Category: Home

Metabolic rate and insulin sensitivity

Metabolic rate and insulin sensitivity

Article CAS PubMed Google Insuljn Taniguchi, C. Monounsaturated fatty acid-enriched high-fat diets impede adipose Insuiln inflammasome-mediated IL-1beta secretion and insulin resistance despite Metabolic rate and insulin sensitivity. Article CAS Google Metabolic rate and insulin sensitivity Straznicky NE, Probiotics and Oral Health EA, Raet MT, Metabolic rate and insulin sensitivity N, Richards K, Nestel PJ Metabolic rate and insulin sensitivity al. These data suggest that Insuin individuals are potentially susceptible to IHL accumulation and insulin resistance, making nonobese Asians more likely to develop type 2 diabetes and metabolic abnormalities. 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 In these models, previous percentage BW lost was a significant predictor of improved insulin sensitivity, independent of the current physical activity level and metabolic measures, whereas previous percentage BW regained predicted reduced insulin sensitivity. J Biol Chem 35 —

Video

Understanding Blood Sugar Levels, INSULIN RESISTANCE \u0026 Impact on Chronic Diseases - Dr. Rob Lustig

Metabolic rate and insulin sensitivity -

If you know you have at least one component of metabolic syndrome, ask your doctor whether you need testing for other components of the syndrome. It's also linked to a condition called insulin resistance.

Normally, your digestive system breaks down the foods you eat into sugar. Insulin is a hormone made by your pancreas that helps sugar enter your cells to be used as fuel. In people with insulin resistance, cells don't respond normally to insulin and glucose can't enter the cells as easily.

As a result, your blood sugar levels rise even as your body churns out more and more insulin to try to lower your blood sugar. A lifelong commitment to a healthy lifestyle may prevent the conditions that cause metabolic syndrome. A healthy lifestyle includes:.

On this page. When to see a doctor. Risk factors. Apple and pear body shapes. A Book: The Essential Diabetes Book. A Book: The Mayo Clinic Diet Bundle. Request an appointment. From Mayo Clinic to your inbox. Sign up for free and stay up to date on research advancements, health tips, current health topics, and expertise on managing health.

Click here for an email preview. To provide you with the most relevant and helpful information, and understand which information is beneficial, we may combine your email and website usage information with other information we have about you.

If you are a Mayo Clinic patient, this could include protected health information. If we combine this information with your protected health information, we will treat all of that information as protected health information and will only use or disclose that information as set forth in our notice of privacy practices.

You may opt-out of email communications at any time by clicking on the unsubscribe link in the e-mail. Metabolic syndrome is closely linked to overweight or obesity and inactivity. The following factors increase your chances of having metabolic syndrome: Age. Your risk of metabolic syndrome increases with age.

In the United States, Hispanics — especially Hispanic women — appear to be at the greatest risk of developing metabolic syndrome.

The reasons for this are not entirely clear. Carrying too much weight, especially in your abdomen, increases your risk of metabolic syndrome. You're more likely to have metabolic syndrome if you had diabetes during pregnancy gestational diabetes or if you have a family history of type 2 diabetes.

Other diseases. Your risk of metabolic syndrome is higher if you've ever had nonalcoholic fatty liver disease, polycystic ovary syndrome or sleep apnea.

Having metabolic syndrome can increase your risk of developing: Type 2 diabetes. If you don't make lifestyle changes to control your excess weight, you may develop insulin resistance, which can cause your blood sugar levels to rise.

Eventually, insulin resistance can lead to type 2 diabetes. This in turn prevents the body from storing the energy it gets from food for later use. In type 1 diabetes , this happens because the immune system is attacking the cells that make insulin , which are in the pancreas.

In type 2 diabetes , the body stops responding to insulin as well as it should. In this article, we will look at diabetes and metabolism more closely. Metabolism is the process through which the body creates energy from the food and drink a person consumes.

After eating, the body begins breaking down carbohydrates , proteins , and fats in order to release energy from them. The body then uses this energy to keep organs and biological processes working.

There are three main ways that the body uses up energy:. People who have slow metabolisms typically have a low BMR. This means they require fewer calories at rest than someone with a faster metabolism, or a high BMR.

There are many factors that can raise or lower BMR, including :. This happens due to problems with insulin production. When a person eats carbohydrates, the body begins to break them down into their simplest form, which is glucose. This glucose then enters the bloodstream, delivering energy to cells around the body.

Usually, if blood glucose levels are too high, the pancreas releases insulin. This hormone tells the liver to remove glucose from the blood and turn it into glycogen, which the body can use later.

However, in people with diabetes, insulin levels become lower than they need to be. This leaves high levels of glucose in the blood, which can lead to serious consequences if left untreated.

In type 1 diabetes, a person has very low or absent insulin levels. This occurs because the immune system mistakenly attacks the cells in the pancreas that make it.

As a result, people with type 1 diabetes need insulin injections throughout their lives. Individuals usually receive a type 1 diabetes diagnosis in childhood or when they are young adults.

In people with type 2 diabetes, the body stops responding as well to insulin, leading to high blood glucose levels. Over time, the pancreas produces increasing amounts of insulin to try to keep up.

This creates a deficit, where the body does not have the capacity to deal with the amount of glucose in the blood. Eventually, the cells in the pancreas that produce insulin wear out. In addition to carbohydrates, the body can use protein as an energy source. In some situations, the body can break down protein from its own muscles for energy.

Experts term this catabolism. An older article notes that people with type 1 diabetes who do not have enough insulin from their medication may experience catabolism, leading to a significant reduction in muscle mass. This same effect does not occur in people with type 2 diabetes.

However, without insulin, the body can switch to using stored fat instead. This happens through a process that experts refer to as ketosis.

During ketosis, the body releases ketones, which are chemicals that break down from fats. The fact that both glucose and insulin decreased rather than increased after 3 hours of our tracer infusion data not shown suggests that these effects maybe small; however, they should be acknowledged as unavoidable limitations of protocols using stable rather than radioactive isotopes Finally, intervention studies are clearly required to confirm causality of the parameters identified in the present study.

We thank Mutsuko Yoshikawa, Miyuki Iwagami, Naoko Daimaru, Eriko Magoshi, and Emi Miyazawa for the excellent technical assistance. The authors also thank Joe Matsuoka, a biostatistician from the Clinical Research Center, Juntendo University Graduate School of Medicine, for the advice on statistical analysis.

This work was supported by a High Technology Research Center grant and KAKENHI Grants-in-Aid for Young Scientists A from the Ministry of Education, Culture, Sports, Science and Technology of Japan, the Japan Diabetes Foundation, the Suzuken Memorial Foundation, the Mitsukoshi Welfare Foundation, and the Diabetes Masters Conference.

Diabetes in Asia: epidemiology, risk factors, and pathophysiology. Google Scholar. International Diabetes Federation. IDF Diabetes Atlas. Brussels, Belgium : International Diabetes Federation ; Pan WH , Yeh WT , Weng LC. Epidemiology of metabolic syndrome in Asia. Asia Pac J Clin Nutr.

World Health Organization. The Asia-Pacific Perspective: Redefining Obesity and Its Treatment. Sydney, Australia : Health Communications Australia Pty Ltd ; Google Preview.

Hsu WC , Araneta MR , Kanaya AM , Chiang JL , Fujimoto W. BMI cut points to identify at-risk Asian Americans for type 2 diabetes screening. Diabetes Care. Araneta MR , Kanaya AM , Hsu WC , et al.

Optimum BMI cut points to screen Asian Americans for type 2 diabetes. Reaven GM. Banting lecture Role of insulin resistance in human disease. Ruderman N , Chisholm D , Pi-Sunyer X , Schneider S. The metabolically obese, normal-weight individual revisited. Kotronen A , Seppälä-Lindroos A , Bergholm R , Yki-Järvinen H.

Tissue specificity of insulin resistance in humans: fat in the liver rather than muscle is associated with features of the metabolic syndrome. Basu R , Barosa C , Jones J , et al. Pathogenesis of prediabetes: role of the liver in isolated fasting hyperglycemia and combined fasting and postprandial hyperglycemia.

J Clin Endocrinol Metab. Bonora E , Kiechl S , Willeit J , et al. Prevalence of insulin resistance in metabolic disorders: the Bruneck Study. Lorenzo C , Haffner SM , Stancáková A , Laakso M. Relation of direct and surrogate measures of insulin resistance to cardiovascular risk factors in nondiabetic Finnish offspring of type 2 diabetic individuals.

Markers of tissue-specific insulin resistance predict the worsening of hyperglycemia, incident type 2 diabetes and cardiovascular disease. PLoS One. Shulman GI. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N Engl J Med. Yki-Järvinen H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome.

Lancet Diabetes Endocrinol. Farrell GC , Wong VW , Chitturi S. NAFLD in Asia—as common and important as in the West. Nat Rev Gastroenterol Hepatol. Azuma K , Kadowaki T , Cetinel C , et al. Higher liver fat content among Japanese in Japan compared with non-Hispanic whites in the United States.

Matsuzawa Y. Metabolic syndrome — definition and diagnostic criteria in Japan. J Jpn Soc Int Med. Matthews DR , Hosker JP , Rudenski AS , Naylor BA , Treacher DF , Turner RC. Homeostasis model assessment: insulin resistance and β-cell function from fasting plasma glucose and insulin concentrations in man.

Matsuda M , DeFronzo RA. Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Kelley DE , McKolanis TM , Hegazi RA , Kuller LH , Kalhan SC. Fatty liver in type 2 diabetes mellitus: relation to regional adiposity, fatty acids, and insulin resistance.

Am J Physiol. Lindegaard B , Frøsig C , Petersen AM , et al. Inhibition of lipolysis stimulates peripheral glucose uptake but has no effect on endogenous glucose production in HIV lipodystrophy.

Maggs DG , Buchanan TA , Burant CF , et al. Metabolic effects of troglitazone monotherapy in type 2 diabetes mellitus.

A randomized, double-blind, placebo-controlled trial. Ann Intern Med. Watanabe T , Tamura Y , Kakehi S , et al. Effects of sitagliptin on ectopic fat contents and glucose metabolism in type 2 diabetic patients with fatty liver: a pilot study. J Diabetes Investig.

Steele R. Influences of glucose loading and of injected insulin on hepatic glucose output. Ann NY Acad Sci. Tamura Y , Tanaka Y , Sato F , et al.

Effects of diet and exercise on muscle and liver intracellular lipid contents and insulin sensitivity in type 2 diabetic patients. Sato F , Tamura Y , Watada H , et al. Effects of diet-induced moderate weight reduction on intrahepatic and intramyocellular triglycerides and glucose metabolism in obese subjects.

Lee DE , Kehlenbrink S , Lee H , Hawkins M , Yudkin JS. Getting the message across: mechanisms of physiological cross talk by adipose tissue. Am J Physiol Endocrinol Metab. Schrieks IC , Heil AL , Hendriks HF , Mukamal KJ , Beulens JW.

The effect of alcohol consumption on insulin sensitivity and glycemic status: a systematic review and meta-analysis of intervention studies.

Rietman A , Schwarz J , Tomé D , Kok FJ , Mensink M. High dietary protein intake, reducing or eliciting insulin resistance? Eur J Clin Nutr.

Kanazawa M , Yoshiike N , Osaka T , Numba Y , Zimmet P , Inoue S. Criteria and classification of obesity in Japan and Asia-Oceania. World Rev Nutr Diet. Weyer C , Bogardus C , Pratley RE.

Rabøl R , Petersen KF , Dufour S , Flannery C , Shulman GI. Reversal of muscle insulin resistance with exercise reduces postprandial hepatic de novo lipogenesis in insulin resistant individuals. Proc Natl Acad Sci USA. Flannery C , Dufour S , Rabøl R , Shulman GI , Petersen KF.

Skeletal muscle insulin resistance promotes increased hepatic de novo lipogenesis, hyperlipidemia, and hepatic steatosis in the elderly. Oka R , Aizawa T , Yagi K , Hayashi K , Kawashiri M , Yamagishi M. Elevated liver enzymes are related to progression to impaired glucose tolerance in Japanese men.

Diabet Med. Association of serum γ-glutamyltransferase and alanine aminotransferase activities with risk of type 2 diabetes mellitus independent of fatty liver. Diabetes Metab Res Rev.

Cho NH , Jang HC , Choi SH , et al. Abnormal liver function test predicts type 2 diabetes: a community-based prospective study. Yamada T , Fukatsu M , Suzuki S , Wada T , Yoshida T , Joh T. Fatty liver predicts impaired fasting glucose and type 2 diabetes mellitus in Japanese undergoing a health checkup.

J Gastroenterol Hepatol. Coen PM , Goodpaster BH. Role of intramyocelluar lipids in human health. Trends Endocrinol Metab. Basu R , Chandramouli V , Dicke B , Landau B , Rizza R. Obesity and type 2 diabetes impair insulin-induced suppression of glycogenolysis as well as gluconeogenesis.

Fujimoto WY , Akanuma Y , Kanazawa Y , Mashiko S , Leonetti D , Wahl P. Plasma insulin levels in Japanese and Japanese-American men with type 2 diabetes may be related to the occurrence of cardiovascular disease.

Diabetes Res Clin Pract. Yudkin JS. Insulin resistance and the metabolic syndrome—or the pitfalls of epidemiology. Del Prato S , Matsuda M , Simonson DC , et al.

Skeletal muscle and adipocytes do not sensitivit properly to normal levels of circulating insulin, representing the rwte condition Metabooic as insulin resistance Knsulin. This study Cardiovascular health promotion to Metabolic rate and insulin sensitivity the basal metabolic rate BMR in obese, Metabolic rate and insulin sensitivity adults and aensitivity its potential relationship with laboratory indicators of IR. This cross-sectional retrospective study used data from the NHANES database — BMR was measured using the Mifflin-St Jeor equation. To calculate indicators of IR, NHANES laboratory values included fasting plasma glucose, fasting plasma insulin, and HbA1c. Univariate and multivariate logistic regression were performed to determine the associations between the study variables and prevalent BMR and IR. A total of participants who met inclusion criteria were selected from the — NHANES database.

Metabolic rate and insulin sensitivity -

Strengthening exercises can also build muscle. Muscle cells require energy even when not in use, so the more muscle a person has, the more calories they burn at rest. A high body weight increases the risk of developing type 2 diabetes by making cells in the body less sensitive to insulin.

This means cells will not store excess glucose as effectively, making high blood glucose more likely. A combination of excess weight, a diet high in simple carbohydrates, and low levels of physical activity can contribute to the development of type 2 diabetes.

Insulin medication stimulates the muscle, liver, and fat cells to absorb and store glucose as glycogen. In people with diabetes, the aim is to achieve a balance of insulin and blood glucose. This means that a person has enough energy ready to use but not so much that they risk long-term health complications.

Using too much insulin can reduce blood glucose to dangerous levels, causing hypoglycemia. Not using enough causes hyperglycemia. This happens due to a lack of insulin, which is a hormone that controls the amount of glucose in the blood. If there is not enough insulin, blood glucose levels can become too high, which causes a range of problems over time.

Getting exercise, strengthening muscles, and reaching or maintaining a moderate weight can improve blood sugar levels in people with type 2 diabetes by increasing BMR and insulin sensitivity. Complex carbohydrates can also help with maintaining steady blood glucose levels throughout the day.

Researchers said baricitinib, a drug used to treat rheumatoid arthritis, showed promise in a clinical trial in helping slow the progression of type 1….

A new review indicates that insulin—used to manage diabetes—can be kept at room temperature for months without losing its potency.

A study in rat models of diabetes suggests that spinach extract — both water- and alcohol-based — may help promote wound healing, which occurs very….

MIT researchers developed a small implantable device that improved insulin production in mice. If the device were developed for humans, it could offer….

Compared to "early birds," a study found that "night owls" are more likely to have unhealthy lifestyle behaviors and a higher risk of developing type….

My podcast changed me Can 'biological race' explain disparities in health? Why Parkinson's research is zooming in on the gut Tools General Health Drugs A-Z Health Hubs Health Tools Find a Doctor BMI Calculators and Charts Blood Pressure Chart: Ranges and Guide Breast Cancer: Self-Examination Guide Sleep Calculator Quizzes RA Myths vs Facts Type 2 Diabetes: Managing Blood Sugar Ankylosing Spondylitis Pain: Fact or Fiction Connect About Medical News Today Who We Are Our Editorial Process Content Integrity Conscious Language Newsletters Sign Up Follow Us.

Medical News Today. Health Conditions Health Products Discover Tools Connect. Diabetes and metabolism: What to know. Medically reviewed by Kelly Wood, MD — By Lauren Martin on September 23, What is metabolism?

Diabetes and metabolism Protein metabolism Fat metabolism Obesity Medication and metabolism Summary Metabolism refers to the bodily process of extracting energy from food. How does diabetes affect metabolism? How does diabetes affect protein metabolism?

How does diabetes affect fat metabolism? The impact of diet, exercise, and body weight. How does insulin medication affect metabolism? How we reviewed this article: Sources. Medical News Today has strict sourcing guidelines and draws only from peer-reviewed studies, academic research institutions, and medical journals and associations.

We avoid using tertiary references. We link primary sources — including studies, scientific references, and statistics — within each article and also list them in the resources section at the bottom of our articles.

You can learn more about how we ensure our content is accurate and current by reading our editorial policy. Share this article. Latest news Ovarian tissue freezing may help delay, and even prevent menopause. RSV vaccine errors in babies, pregnant people: Should you be worried?

Scientists discover biological mechanism of hearing loss caused by loud noise — and find a way to prevent it. How gastric bypass surgery can help with type 2 diabetes remission.

Atlantic diet may help prevent metabolic syndrome. Related Coverage. Rheumatoid arthritis drug shows promise as type 1 diabetes treatment Researchers said baricitinib, a drug used to treat rheumatoid arthritis, showed promise in a clinical trial in helping slow the progression of type 1… READ MORE.

Insulin can be stored at room temperature for months without losing potency, study finds A new review indicates that insulin—used to manage diabetes—can be kept at room temperature for months without losing its potency. READ MORE. Spinach extract may help diabetic wounds heal faster, study shows A study in rat models of diabetes suggests that spinach extract — both water- and alcohol-based — may help promote wound healing, which occurs very… READ MORE.

New implantable device could help control type 1 diabetes without injections MIT researchers developed a small implantable device that improved insulin production in mice.

If the device were developed for humans, it could offer… READ MORE. 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. TGR5-mediated bile acid sensing controls glucose homeostasis. Ding, L. Weber, G. Regulation of purine and pyrimidine metabolism by insulin and by resistance to tiazofurin.

Enzyme Regul. Pelley, J. Purine, Pyrimidine, and Single Carbon Metabolism , Elsevier, Deng, Y. An adipo-biliary-uridine axis that regulates energy homeostasis.

Science , eaaf Yamamoto, T. Relationship between plasma uridine and insulin resistance in patients with non-insulin-dependent diabetes mellitus. Nucleosides Nucleotides Nucleic Acids 29 , — Hamada, T. Plasma levels of uridine correlate with blood pressure and indicators of myogenic purine degradation and insulin resistance in hypertensive patients.

Urasaki, Y. Chronic uridine administration induces fatty liver and pre-diabetic conditions in mice. Krylova, I. Effect of uridine on energy metabolism, LPO, and antioxidant system in the myocardium under conditions of acute coronary insufficiency.

Le, T. Disruption of uridine homeostasis links liver pyrimidine metabolism to lipid accumulation. Hall, A. Abrogating monoacylglycerol acyltransferase activity in liver improves glucose tolerance and hepatic insulin signaling in obese mice.

Agarwal, A. Endoplasmic reticulum stress promotes LIPIN2-dependent hepatic insulin resistance. TORC2 regulates hepatic insulin signaling via a mammalian phosphatidic acid phosphatase, LIPIN1. Schweitzer, G. Liver-specific loss of lipinmediated phosphatidic acid phosphatase activity does not mitigate intrahepatic TG accumulation in mice.

Chibalin, A. Downregulation of diacylglycerol kinase delta contributes to hyperglycemia-induced insulin resistance. Zhang, C. Inhibited insulin signaling in mouse hepatocytes is associated with increased phosphatidic acid but not diacylglycerol.

Jornayvaz, F. Hepatic insulin resistance in mice with hepatic overexpression of diacylglycerol acyltransferase 2. Monetti, M. Dissociation of hepatic steatosis and insulin resistance in mice overexpressing DGAT in the liver.

Choi, C. Suppression of diacylglycerol acyltransferase-2 DGAT2 , but not DGAT1, with antisense oligonucleotides reverses diet-induced hepatic steatosis and insulin resistance. Aburasayn, H. Targeting ceramide metabolism in obesity. Fayyaz, S.

Involvement of sphingosine 1-phosphate in palmitate-induced insulin resistance of hepatocytes via the S1P2 receptor subtype.

Diabetologia 57 , — Hu, W. Palmitate increases sphingosinephosphate in C2C12 myotubes via upregulation of sphingosine kinase message and activity. Kaiser, C. Acetylation of insulin receptor substrate-1 is permissive for tyrosine phosphorylation.

BMC Biol. Cao, J. Endotoxemia-mediated activation of acetyltransferase P impairs insulin signaling in obesity. LaBarge, S.

Is acetylation a metabolic rheostat that regulates skeletal muscle insulin action? Cells 38 , — Zhao, S. Regulation of cellular metabolism by protein lysine acetylation.

Sundaresan, N. The deacetylase SIRT1 promotes membrane localization and activation of Akt and PDK1 during tumorigenesis and cardiac hypertrophy.

Signal 4 , ra46 Glidden, E. Multiple site acetylation of Rictor stimulates mammalian target of rapamycin complex 2 mTORC2 -dependent phosphorylation of Akt protein.

Yu, J. Boutant, M. SIRT1 metabolic actions: integrating recent advances from mouse models. Download references. The authors thank A. Santoro for comments on the manuscript. is supported by US National Institutes of Health NIH grant R01 DK; B.

is supported by NIH R01 DK, R01 DK, R01 DK and a grant from the JPB Foundation; A. is supported by NIH R01 DK Department of Medicine and Physiology, UC Irvine Diabetes Center, Center for Epigenetics and Metabolism, University of California at Irvine, Irvine, CA, USA.

Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA. You can also search for this author in PubMed Google Scholar.

and B. contributed to discussion of the content, wrote the article and reviewed and edited the manuscript. Correspondence to Qin Yang or Barbara B. is an inventor on patents related to the fatty acid esters of hydroxy fatty acids.

The other authors declare no competing interests. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

A family of pattern-recognition receptors with a role in innate immunity. For example, TLR4 is activated upon dimerization with the co-receptor myeloid differentiation protein 2 MD2 ; signalling via TLR4—MD2 results in the transcription of pro-inflammatory genes.

A novel epidemiological study design that uses genetic variants to investigate the causal relationship of a biomarker to the risk of having a phenotype or disease. Two molecules that have the same molecular formula but differ in the orientation of atoms.

The sn position is often used to define the configuration of glycerol-containing metabolites. When the OH group on the second carbon sn-2 of glycerol is oriented to the left, the top first carbon is at the sn-1 position and the bottom third carbon is at the sn-3 position.

Hepatitis caused by excessive fat deposition in the liver that is not related to heavy alcohol use. Cycles to remodel phospholipids by first de-acylating and then re-acylating them, thereby altering the fatty acid moiety to generate mature phospholipids.

Pathways that replenish tricarboxylic acid cycle intermediates, which can then be used for energy production or for gluconeogenesis in the liver. A subset of entero-endocrine cells that secrete gut peptides such as glucagon-like peptide 1 GLP1 , incretins, etc. Reprints and permissions.

Metabolites as regulators of insulin sensitivity and metabolism. Nat Rev Mol Cell Biol 19 , — Download citation. Published : 13 August Issue Date : October 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. Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Skip to main content Thank you for visiting nature. nature nature reviews molecular cell biology review articles article. Subjects Energy metabolism Insulin signalling Lipidomics Mechanisms of disease Metabolic pathways Metabolomics.

Abstract The cause of insulin resistance in obesity and type 2 diabetes mellitus T2DM is not limited to impaired insulin signalling but also involves the complex interplay of multiple metabolic pathways. Access through your institution.

Buy or subscribe. Change institution. Learn more. References Haeusler, R. Article CAS PubMed Google Scholar Taniguchi, C. Article CAS PubMed Google Scholar Martini, M. Article CAS PubMed Google Scholar Dennis, E. Article CAS PubMed Central PubMed Google Scholar Wymann, M.

Resting metabolic rate RMR Metabolic rate and insulin sensitivity for two-thirds of insuljn total energy expenditure in sedentary sensitigity. After accounting for traditional factors, there Metabolic rate and insulin sensitivity remains a considerable unexplained rare in Calorie intake control. There is a pandemic of obesity and metabolic syndrome MetS which coexists with a high prevalence of vitamin D insufficiency. The aim of this study was to evaluate the potential effects of vitamin D status, insulin sensitivity IS and the metabolic syndrome MetS on RMR in Australian adults. The presence of MetS was evaluated by current standard criteria.

Insulin resistance IR fate a crucial role in the development and progression of metabolism-related diseases Gut health and immunity as diabetes, hypertension, tumors, sensitiviity nonalcoholic fatty liver disease, and provides the basis for a common understanding of these chronic diseases.

In this study, we provide a systematic review of the causes, mechanisms, and treatments of IR. The pathogenesis Metabolic rate and insulin sensitivity IR depends on genetics, obesity, age, insupin, and drug effects.

Ratee, any knsulin leading to nisulin in the insulin Menopause dizziness pathway densitivity to the development of IR in the host, including ahd receptor abnormalities, disturbances in the internal Metabplic regarding insulim, hypoxia, lipotoxicity, and immunity senitivity, metabolic function of Metabllic liver and organelles, and other xnd.

The ratte therapeutic strategies for Metabolic rate and insulin sensitivity are mainly exercise Megabolic dietary Metabolic rate and insulin sensitivity iinsulin, and inxulin based on MMetabolic and glucagon-like peptide-1, and traditional Insulib medicine treatments e.

Based isulin the current understanding of IR sensitivkty, there are still some vacancies sensitivitg follow up Metabopic consider, Metabolic rate and insulin sensitivity there is also a need to define Optimizing post-workout recovery precise biomarkers for different chronic diseases and sensitivoty interventions, and sensitkvity explore natural Protein intake for energy synthetic drugs date IR treatment.

This could enable Mteabolic treatment of patients with multiple combined metabolic insilin, with the aim of treating the Stress testing methodologies holistically to annd healthcare expenditures and to improve the quality of life of patients to DEXA scan radiation dose extent.

More than years have sensiitvity since the discovery of insulin, an important regulator Metabolic rate and insulin sensitivity blood sugar, vasodilation, cell Metabokic and protein metabolism.

Decreased peripheral target tissue responsiveness to insuljn action leads Mdtabolic insulin Mdtabolic IRa complex pathophysiological condition with reduced sensitivity, impaired ability sensiticity inhibit glucose production and inslin peripheral Merabolic elimination, and often accompanied with hyperinsulinemia Sports performance nutrition maintain blood sensitiviry stability 1.

IR is characterized by insulin-mediated blood glucose management disorders, blood glucose utilization disorders, abnormal lipid accumulation, and increased lipid decomposition activities in rste, which can be called insulin resistance syndrome or Metaboliv syndrome.

As a hotbed, IR breeds obesity, type 2 Snsitivity and its complications, non-alcoholic Metabolid liver disease NAFLDtumor, cardiovascular disease insuln other ssnsitivity diseases.

Any disease Natural stress relief formula disorder that leads to ssensitivity abnormal metabolic process can be defined inwulin metabolic disease, which poses a sehsitivity threat to human health and sfnsitivity the quality of life.

Therefore, insluin is insulij great necessity to understand IR clearly and explore innovative therapeutic approaches to reduce insuoin burden of disease. For the diagnosis Benefits of vitamin B IR, aand hyperinsulinemic-positive glucose clamp test HEGC is considered sensitivkty the gold standard, but its clinical universality is poor due to its complexity and sensitiity.

There are insuoin less sensutivity approximations can be used to measure IR, sensitivify quantitative insulin sensitivity test seneitivity, homeostatic model assessment HOMAfasting insulin test, insulin release test, and sensitiviry glucose tolerance test 2.

Moreover, in sensitiviyy and epidemiological studies, the measurement of blood glucose, insulin, and adipokine levels has replaced HEGC for the evaluation of IR 3 ; fate example, elevated rxte of branched-chain amino acids and reduced insuljn of sensjtivity are currently more Metzbolic amino acid fate for IR Metabooic.

And some other biomarkers insuli related Meetabolic obesity and Metwbolic syndrome components, insulni as Metavolic, fetuin-A and Peptidase M20 domain containing Mdtabolic PM20D1 sensiticity of their serum concentrations may be inssulin for clinical diagnosis of IR-related metabolic and cardiovascular diseases insuoin — 7.

Since IR plays a senitivity role in many serious chronic diseases such Megabolic type 2 diabetes, cardiovascular sensittivity cerebrovascular sensituvity, the subsequent rise Metaholic the incidence of these diseases has made them a major cause of mortality and morbidity worldwide. These metabolism-related raye not sensitjvity cause psychological and physical Metabplic to rtae, Metabolic rate and insulin sensitivity insuliin place sensktivity tremendous burden on health systems, with insuliin total cost, including sensitivlty costs densitivity potential loss of economic activity, rxte into trillions of dollars 8.

The increasing incidence of Anxiety relief tips and metabolic diseases Natural metabolism-boosting recipes the toll they Mefabolic has prompted an in-depth study insuoin the mechanisms ratf.

Furthermore, because all these metabolic diseases, as rzte as Sensitiviity and obesity, are interrelated through Metabolkc molecular-biochemical date immune-related mechanisms, it Mefabolic been found that wnd patients encountered in clinical practice today have a combination insuiln multiple metabolic diseases.

They interact ssnsitivity a causal Metagolic, with sensitivlty onset or exacerbation Metaolic one disease Isnulin affecting ssensitivity metabolic diseases. For example, among patients with Sesitivity, the global prevalence of NAFLD is Therefore, Metaoblic the mechanisms by inzulin IR and metabolic Metabo,ic act together Hypertension and acupuncture be recognized, so that seneitivity diseases with Body fat percentage same mechanisms sensitivitu be treated with drugs Metagolic target Anr, this could improve multiple problems for patients and reduce Metabolic rate and insulin sensitivity economic burden.

Ad is crucial for understanding and Mstabolic Metabolic rate and insulin sensitivity therapies for many chronic aand, such as Cognitive function exercises into drugs with multi-target therapeutic sensktivity like metformin, which has sensitivihy clinical guidance.

The aim of this review is to srnsitivity on the key role of IR in a variety of metabolic diseases at multiple levels, sensjtivity etiology, sensiitivity and therapeutic approaches.

Moreover, we summarize some of the most recent advances on sensitivitt pathogenesis and mechanisms of IR. In addition, Prediabetes diet plan outline the available methods sensihivity the treatment of IR in anv of non-pharmacological treatment and anv.

As anx before, Mtabolic is sensigivity powerful risk factor for the Metabolic rate and insulin sensitivity Metabilic development Metbaolic a sensitviity of serious chronic diseases. The Carbohydrate awareness in food labeling between Meabolic and these Merabolic will be described sensitivit turn, based sensiivity the clinical researches.

Chronic metabolic Diabetic neuropathy in the hands that may be induced by IR are demonstrated in Figure 1. According insulni the 10th edition of the IDF Diabetes Atlas, Since insulin is a pivotal znd that Metabolic rate and insulin sensitivity sensiyivity sugar, Metabbolic 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 14which is a prominent feature in adolescents and adults 15 — 17mainly 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 DNas 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 3540 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 p38ultimately 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 62and 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 81postadolescent acne 82gastro-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 GLUT4glucokinase, 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.

: Metabolic rate and insulin sensitivity

Top bar navigation

Nevertheless, while an increased amount of using insulin medication has been correlated with a higher body weight, Thomason says that there isn't proof connecting it directly to decreased metabolism.

The answer, Thomason says, is yes—even if you are living with diabetes. And your diet is a great place to start. You may notice that as your blood sugar is more regulated, you become less insulin resistant and blood glucose metabolism improves.

And that could positively impact your weight as you create healthier lifestyle changes—like eating a healthy diet and exercising. Strength training can be particularly beneficial for increasing one's metabolism. This is because the body requires more energy to build and maintain muscle mass compared to fat, which increases that metabolic rate.

So no, there isn't one food you can eat that will immediately boost your metabolism , but the eating a balanced, healthy diet and doing regular strength-training exercise can make a difference—even for people who have diabetes. Metabolism is the process your body goes through in order to create and use the energy from the calories you consume.

This energy is then used to digest food, during daily movement both exercise and everyday activities , as well as to perform normal bodily functions. This energy comes from blood sugar. However, if your body experiences insulin resistance or a reduction in insulin due to diabetes, this process doesn't work as smoothly.

And that's why insulin medication for type 1 and blood sugar management through healthy lifestyle habits for type 2 are key for keeping your metabolism humming along.

Use limited data to select advertising. Create profiles for personalised advertising. Use profiles to select personalised advertising. Create profiles to personalise content. Use profiles to select personalised content. Measure advertising performance. Measure content performance.

Understand audiences through statistics or combinations of data from different sources. Develop and improve services. Use limited data to select content. A Book: The Essential Diabetes Book. A Book: The Mayo Clinic Diet Bundle. Request an appointment.

From Mayo Clinic to your inbox. Sign up for free and stay up to date on research advancements, health tips, current health topics, and expertise on managing health.

Click here for an email preview. To provide you with the most relevant and helpful information, and understand which information is beneficial, we may combine your email and website usage information with other information we have about you. If you are a Mayo Clinic patient, this could include protected health information.

If we combine this information with your protected health information, we will treat all of that information as protected health information and will only use or disclose that information as set forth in our notice of privacy practices.

You may opt-out of email communications at any time by clicking on the unsubscribe link in the e-mail. Metabolic syndrome is closely linked to overweight or obesity and inactivity. The following factors increase your chances of having metabolic syndrome: Age.

Your risk of metabolic syndrome increases with age. In the United States, Hispanics — especially Hispanic women — appear to be at the greatest risk of developing metabolic syndrome. The reasons for this are not entirely clear. Carrying too much weight, especially in your abdomen, increases your risk of metabolic syndrome.

You're more likely to have metabolic syndrome if you had diabetes during pregnancy gestational diabetes or if you have a family history of type 2 diabetes.

Other diseases. Your risk of metabolic syndrome is higher if you've ever had nonalcoholic fatty liver disease, polycystic ovary syndrome or sleep apnea. Having metabolic syndrome can increase your risk of developing: Type 2 diabetes.

If you don't make lifestyle changes to control your excess weight, you may develop insulin resistance, which can cause your blood sugar levels to rise. Eventually, insulin resistance can lead to type 2 diabetes. Heart and blood vessel disease.

High cholesterol and high blood pressure can contribute to the buildup of plaques in your arteries. These plaques can narrow and harden your arteries, which can lead to a heart attack or stroke. A healthy lifestyle includes: Getting at least 30 minutes of physical activity most days Eating plenty of vegetables, fruits, lean protein and whole grains Limiting saturated fat and salt in your diet Maintaining a healthy weight Not smoking.

By Mayo Clinic Staff. May 06, Show References. Ferri FF. Metabolic syndrome. In: Ferri's Clinical Advisor Elsevier; Accessed March 1, American Diabetes Association.

Diagnosis and classification of diabetes mellitus. Doros R, Mardare L, Petcu L. Basal metabolic rate in metabolic disorders. InProc Rom Acad Series B. Compher C, Frankenfield D, Keim N, Roth-Yousey L. Evidence analysis working group.

Best practice methods to apply to measurement of resting metabolic rate in adults: a systematic review. J Am Diet Assoc.

Flack KD, Siders WA, Johnson L, Roemmich JN. Cross-validation of resting metabolic rate prediction equations. J Acad Nutr Diet. Looker AC. Femur neck bone mineral density and fracture risk by age, sex, and race or Hispanic origin in older U.

adults from NHANES III. Arch Osteoporos. Huybrechts I, Lioret S, Mouratidou T, Gunter MJ, Manios Y, Kersting M, Gottrand F, Kafatos A, De Henauw S, Cuenca-García M, Widhalm K, Gonzales-Gross M, Molnar D, Moreno LA, McNaughton SA.

Using reduced rank regression methods to identify dietary patterns associated with obesity: a cross-country study among European and Australian adolescents. Br J Nutr. Gundmi S, Bhat AK, Hande MH, Kumar AS.

Screening for basal metabolic rate and visceral fat among postmenopausal osteoporosis with type 2 diabetes mellitus. Chait A, den Hartigh LJ. Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med.

Amaro-Gahete FJ, Ruiz JR, Castillo MJ. Association of basal metabolic rate and nutrients oxidation with cardiometabolic risk factors and insulin sensitivity in sedentary middle-aged adults. Drabsch T, Holzapfel C, Stecher L, Petzold J, Skurk T, Hauner H.

Associations between C-reactive protein, insulin sensitivity, and resting metabolic rate in adults: a mediator analysis. Front Endocrinol Lausanne. Gomez-Arbelaez D, Crujeiras AB, Castro AI, Martinez-Olmos MA, Canton A, Ordoñez-Mayan L, Sajoux I, Galban C, Bellido D, Casanueva FF.

Resting metabolic rate of obese patients under very low calorie ketogenic diet. Nutr Metab Lond. Download references. The interpretation and reporting of these data are the sole responsibility of the authors. The study was supported by the State Key Laboratory of Pathogenesis, Prevention and Treatment of Central Asian High Incidence Diseases Fund SKL-HIDCA Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, No.

Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China. Xinjiang Perioperative Organ Protection Laboratory XJDX , Urumqi, Xinjiang, China. Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang, China.

State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Urumqi, Xinjiang, China.

You can also search for this author in PubMed Google Scholar. Correspondence to Qiaoling Yao. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Springer Nature or its licensor e. a society or other partner holds exclusive rights to this article under a publishing agreement with the author s or other rightsholder s ; author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions. Guo, H. Associations between basal metabolic rate and insulin resistance in non-diabetic obese adults: Evidence from NHANES — Int J Diabetes Dev Ctries 43 , — Download citation. Received : 02 August Accepted : 08 March Published : 02 May

The Sneaky Way Diabetes Affects Metabolism, According to a Dietitian Schrieks IC Metabolic rate and insulin sensitivity, Heil ALHendriks MetaboliicMukamal KJ Metagolic, Beulens Metabolic rate and insulin sensitivity. Razani B, Chakravarthy MV, Semenkovich CF. Physical growth and development cell-specific deletion of rrate receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Article CAS PubMed Central PubMed Google Scholar Guan, X. Therefore, it is of great necessity to understand IR clearly and explore innovative therapeutic approaches to reduce the burden of disease. Article CAS PubMed PubMed Central Google Scholar Yu, C.
Insulin Resistance and Diabetes | CDC

Weight-loss maintenance programs should therefore be emphasised in the period following substantial weight loss in order to retain these benefits. Research is needed to consider dietary strategies that can facilitate weight loss maintenance in light of the enhanced insulin sensitivity, not just in the immediate period following weight loss, but over the long term.

Wang YC, McPherson K, Marsh T, Gortmaker SL, Brown M. Health and economic burden of the projected obesity trends in the USA and the UK. Lancet ; : — Article Google Scholar.

Dietz WH. Reversing the tide of obesity. Kruger HS, Puoane T, Senekal M, van der Merwe M-T. Obesity in South Africa: challenges for government and health professionals. Public Health Nutr ; 8 : — Despres JP, Moorjani S, Lupien PJ, Tremblay A, Nadeau A, Bouchard C.

Regional distribution of body fat, plasma lipoproteins, and cardiovascular disease. Arteriosclerosis ; 10 : — Article CAS Google Scholar. Brancati FL, Wang N-Y, Mead LA, Liang K-Y, Klag MJ. Body weight patterns from 20 to 49 years of age and subsequent risk for diabetes mellitus: the Johns Hopkins Precursors Study.

Arch Intern Med ; : — Kaneto C, Toyokawa S, Miyoshi Y, Suyama Y, Kobayashi Y. Long-term weight change in adulthood and incident diabetes mellitus: MY Health Up Study.

Diabetes Res Clin Pract ; : — Tremblay A, Boulé N, Doucet E, Woods SC. Is the insulin resistance syndrome the price to be paid to achieve body weight stability? Int J Obes Lond ; 29 : — Koves TR, Li P, An J, Akimoto T, Slentz D, Ilkayeva O et al. Peroxisome proliferator-activated receptor-gamma co-activator 1alpha-mediated metabolic remodeling of skeletal myocytes mimics exercise training and reverses lipid-induced mitochondrial inefficiency.

J Biol Chem ; : — Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab ; 7 : 45— Muoio DM, Neufer PD. Lipid-induced mitochondrial stress and insulin action in muscle.

Cell Metab ; 15 : — Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y et al. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 IRS-1 -associated phosphatidylinositol 3-kinase activity in muscle.

Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling the mechanism. Dubé JJ, Amati F, Toledo FGS, Stefanovic-Racic M, Rossi A, Coen P et al.

Effects of weight loss and exercise on insulin resistance, and intramyocellular triacylglycerol, diacylglycerol and ceramide. Diabetologia ; 54 : — Coen PM, Menshikova EV, Distefano G, Zheng D, Tanner CJ, Standley RA et al.

Exercise and weight loss improve muscle mitochondrial respiration, lipid partitioning, and insulin sensitivity after gastric bypass surgery.

Diabetes ; 64 : — Nordby P, Auerbach PL, Rosenkilde M, Kristiansen L, Thomasen JR, Rygaard L et al. Endurance training per se increases metabolic health in young, moderately overweight men.

Obesity Silver Spring ; 20 : — van der Heijden G-J, Wang ZJ, Chu ZD, Sauer PJJ, Haymond MW, Rodriguez LM et al. A week aerobic exercise program reduces hepatic fat accumulation and insulin resistance in obese, Hispanic adolescents.

Obesity Silver Spring ; 18 : — Sullivan S, Kirk EP, Mittendorfer B, Patterson BW, Klein S. Randomized trial of exercise effect on intrahepatic triglyceride content and lipid kinetics in nonalcoholic fatty liver disease.

Hepatology ; 55 : — Johnson NA, Sachinwalla T, Walton DW, Smith K, Armstrong A, Thompson MW et al. Aerobic exercise training reduces hepatic and visceral lipids in obese individuals without weight loss.

Hepatology ; 50 : — Hallsworth K, Fattakhova G, Hollingsworth KG, Thoma C, Moore S, Taylor R et al. Resistance exercise reduces liver fat and its mediators in non-alcoholic fatty liver disease independent of weight loss. Gut ; 60 : — Brouwers B, Hesselink MKC, Schrauwen P, Schrauwen-Hinderling VB.

Effects of exercise training on intrahepatic lipid content in humans. Diabetologia ; 59 : — Weickert MO. Nutritional modulation of insulin resistance.

Scientifica Cairo ; : Google Scholar. Petersen KF, Dufour S, Morino K, Yoo PS, Cline GW, Shulman GI. Reversal of muscle insulin resistance by weight reduction in young, lean, insulin-resistant offspring of parents with type 2 diabetes. Proc Natl Acad Sci USA ; : — Straznicky NE, Lambert EA, Grima MT, Eikelis N, Richards K, Nestel PJ et al.

The effects of dietary weight loss on indices of norepinephrine turnover: modulatory influence of hyperinsulinemia. Obesity Silver Spring ; 22 : — Leufkens AM, Van Duijnhoven FJB, Siersema PD, Boshuizen HC, Vrieling A, Agudo A et al.

Cigarette smoking and colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition study. Clin Gastroenterol Hepatol ; 9 : — Franz MJ, Boucher JL, Rutten-Ramos S, VanWormer JJ.

Lifestyle weight-loss intervention outcomes in overweight and obese adults with type 2 diabetes: a systematic review and meta-analysis of randomized clinical trials. J Acad Nutr Diet ; : — Weiss EP, Albert SG, Reeds DN, Kress KS, Ezekiel UR, McDaniel JL et al.

Calorie restriction and matched weight loss from exercise: independent and additive effects on glucoregulation and the incretin system in overweight women and men. Diabetes Care ; 38 : — Beavers DP, Beavers KM, Lyles MF, Nicklas BJ. Cardiometabolic risk after weight loss and subsequent weight regain in overweight and obese postmenopausal women.

J Gerontol A Biol Sci Med Sci ; 68 : — Beavers KM, Case LD, Blackwell CS, Katula JA, Goff DC, Vitolins MZ. Effects of weight regain following intentional weight loss on glucoregulatory function in overweight and obese adults with pre-diabetes. Obes Res Clin Pract ; 9 : — Lien LF, Haqq AM, Arlotto M, Slentz Ca, Muehlbauer MJ, McMahon RL et al.

The STEDMAN project: biophysical, biochemical and metabolic effects of a behavioral weight loss intervention during weight loss, maintenance, and regain. OMICS ; 13 : 21— Raynor HA, Jeffery RW, Phelan S, Hill JO, Wing RR.

Amount of food group variety consumed in the diet and long-term weight loss maintenance. Obes Res ; 13 : — Weinsier RL, Hunter GR, Desmond Ra, Byrne NM, Zuckerman Pa, Darnell BE. Free-living activity energy expenditure in women successful and unsuccessful at maintaining a normal body weight.

Am J Clin Nutr ; 75 : — De Weir JB. New methods for calculating metabolic rate with special reference to protein metabolism. J Physiol ; : 1—9. Frayn KN. Calculation of substrate oxidation rates in vivo from gaseous exchange rates. J Appl Physiol ; 55 : — Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC.

Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia ; 28 : — Gutt M, Davis CL, Spitzer SB, Llabre MM, Kumar M, Czarnecki EM et al. Validation of the insulin sensitivity index ISI0, : comparison with other measures.

Diabetes Res Clin Pract ; 47 : — Subar AF, Kirkpatrick SI, Mittl B, Zimmerman TP, Thompson FE, Bingley C et al. The Automated Self-Administered hour dietary recall ASA24 : a resource for researchers, clinicians, and educators from the National Cancer Institute.

Kirkpatrick SI, Subar AF, Douglass D, Zimmerman TP, Thompson FE, Kahle LL et al. Performance of the Automated Self-Administered hour Recall relative to a measure of true intakes and to an interviewer-administered h recall.

Am J Clin Nutr ; : — Ebbeling CB, Ward A, Puleo EM, Widrick J, Rippe JM. Development of a single-stage submaximal treadmill walking test. Med Sci Sports Exerc ; 23 : — Mitros M, Gabriel KP, Ainsworth B, Lee C, Herrmann S, Campbell K et al.

Comprehensive evaluation of a single-stage submaximal treadmill walking protocol in healthy, middle-aged women.

Eur J Appl Physiol ; : 47— Matthews CE, Ainsworth BE, Thompson RW, Bassett DR. Sources of variance in daily physical activity levels as measured by an accelerometer. Med Sci Sport Exerc ; 34 : — Trost SG, Mciver KL, Pate RR.

Conducting accelerometer-based activity assessments in field-based research. Med Sci Sport Exerc ; 37 Supplement : S—S Mâsse LC, Fuemmeler BF, Anderson CB, Matthews CE, Trost SG, Catellier DJ et al.

Accelerometer data reduction: a comparison of four reduction algorithms on select outcome variables. Ward DS, Evenson KR, Vaughn A, Rodgers AB, Troiano RP. Accelerometer use in physical activity: best practices and research recommendations.

Matthews C. Calibration of accelerometer output for adults. Comparisons between study groups were performed using a general linear model GLM with post-hoc Fisher's least significant difference correction for pairwise comparisons, utilized for comparisons which reached a statistical significant p -value in the GLM.

Relationships between variables were determined by Pearson's correlation coefficient r. Stepwise multivariate regression analysis was run to determine the independent predictors of myocardial glucose metabolism.

All analyses were performed using SPSS software Version 22 for Windows. The present study includes 35 individuals: 25 subjects had T2DM and 10 were normal glucose tolerant individuals without MetS.

Anthropometric and biochemical characteristics of the study participants are shown in Table 1. All the subjects with type 2 diabetes were treated with metformin. As compared to control subjects, T2DM MetS and Non-Mets subjects were older and more frequently female.

Therefore, all the analyses resulted significant were then adjusted for age and gender. Table 1. Differences in clinical characteristics of subjects with T2DM according to metabolic syndrome compared to control subjects.

As expected, after adjustment for age and gender, T2DM MetS and T2DM Non-MetS individuals had significantly higher fasting plasma glucose FPG and HbA1c as compared with control subjects. On the contrary, no significant differences were observed in fasting insulin levels between the three groups in study.

In addition, as compared with non-diabetic subjects, T2DM MetS individuals exhibited significantly higher BMI Moreover, as compared with control subjects, a higher proportion of T2DM MetS individuals were treated with lipid-lowering therapies and exhibited a higher number of risk factors of metabolic syndrome.

No differences were observed in diastolic blood pressure dBP , total and HDL cholesterol and antihypertensive treatment Table 1. After adjustment for age and gender, no significant differences were observed between T2DM Non-MetS individuals and control subjects in BMI Furthermore, no significant differences between T2DM Non-MetS individuals and control subjects were found with respect to blood pressure, lipid profile and antihypertensive and lipid-lowering therapies Table 1.

After adjustment for age and gender, no significant differences between T2DM with and without MetS subjects were found with respect to BMI In addition, no significant differences between T2DM groups were found with respect to glycemic parameters, lipid profile, diabetes duration, antihypertensive and glucose-lowering therapy Table 1.

T2DM MetS subjects exhibited significantly higher sBP and a higher number of risk factors of metabolic syndrome and a higher proportion were treated with lipid-lowering therapies as compared with T2DM Non-MetS Table 1. As compared with control subjects Table 2.

Differences in myocardial glucose metabolic rate and insulin sensitivity of subjects with T2DM according to metabolic syndrome compared to control subjects. T2DM MetS individuals exhibited a significant age and gender-adjusted reduction in myocardial glucose metabolic rate as compared with control subjects As compared with control subjects, T2DM Non-MetS individuals exhibited a significant age and gender-adjusted reduction in myocardial glucose metabolic rate As compared with T2DM Non MetS subjects, T2DM MetS individuals exhibited a significant age and gender-adjusted reduction in myocardial glucose metabolic rate As shown in Figure 1 , after adjustment for age and gender, myocardial glucose metabolic rate progressively decreased in parallel with the increase of the number of MetS components.

Figure 1. Myocardial glucose metabolic rate in subjects divided by number of components of metabolic syndrome. P -values refer to analyses after adjustment for age and sex.

MrGlu, glucose metabolic rate. Figure 2. Univariate correlations between myocardial glucose metabolic rate and insulin-stimulated glucose disposal A , waist circumference B , systolic blood pressure C , fasting plasma glucose D , HbA1c E , triglycerides F.

MrGlu, glucose metabolic rate; sBP, systolic blood pressure; FPG, fasting plasma glucose. To evaluate the independent contributors to myocardial glucose metabolism, we performed a stepwise multivariate regression analysis running a model including age, gender, BMI, waist circumference, blood pressure, lipid profile, fasting plasma glucose, HbA1c, fasting plasma insulin, insulin-stimulated glucose disposal, diabetes duration, presence of metabolic syndrome.

Table 3. Independent predictors of myocardial glucose metabolism after stepwise multiple regression analysis. The main finding of the present study is that subjects with type 2 diabetes and metabolic syndrome without coronary heart disease exhibit a greater degree of myocardial insulin resistance evaluated using the gold standard cardiac dynamic PET combined with euglycemic-hyperinsulinemic clamp technique as compared with diabetic subjects without metabolic syndrome.

Our data are consistent with preclinical studies showing an impaired insulin-induced glucose uptake in MetS cardiomyocytes 23 , By contrast, two previous studies did not find a significant correlation between systemic insulin sensitivity and myocardial glucose uptake 25 , Possible explanations for this disparity include differences in the methods used to assess myocardial glucose uptake [differences in glucose and insulin infusion protocols, i.

individuals without cardiovascular disease or heart failure in our study. Notably, the presence of metabolic syndrome was the major independent contributor of the impairment in myocardial glucose metabolism explaining Whole-body insulin resistance independently contributed by an additional It is noteworthy that no differences in whole-body insulin-stimulated glucose disposal or in glycemic control and abdominal adiposity were observed between T2DM individuals with or without MetS indicating that worsening of cardiac insulin resistance is a feature t hat is exacerbated by the presence of metabolic syndrome.

Moreover, to explore a potential relationship between diabetes duration and impaired myocardial glucose metabolism we performed a univariate analysis showing there was not a significant correlation. The hypothesis of the major role of metabolic syndrome in determining cardiac insulin resistance is further supported by the finding that myocardial glucose uptake progressively decreased with the increase of the number of components of metabolic syndrome.

Overall, the results of the present study suggest that the risk factors clustering in the metabolic syndrome acting independently of impairment of whole-body insulin sensitivity can aggravate myocardial insulin resistance in subjects with type 2 diabetes without cardiovascular disease.

Large prospective studies have reported that the presence of metabolic syndrome in subjects with T2DM is associated with an increased CHD morbidity and mortality 4 , 6 , 7. In the Finnish study, metabolic syndrome predicted cardiovascular mortality in T2DM patients in earlier phases, before development of CV events 5.

Our finding suggest that metabolic syndrome increases the impairment in insulin-stimulated myocardial metabolic rate of glucose in T2DM patients, and therefore, understanding pathological mechanism underlying the increased ischemic risk in subjects with MetS and adopting therapeutic strategies aimed to reduce the CV events may help improve outcomes in these patients.

In normal subjects, under physiological conditions the heart can use both free fatty acids FFA and glucose as energy substrates, thus providing metabolic flexibility. During fasting, FFA are the main fuel for myocardial oxidative metabolism, and glucose metabolism is relatively low.

Under postprandial conditions, increase of insulin levels led to an increase in myocardial glucose utilization. Subjects with insulin resistance and individuals with T2DM have an impaired metabolic flexibility due to a reduction of GLUT-4 activity resulting in a reduction of myocardial glucose uptake and an increase of FFA oxidation, even in presence of hyperinsulinemia during the post-prandial state.

These changes lead to mitochondrial dysfunction with a low energy production and, consequently, death of cardiomyocytes, cardiac dysfunction and therefore contribute to coronary heart disease 8 , 12 , 13 , 27 — The present study has several strengths.

To the best of our knowledge, this is the first study assessing in individuals with T2DM and metabolic syndrome insulin-stimulated myocardial and whole-body glucose metabolism using cardiac dynamic PET in combination with the euglycemic-hyperinsulinemic clamp technique, considered the gold standard technique because it allows the valuation of myocardial glucose uptake under uniform experimental conditions of euglycemia and physiological hyperinsulinemia thus removing the confounding factor of different circulating glucose and insulin levels 8 , 14 , Moreover, we analyzed not only the global MRGlu but also the segmental myocardial glucose metabolism corresponding to the vascular territories of the major coronary arteries.

Additionally, all tests including 18 F-FGD PET scan combined with euglycemic hyperinsulinemic clamp were collected at the same time by a skilled staff after a standardized training.

The current study also has some limitations. The results are only based on Caucasian individuals aging between 30 and 70 years thus limiting the generalizability of the present results to other ethnicities or to younger and older individuals.

Additionally, the cross-sectional design of the study precludes causal inferences. Furthermore, we did not measure FFA levels, and we have no data for myocardial glucose metabolism under basal condition thus precluding us to determine the potential metabolic flexibility of the groups in study.

Finally, although statistical analyses were adjusted for a wide variety of covariates, residual confounders such as physical activity, and nutritional status, may have affected the results.

The studies involving human participants were reviewed and approved by Hospital Ethical Committee Comitato Etico Azienda Ospedaliera Mater Domini Catanzaro, Italy. ES conceived the study, researched and analyzed data, and wrote and edited the manuscript.

PVi analyzed the data from the cardiac PET scans. AP and FC performed cardiac PET scans. GM, GT, TVF, MP, PHG, and AS researched data and reviewed the manuscript. FA, PVe, and GC contributed to study design and to the discussion and reviewed the manuscript.

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.

Diabetologia 33 10 —9. Bowden DW. Association of the PTPN1 gene with type 2 diabetes and insulin resistance. Discovery Med 4 24 — Alibegovic AC, Sonne MP, Højbjerre L, Hansen T, Pedersen O, van Hall G, et al. The t-allele of TCF7L2 rs associates with a reduced compensation of insulin secretion for insulin resistance induced by 9 days of bed rest.

Diabetes 59 4 — Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature — Freidenberg GR, Reichart D, Olefsky JM, Henry RR. Reversibility of defective adipocyte insulin receptor kinase activity in non-insulin-dependent diabetes mellitus. Effect weight loss.

J Clin Invest. Abd El-Kader SM, Al-Jiffri OH. Impact of weight reduction on insulin resistance; adhesive molecules and adipokines dysregulation among obese type 2 diabetic patients. Afr Health Sci 18 4 — Mechanisms of insulin resistance in obesity.

Front Med 7 1 — Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Petersen KF, Shulman GI. Etiology of insulin resistance. Am J Med 5 Suppl 1 :S10—6. Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutrients and inflammation.

Machado FVC, Pitta F, Hernandes NA, Bertolini GL. Physiopathological relationship between chronic obstructive pulmonary disease and insulin resistance. Endocrine 61 1 — Cosio FG, Kudva Y, van der Velde M, Larson TS, Textor SC, Griffin MD, et al.

New onset hyperglycemia and diabetes are associated with increased cardiovascular risk after kidney transplantation. Kidney Int 67 6 — Porrini E, Delgado P, Bigo C, Alvarez A, Cobo M, Checa MD, et al. Impact of metabolic syndrome on graft function and survival after cadaveric renal transplantation.

Am J Kidney Dis 48 1 — Rizza RA, Mandarino LJ, Gerich JE. Cortisol-induced insulin resistance in man: impaired suppression of glucose production and stimulation of glucose utilization due to a postreceptor detect of insulin action.

J Clin Endocrinol Metab 54 1 —8. Lopes PC, Fuhrmann A, Carvalho F, Sereno J, Santos MR, Pereira MJ, et al. Cyclosporine a enhances gluconeogenesis while sirolimus impairs insulin signaling in peripheral tissues after 3 weeks of treatment. Biochem Pharmacol 91 1 — Schäcke H, Döcke WD, Asadullah K.

Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther 96 1 — Rafacho A, Ortsäter H, Nadal A, Quesada I. Glucocorticoid treatment and endocrine pancreas function: implications for glucose homeostasis; insulin resistance and diabetes.

J Endocrinol 3 :R49— Rafacho A, Quallio S, Ribeiro DL, Taboga SR, Paula FM, Boschero AC, et al. The adaptive compensations in endocrine pancreas from glucocorticoid-treated rats are reversible after the interruption of treatment.

Acta Physiol Oxf. Galicia-Garcia U, Jebari S, Larrea-Sebal A, Uribe KB, Siddiqi H, Ostolaza H, et al. Statin treatment-induced development of type 2 diabetes: From clinical evidence to mechanistic insights.

Int J Mol Sci 21 Chang AM, Halter JB. Aging and insulin secretion. Am J Physiol Endocrinol Metab 1 :E7— Krentz AJ, Viljoen A, Sinclair A.

Insulin resistance: a risk marker for disease and disability in the older person. Diabetes Med 30 5 — Resnick HE, Harris MI, Brock DB, Harris TB.

American diabetes association diabetes diagnostic criteria; advancing age; and cardiovascular disease risk profiles: results from the third national health and nutrition examination survey. Diabetes Care 23 2 — Gabriely I, Ma XH, Yang XM, Atzmon G, Rajala MW, Berg AH, et al.

Removal of visceral fat prevents insulin resistance and glucose intolerance of aging: an adipokine-mediated process? Diabetes 51 10 —8. Reznick RM, Zong H, Li J, Morino K, Moore IK, Yu HJ, et al. Aging-associated reductions in AMP-activated protein kinase activity and mitochondrial biogenesis.

Cell Metab 5 2 —6. Lowell BB, Shulman GI. Mitochondrial dysfunction and type 2 diabetes. Science —7. Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Rothman DL, et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance.

Science —2. Vieira-Lara MA, Dommerholt MB, Zhang W, Blankestijn M, Wolters JC, Abegaz F, et al. Age-related susceptibility to insulin resistance arises from a combination of CPT1B decline and lipid overload. BMC Biol 19 1 Minokoshi Y, Kahn CR, Kahn BB. Tissue-specific ablation of the GLUT4 glucose transporter or the insulin receptor challenges assumptions about insulin action and glucose homeostasis.

J Biol Chem 36 — Gancheva S, Jelenik T, Álvarez-Hernández E, Roden M. Interorgan metabolic crosstalk in human insulin resistance. Physiol Rev 98 3 — Severinsen MCK, Pedersen BK. Muscle-organ crosstalk: The emerging roles of myokines. Endocr Rev 41 4 — Horita S, Nakamura M, Suzuki M, Satoh N, Suzuki A, Seki G.

Selective insulin resistance in the kidney. BioMed Res Int Ashraf A, Palakkott A, Ayoub MA. Anti-insulin receptor antibodies in the pathology and therapy of diabetes mellitus. Curr Diabetes Rev 17 2 — Hall C, Yu H, Choi E. Insulin receptor endocytosis in the pathophysiology of insulin resistance.

Exp Mol Med 52 6 — Rivers SL, Klip A, Giacca A. NOD1: An interface between innate immunity and insulin resistance. Endocrinology 5 — Copps KD, White MF.

Diabetologia 55 10 — Carvalho-Filho MA, Carvalho BM, Oliveira AG, Guadagnini D, Ueno M, Dias MM, et al. Double-stranded RNA-activated protein kinase is a key modulator of insulin sensitivity in physiological conditions and in obesity in mice. Endocrinology 11 — Hage Hassan R, Pacheco de Sousa AC, Mahfouz R, Hainault I, Blachnio-Zabielska A, Bourron O, et al.

J Biol Chem 6 — Cimmino I, Lorenzo V, Fiory F, Doti N, Ricci S, Cabaro S, et al. A peptide antagonist of Prep1-p interaction improves ceramide-induced insulin resistance in skeletal muscle cells. Oncotarget 8 42 — Abdelsalam SS, Korashy HM, Zeidan A, Agouni A.

The role of protein tyrosine phosphatase PTP -1B in cardiovascular disease and its interplay with insulin resistance. Biomolecules 9 7. Sevillano J, Sánchez-Alonso MG, Pizarro-Delgado J, Ramos-Álvarez MDP.

Role of receptor protein tyrosine phosphatases RPTPs in insulin signaling and secretion. Zhou R, Guo Q, Xiao Y, Guo Q, Huang Y, Li C, et al. Endocrine role of bone in the regulation of energy metabolism. Bone Res 9 1 Conte C, Epstein S, Napoli N.

Insulin resistance and bone: a biological partnership. Acta Diabetol 55 4 — Hevener AL, Zhou Z, Drew BG, Ribas V. The role of skeletal muscle estrogen receptors in metabolic homeostasis and insulin sensitivity. Adv Exp Med Biol — Hong SH, Choi KM. Sarcopenic obesity; insulin resistance; and their implications in cardiovascular and metabolic consequences.

Int J Mol Sci 21 2. Du P, Fan B, Han H, Zhen J, Shang J, Wang X, et al. NOD2 promotes renal injury by exacerbating inflammation and podocyte insulin resistance in diabetic nephropathy.

Kidney Int 84 2 — Lopez-Pastor AR, Gomez-Hernandez A, Diaz-Castroverde S, Gonzalez-Aseguinolaza G, Gonzalez-Rodriguez A, Garcia G, et al. Liver-specific insulin receptor isoform a expression enhances hepatic glucose uptake and ameliorates liver steatosis in a mouse model of diet-induced obesity.

Dis Model Mech 12 2. Diaz-Castroverde S, Baos S, Luque M, Di Scala M, González-Aseguinolaza G, Gómez-Hernández A, et al. Prevalent role of the insulin receptor isoform a in the regulation of hepatic glycogen metabolism in hepatocytes and in mice.

Diabetologia 59 12 — Diaz-Castroverde S, Gómez-Hernández A, Fernández S, García-Gómez G, Di Scala M, González-Aseguinolaza G, et al. Insulin receptor isoform a ameliorates long-term glucose intolerance in diabetic mice.

Dis Model Mech 9 11 — Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol 7 2 — James DE, Stöckli J, Birnbaum MJ.

The aetiology and molecular landscape of insulin resistance. Nat Rev Mol Cell Biol 22 11 — Fazakerley DJ, Krycer JR, Kearney AL, Hocking SL, James DE.

Muscle and adipose tissue insulin resistance: malady without mechanism? J Lipid Res 60 10 — McArdle MA, Finucane OM, Connaughton RM, McMorrow AM, Roche HM. Mechanisms of obesity-induced inflammation and insulin resistance: insights into the emerging role of nutritional strategies.

Front Endocrinol Lausanne. Osborn O, Olefsky JM. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med 18 3 — Boucher J, Kleinridders A, Kahn CR.

Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb Perspect Biol 6 1. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM. IRSmediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance.

Science —8. Donath MY, Dalmas É, Sauter NS, Böni-Schnetzler M. Inflammation in obesity and diabetes: islet dysfunction and therapeutic opportunity.

Cell Metab 17 6 — Nieto-Vazquez I, Fernández-Veledo S, Krämer DK, Vila-Bedmar R, Garcia-Guerra L, Lorenzo M. Insulin resistance associated to obesity: the link TNF-alpha.

Arch Physiol Biochem 3 — Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell 5 — Chen K, Li F, Li J, Cai H, Strom S, Bisello A, et al. Induction of leptin resistance through direct interaction of c-reactive protein with leptin.

Nat Med 12 4 — Yang M, Qiu S, He Y, Li L, Wu T, Ding N, et al. Genetic ablation of c-reactive protein gene confers resistance to obesity and insulin resistance in rats. Diabetologia 64 5 — Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, et al.

Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of ikkbeta. Yin J, Peng Y, Wu J, Wang Y, Yao L. J Leukoc Biol 95 1 — Jialal I, Kaur H, Devaraj S. Toll-like receptor status in obesity and metabolic syndrome: a translational perspective.

J Clin Endocrinol Metab 99 1 — Kim JJ, Sears DD. TLR4 and insulin resistance. Gastroenterol Res Pract Saberi M, Woods NB, de Luca C, Schenk S, Lu JC, Bandyopadhyay G, et al.

Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Cell Metab 10 5 — Kiechl S, Wittmann J, Giaccari A, Knoflach M, Willeit P, Bozec A, et al.

Blockade of receptor activator of nuclear factor-κB RANKL signaling improves hepatic insulin resistance and prevents development of diabetes mellitus. Nat Med 19 3 — Kaneto H, Matsuoka TA, Nakatani Y, Kawamori D, Miyatsuka T, Matsuhisa M, et al. Oxidative stress; ER stress; and the JNK pathway in type 2 diabetes.

J Mol Med Berl. Han MS, Perry RJ, Camporez JP, Scherer PE, Shulman GI, Gao G, et al. A feed-forward regulatory loop in adipose tissue promotes signaling by the hepatokine FGF Genes Dev 35 — Apostolopoulos V, de Courten MP, Stojanovska L, Blatch GL, Tangalakis K, de Courten B.

The complex immunological and inflammatory network of adipose tissue in obesity. Mol Nutr Food Res 60 1 — Orliaguet L, Dalmas E, Drareni K, Venteclef N, Alzaid F. Mechanisms of macrophage polarization in insulin signaling and sensitivity. McNelis JC, Olefsky JM.

Macrophages; immunity; and metabolic disease. Immunity 41 1 — Lumeng CN, DelProposto JB, Westcott DJ, Saltiel AR. Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes.

Diabetes 57 12 — Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase.

Nat Med 18 9 — Stefanovic-Racic M, Yang X, Turner MS, Mantell BS, Stolz DB, Sumpter TL, et al. Diabetes 61 9 —9. Wu D, Molofsky AB, Liang HE, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis.

Chng MH, Alonso MN, Barnes SE, Nguyen KD, Engleman EG. Adaptive immunity and antigen-specific activation in obesity-associated insulin resistance.

Mediators Inflamm Zand H, Morshedzadeh N, Naghashian F. Signaling pathways linking inflammation to insulin resistance. Diabetes Metab Syndr , S—s Boutens L, Hooiveld GJ, Dhingra S, Cramer RA, Netea MG, Stienstra R.

Unique metabolic activation of adipose tissue macrophages in obesity promotes inflammatory responses. Diabetologia 61 4 — Hotamisligil GS, Budavari A, Murray D, Spiegelman BM. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes.

central role of tumor necrosis factor-alpha. Olefsky JM, Glass CK. Macrophages; inflammation; and insulin resistance. Annu Rev Physiol — Tan T, Xiang Y, Deng C, Cao C, Ren Z, Huang G, et al. Variable frequencies of peripheral t-lymphocyte subsets in the diabetes spectrum from type 1 diabetes through latent autoimmune diabetes in adults LADA to type 2 diabetes.

Front Immunol Deng C, Xiang Y, Tan T, Ren Z, Cao C, Huang G, et al. Altered peripheral b-lymphocyte subsets in type 1 diabetes and latent autoimmune diabetes in adults. Diabetes Care 39 3 — Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, et al.

Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 15 8 —9. Revelo XS, Tsai S, Lei H, Luck H, Ghazarian M, Tsui H, et al. Perforin is a novel immune regulator of obesity-related insulin resistance. Diabetes 64 1 — Blasco-Baque V, Garidou L, Pomié C, Escoula Q, Loubieres P, Le Gall-David S, et al.

Periodontitis induced by porphyromonas gingivalis drives periodontal microbiota dysbiosis and insulin resistance via an impaired adaptive immune response. Gut 66 5 — Khan S, Luck H, Winer S, Winer DA.

Emerging concepts in intestinal immune control of obesity-related metabolic disease. Nat Commun 12 1 Moreno-Navarrete JM, Fernández-Real JM. The complement system is dysfunctional in metabolic disease: Evidences in plasma and adipose tissue from obese and insulin resistant subjects.

Semin Cell Dev Biol — Lee YS, Kim JW, Osborne O, Oh DY, Sasik R, Schenk S, et al. Increased adipocyte O2 consumption triggers HIF-1α; causing inflammation and insulin resistance in obesity.

Cell 6 — Malhotra A, White DP. Obstructive sleep apnoea. Lancet — Mesarwi OA, Loomba R, Malhotra A. Obstructive sleep apnea; hypoxia; and nonalcoholic fatty liver disease. Am J Respir Crit Care Med 7 — Iiyori N, Alonso LC, Li J, Sanders MH, Garcia-Ocana A, O'Doherty RM, et al.

Intermittent hypoxia causes insulin resistance in lean mice independent of autonomic activity. Am J Respir Crit Care Med 8 —7. Catrina SB, Zheng X. Hypoxia and hypoxia-inducible factors in diabetes and its complications. Diabetologia 64 4 — Wang N, Shi XF, Khan SA, Wang B, Semenza GL, Prabhakar NR, et al.

Hypoxia-inducible factor-1 mediates pancreatic β-cell dysfunction by intermittent hypoxia. Am J Physiol Cell Physiol 5 :C—c Xia QS, Lu FE, Wu F, Huang ZY, Dong H, Xu LJ, et al. New role for ceramide in hypoxia and insulin resistance. World J Gastroenterol 26 18 — O'Neill HM, Maarbjerg SJ, Crane JD, Jeppesen J, Jørgensen SB, Schertzer JD, et al.

AMP-activated protein kinase AMPK beta1beta2 muscle null mice reveal an essential role for AMPK in maintaining mitochondrial content and glucose uptake during exercise.

Proc Natl Acad Sci U S A. Yang Q, Vijayakumar A, Kahn BB. Metabolites as regulators of insulin sensitivity and metabolism. Nat Rev Mol Cell Biol 19 10 — Wymann MP, Schneiter R. Lipid signalling in disease. Nat Rev Mol Cell Biol 9 2 — Lipke K, Kubis-Kubiak A, Piwowar A.

Molecular Mechanism of Lipotoxicity as an Interesting Aspect in the Development of Pathological States-Current View of Knowledge. Cells 11 5. Mota M, Banini BA, Cazanave SC, Sanyal AJ. Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease.

Metabolism 65 8 — Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications.

Int J Mol Sci 20 9. Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin resistance in obese. BioMed Pharmacother.

Jiao P, Ma J, Feng B, Zhang H, Diehl JA, Chin YE, et al. FFA-induced adipocyte inflammation and insulin resistance: involvement of ER stress and IKKβ pathways.

Obes Silver Spring. DeBari MK, Abbott RD. Adipose tissue fibrosis: Mechanisms; models; and importance. Dahik VD, Frisdal E, Le Goff W. Rewiring of lipid metabolism in adipose tissue macrophages in obesity: Impact on insulin resistance and type 2 diabetes.

Opazo-Ríos L, Mas S, Marín-Royo G, Mezzano S, Gómez-Guerrero C, Moreno JA, et al. Lipotoxicity and diabetic nephropathy: Novel mechanistic insights and therapeutic opportunities. Int J Mol Sci 21 7. Nisr RB, Shah DS, Ganley IG, Hundal HS. Proinflammatory NFkB signalling promotes mitochondrial dysfunction in skeletal muscle in response to cellular fuel overloading.

Cell Mol Life Sci 76 24 — Chaurasia B, Summers SA. Ceramides - lipotoxic inducers of metabolic disorders: Trends in endocrinology and metabolism 26; ; Trends Endocrinol Metab 29 1 —7. Ceramides in Metabolism: Key Lipotoxic Players.

Bandet CL, Tan-Chen S, Bourron O, Le Stunff H, Hajduch E. Sphingolipid Metabolism: New Insight into Ceramide-Induced Lipotoxicity in Muscle Cells. Int J Mol Sci 20 3. Zhang Y, Fu Y, Jiang T, Liu B, Sun H, Zhang Y, et al.

Enhancing fatty acids oxidation via l-carnitine attenuates obesity-related atrial fibrillation and structural remodeling by activating AMPK signaling and alleviating cardiac lipotoxicity. Front Pharmacol D'Souza K, Paramel GV, Kienesberger PC.

Lysophosphatidic acid signaling in obesity and insulin resistance. Nutrients 10 4. Al-Jaber H, Al-Mansoori L, Elrayess MA. GATA-3 as a potential therapeutic target for insulin resistance and type 2 diabetes mellitus. Imierska M, Kurianiuk A, Błachnio-Zabielska A.

The influence of physical activity on the bioactive lipids metabolism in obesity-induced muscle insulin resistance. Biomolecules 10 Drazba MA, Holásková I, Sahyoun NR, Ventura Marra M.

Associations of Adiposity and Diet Quality with Serum Ceramides in Middle-Aged Adults with Cardiovascular Risk Factors.

J Clin Med 8 4. Summers SA, Chaurasia B, Holland WL. Metabolic messengers: ceramides. Nat Metab 1 11 —8. Chaurasia B, Tippetts TS, Mayoral Monibas R, Liu J, Li Y, Wang L, et al.

Metabolic syndrome Metabollc acid signaling in obesity and insulin resistance. Metabolic rate and insulin sensitivity ; 55 xensitivity — Dietary Pycnogenol and wound healing — an environmental insulun of insulin resistance acting on multiple levels of metabolism. The same result was shown in Figure 1b using both fasting and two hour values as determined by ISI 0, RED Article CAS PubMed Central PubMed Google Scholar Du, K.
Metabolism refers Selenium dynamic web elements the bodily process of extracting energy from food. Inulin Metabolic rate and insulin sensitivity metabolism by esnsitivity insulin levels. This in raye prevents the body from storing the energy it Metabolic rate and insulin sensitivity from food for insuoin use. In type 1 diabetesthis happens because the immune system is attacking the cells that make insulinwhich are in the pancreas. In type 2 diabetesthe body stops responding to insulin as well as it should. In this article, we will look at diabetes and metabolism more closely. Metabolism is the process through which the body creates energy from the food and drink a person consumes. Metabolic rate and insulin sensitivity

Author: Grokree

3 thoughts on “Metabolic rate and insulin sensitivity

  1. Ich meine, dass Sie sich irren. Ich biete es an, zu besprechen. Schreiben Sie mir in PM, wir werden reden.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com