Category: Home

Insulin sensitivity and insulin response

Insulin sensitivity and insulin response

Biochimie 8781—86 Signal Transduct. Nisulin are a number of risk factors for insulin resistance, including being overweight or obese or having a sedentary lifestyle.

Video

Intermitted Fasting And How To Do It

Insulin sensitivity and insulin response -

A review investigated the effects of two methods of intermittent fasting in overweight and obese adults. The first involved restricting calorie intake for 1—3 days per week and eating freely on the remaining days. As with a daily calorie-restricted diet, the researchers found that both types of intermittent fasting reduced insulin resistance.

However, this type of eating had no meaningful effect on blood glucose levels, so the authors concluded that more research is necessary. In addition to changing the foods in their diet, people looking to increase their insulin sensitivity may benefit from taking dietary supplements.

Taking probiotics or omega-3 fatty acid supplements may improve insulin sensitivity in people who are overweight. A clinical trial investigated the effects of both omega-3 fatty acids and probiotics on insulin sensitivity in 60 adults who were overweight but otherwise healthy.

The researchers reported that taking either a probiotic or omega-3 supplement for 6 weeks led to significant improvements in insulin sensitivity in comparison with a placebo. The increase in insulin sensitivity was even greater in those who took both supplements together. Learn everything you need to know about probiotics.

Magnesium supplements may also be beneficial for people wanting to improve their insulin sensitivity. A systematic review found that taking magnesium supplements for more than 4 months significantly improved insulin resistance in people with and without diabetes.

Read more about magnesium glycinate, a popular supplement. Resveratrol is a natural compound that occurs in the skin of red grapes. It is also available as a dietary supplement.

A meta-analysis of 11 studies found that taking resveratrol supplements significantly improved glucose control and insulin sensitivity in people with diabetes. However, the researchers did not observe the same effects in people without diabetes. They concluded that there is a need for more research on the effects of resveratrol supplementation in humans.

Low insulin sensitivity is a risk factor for developing type 2 diabetes. Exercising well, getting enough sleep, and eating a nutritious diet high in unsaturated fats and soluble fiber may help improve insulin sensitivity in people with and without diabetes. Certain dietary supplements may also be beneficial.

Many of these supplements are available to purchase online:. However, a person should be aware that the Food and Drug Administration FDA does not regulate supplements. Therefore, they should speak with their doctor before taking any supplement. Individuals can discover more resources for living with type 2 diabetes by downloading the free T2D Healthline app.

It provides access to expert content and peer support through one-on-one conversations and live group discussions. Download the app for iPhone or Android. Find out here about the differences and…. Many people avoid eating carbohydrates to help them lose weight. However, some carbohydrates are beneficial and can be healthful when included in the….

A study in mice suggests a potential mechanism that could explain why only some individuals with obesity develop type 2 diabetes. A type of medication used to treat type 2 diabetes could help lower the risk of developing kidney stones, a new study suggests.

Some recent evidence suggest that 4 grams of cinnamon per day, in the form of supplements, could help lower blood sugar levels in people with obesity…. World J. Rencurel, F. Requirement of glucose metabolism for regulation of glucose transporter type 2 GLUT2 gene expression in liver.

Thorens, B. Diabetologia 58 , — Eberlé, D. SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 86 , — Horton, J. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver.

Ferré, P. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes. Tobe, K. Dentin, R.

Carbohydrate responsive element binding protein ChREBP and sterol regulatory element binding protein-1c SREBP-1c : two key regulators of glucose metabolism and lipid synthesis in liver. Biochimie 87 , 81—86 Hepatic glucokinase is required for the synergistic action of ChREBP and SREBP-1c on glycolytic and lipogenic gene expression.

Herman, M. A novel ChREBP isoform in adipose tissue regulates systemic glucose metabolism. Iizuka, K. Deficiency of carbohydrate response element-binding protein ChREBP reduces lipogenesis as well as glycolysis.

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

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

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

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

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

Antuna-Puente, B. Adipokines: the missing link between insulin resistance and obesity. Diabetes Metab. Rabe, K. Adipokines and insulin resistance. Adipokines mediate inflammation and insulin resistance.

Lausanne 4 , 71 Li, S. Adiponectin levels and risk of type 2 diabetes: a systematic review and meta-analysis. JAMA , — Hotta, K. Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys.

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

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

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

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

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

Seufert, J. Leptin effects on pancreatic beta-cell gene expression and function. Diabetes 53 , S—S Myers, M. Mechanisms of leptin action and leptin resistance.

Burguera, B. Obesity is associated with a decreased leptin transport across the blood-brain barrier in rats. Gainsford, T. Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells. USA 93 , — Scherer, P. Adipose tissue: from lipid storage compartment to endocrine organ.

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

Nutrients 7 , — Frayn, K. Adipose tissue and the insulin resistance syndrome. Roden, M. Mechanism of free fatty acid-induced insulin resistance in humans. Invest 97 , — Goossens, G.

The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. RANDLE, P. The glucose fatty-acid cycle.

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

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

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

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

Yen, C. The triacylglycerol synthesis enzyme DGAT1 also catalyzes the synthesis of diacylglycerols, waxes, and retinyl esters. Lipid Res. Timmers, S. Muscular diacylglycerol metabolism and insulin resistance. Liu, L. Upregulation of myocellular DGAT1 augments triglyceride synthesis in skeletal muscle and protects against fat-induced insulin resistance.

Sokolowska, E. The Role of Ceramides in Insulin Resistance. Lausanne 10 , Schubert, K. Chaurasia, B. Trends Endocrinol. Stiban, J. Ceramide synthases: roles in cell physiology and signaling. Med Biol.

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

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

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

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

Yousef, A. IRS-1 genetic polymorphism r. Appl Clin. Genet 11 , 99— Hashemian, L. The role of the PPARG Pro12Ala common genetic variant on type 2 diabetes mellitus risk.

Zeng, Q. Association Between Insulin-like Growth Factor-1 rs Polymorphism and Type 2 Diabetes Mellitus Susceptibility: A Meta-Analysis. Front Genet 12 , Knowles, J. Identification and validation of N-acetyltransferase 2 as an insulin sensitivity gene.

Invest , Richards, J. A genome-wide association study reveals variants in ARL15 that influence adiponectin levels. PLoS Genet 5 , e Eichler, E. Missing heritability and strategies for finding the underlying causes of complex disease.

Genet 11 , — Flannick, J. Exome sequencing of 20, cases of type 2 diabetes and 24, controls. Nature , 71—76 Viñuela, A. Genetic variant effects on gene expression in human pancreatic islets and their implications for T2D.

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

Huypens, P. Epigenetic germline inheritance of diet-induced obesity and insulin resistance. Genet 48 , — Ling, C. Epigenetics in human obesity and type 2 diabetes.

Ahmed, S. The role of DNA methylation in the pathogenesis of type 2 diabetes mellitus. Epigenetics 12 , 1—23 Cierzniak, A. DNA methylation in adipocytes from visceral and subcutaneous adipose tissue influences insulin-signaling gene expression in obese individuals.

Int J. Epigenetic regulation of insulin action and secretion-role in the pathogenesis of type 2 diabetes. Intern Med , — Zhao, J.

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

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

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

Res , 13—24 Ma, X. Functional implications of DNA methylation in adipose biology. Gao, H. Long non-coding RNAs associated with metabolic traits in human white adipose tissue.

EBioMedicine 30 , — Altered adipose tissue DNA methylation status in metabolic syndrome: relationships between global DNA methylation and specific methylation at adipogenic, lipid metabolism and inflammatory candidate genes and metabolic variables.

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

Ali, M. DNA methylation profile of genes involved in inflammation and autoimmunity correlates with vascular function in morbidly obese adults. Epigenetics 17 , 93— Panchal, S. DNA methylation in adipose tissue and metabolic syndrome. Petrus, P. Adipocyte expression of SLC19A1 links DNA hypermethylation to adipose tissue inflammation and insulin resistance.

Ramos-Lopez, O. DNA methylation signatures at endoplasmic reticulum stress genes are associated with adiposity and insulin resistance. Genet Metab. Zhang, Y. Methylation of HIF3A promoter CpG islands contributes to insulin resistance in gestational diabetes mellitus. Genet Genom.

van Dijk, S. DNA methylation in blood from neonatal screening cards and the association with BMI and insulin sensitivity in early childhood. Endoplasmic reticulum stress epigenetics is related to adiposity, dyslipidemia, and insulin resistance.

Adipocyte 7 , — Stillman, B. Histone modifications: insights into their influence on gene expression. Cell , 6—9 Małodobra-Mazur, M. Histone modifications influence the insulin-signaling genes and are related to insulin resistance in human adipocytes.

Biochem Cell Biol. Davegårdh, C. DNA methylation in the pathogenesis of type 2 diabetes in humans. David, V. Gluco-incretinsregulate beta-cell glucose competence by epigenetic silencing of fxyd3expression. PLoS ONE 9 , e Emamgholipour, S.

Acetylation and insulin resistance: a focus on metabolic and mitogenic cascades of insulin signaling. Lab Sci. Zhou, S. Sirtuins and insulin resistance. Human adipose tissue H3K4me3 histone mark in adipogenic, lipid metabolism and inflammatory genes is positively associated with BMI and HOMA-IR.

PloS one 14 , e Maude, H. Epigenetics of Hepatic Insulin Resistance. Cech, T. The noncoding RNA revolution-trashing old rules to forge new ones.

Cell , 77—94 Formichi, C. Non-Coding RNAs: Novel Players in Insulin Resistance and Related Diseases. Ghafouri-Fard, S. The expression profile and role of non-coding RNAs in obesity. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Lausanne 9 , Agbu, P.

MicroRNA-mediated regulation of glucose and lipid metabolism. Poy, M. A pancreatic islet-specific microRNA regulates insulin secretion. Jiménez-Lucena, R. Lee, C. Foxa2 controls Pdx1 gene expression in pancreatic β-cells in vivo. Diabetes 51 , — Xu, G. Thioredoxin-interacting protein regulates insulin transcription through microRNA Med 19 , — Bao, L.

MicroRNA targets SOCS3 to inhibit beta-cell dysfunction in diabetes. PloS One 10 , e Iacomino, G. Role of microRNAs in obesity and obesity-related diseases. Genes Nutr.

Suksangrat, T. miRNA regulation of glucose and lipid metabolism in relation to diabetes and non-alcoholic fatty liver disease. Chen, Y. miRNA inhibits GLUT4 and is overexpressed in adipose tissue of polycystic ovary syndrome patients and women with insulin resistance. Zhou, Y. MicroRNAa induces insulin resistance by targeting PPARδ in skeletal muscle cells.

Wang, S. Li, C. Decreased glycogenolysis by miRp promotes regional glycogen accumulation within the spinal cord of amyotrophic lateral sclerosis mice. Front Mol. Zheng, H. MicroRNAp inhibits hepatic gluconeogenesis and reduces fasting blood glucose levels by suppressing G6Pase.

Theranostics 11 , Wagschal, A. Genome-wide identification of microRNAs regulating cholesterol and triglyceride homeostasis. Rayner, K. MiR contributes to the regulation of cholesterol homeostasis.

Over time, though, insulin resistance tends to get worse, and the pancreatic beta cells that make insulin can wear out. Eventually, the pancreas no longer produces enough insulin to overcome the cells' resistance. The result is higher blood glucose levels, and ultimately prediabetes or type 2 diabetes.

Insulin has other roles in the body besides regulating blood glucose levels, and the effects of insulin resistance are thought to go beyond diabetes. For example, some research has shown that insulin resistance, independent of diabetes, is associated with heart disease.

Scientists are beginning to get a better understanding of how insulin resistance develops. For starters, several genes have been identified that make a person more or less likely to develop the condition.

It's also known that older people are more prone to insulin resistance. Lifestyle can play a role, too. Being sedentary, overweight or obese increases the risk for insulin resistance. It's not clear, but some researchers theorize that extra fat tissue may cause inflammation, physiological stress or other changes in the cells that contribute to insulin resistance.

There may even be some undiscovered factor produced by fat tissue, perhaps a hormone, that signals the body to become insulin resistant. Doctors don't usually test for insulin resistance as a part of standard diabetes care. In clinical research, however, scientists may look specifically at measures of insulin resistance, often to study potential treatments for insulin resistance or type 2 diabetes.

They typically administer a large amount of insulin to a subject while at the same time delivering glucose to the blood to keep levels from dipping too low.

Last Updated Insulin sensitivity and insulin response This article was created by familydoctor. org editorial staff and reviewed by Andd Bradford Black pepper extract for arthritis, MD, FAAFP. Insulin resistance is a condition sdnsitivity to prediabetes and type 2 diabetes. Insulin resistance means your body is unable to respond to the amount of the hormone insulin it is producing. It helps protect your body from getting too much sugar glucose. Glucose gives you energy. However, too much sugar is harmful to your health. Glucose comes from the annd you eat. When blood glucose, also called responae sugar, levels rise Insulin sensitivity and insulin response ijsulin eat, your pancreas releases Immune system support into the blood. Insulin then Insulin sensitivity and insulin response blood glucose to keep it in the normal range. As a result, your pancreas makes more insulin to help glucose enter your cells. Prediabetes means your blood glucose levels are higher than normal but not high enough to be diagnosed as diabetes. Without enough insulin, extra glucose stays in your bloodstream rather than entering your cells.

Author: Kajisida

0 thoughts on “Insulin sensitivity and insulin response

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com