paediatric age: Its magnitude and implications. World J Diabetes. Kuglin B, Kolb H, Greenbaum C, Maclaren NK, Lernmark A, Palmer JP.

The fourth international workshop on the standardisation of insulin autoantibody workshop.

Metabolic syndrome is a cluster andd conditions that occur together, increasing your risk of heart Metabolism and insulin sensitivity, stroke ssnsitivity type 2 diabetes. These Diabetic meal prep ideas include increased blood pressure, high blood sugar, excess body fat around the seensitivity, and abnormal cholesterol or Semsitivity levels. People who have metabolic syndrome typically have apple-shaped bodies, meaning they have larger waists and carry a lot of weight around their abdomens. It's thought that having a pear-shaped body that is, carrying more of your weight around your hips and having a narrower waist doesn't increase your risk of diabetes, heart disease and other complications of metabolic syndrome. Having just one of these conditions doesn't mean you have metabolic syndrome. But it does mean you have a greater risk of serious disease. And if you develop more of these conditions, your risk of complications, such as type 2 diabetes and heart disease, rises even higher.

Author: Dikree

1 thoughts on “Metabolism and insulin sensitivity

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com