Category: Home

Promoting insulin function

Promoting insulin function

Proomting Spring Harb Perspect Biol. Neutrophils Fat metabolism and metabolic syndrome insulin resistance in mice fed a high-fat diet through secreted elastase. This is the form in which insulin is stored in beta cells.

Promoting insulin function -

A relatively safe and well accepted approach in the prevention and treatment of IR is via lifestyle interventions. Nutritional intervention is an important first step that emphasizes a low-calorie and low-fat diet that stimulates excessive insulin demands.

In addition, increased physical activity is recommended to help increase energy expenditures and improve muscle insulin sensitivity, this two approach represent the fundamental treatment for IR.

In this review, the mechanism of insulin action and IR are first described to promote the development of new therapeutic strategies. Further, the direct and indirect effects of insulin on target tissues are discussed to better understand the pivotal role of tissue crosstalk in systemic insulin action.

Lastly, diseases associated with IR are discussed and summarized. Many methods and multiple surrogate markers have been developed to calculate the IR. We then summarize the current measurements and potential biomarkers of IR to facilitate the clinical diagnosis.

Finally, we provide the general approaches including lifestyle intervention, specific pharmacologic interventions and clinical trials to reduce IR. Insulin is an endocrine peptide hormone with 51 amino acids and composed of an α and a β chain linked together as a dimer by two disulfide bridges 18 along with a third intrachain disulfide bridge in the α chain.

Accumulation of reports have demonstrated that IR is a complex metabolic disorder with integrated pathophysiology. The exact causes of IR has not been fully determined, 36 , 37 , 38 but ongoing research seeks to better understand how IR develops.

Here, we focus on the underlying mechanism of IR including direct defective of insulin signaling, epidemiological factors, interorgan metabolic crosstalk, metabolic mediators, genetic mutation, epigenetic dysregulation, non-coding RNAs, and gut microbiota dysbiosis.

As has been mentioned, the proper modulators acting on different steps of the signaling pathway ensure appropriate biological responses to insulin in different tissues. Thus, the diverse defect in signal transduction contributes to IR. Insulin exerts its biological effects by binding to its cell-surface receptors, therby activating specific adapter proteins, such us the insulin receptor substrate IRS proteins principally IRS1 and IRS2 , Src-homology 2 SH2 and protein-tyrosine phosphatase 1B PTP1B , eventually promoting downstream insulin signaling involving glucose homeostasis.

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

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

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

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

Specifically, insulin sensitivity and capacities for insulin responses in women is significantly higher than men. Male homozygous for the polymorphism of PPP1R3A gene that involved in glycogen synthase activity are significantly younger at diagnosis than female. Thus, additional studies are required to understand mechanisms underlying sex differences and IR development.

South Asian children exhibit greater IR compared with white European children, while girls are more insulin resistant than boys, with sex and ethnicity differences related to insulin sensitivity and body composition.

Despite the above objective factors, some modifiable lifestyle factors including diet, exercise, smoking, sleep and stress are also considered to contribute to IR. Further, circadian clocks disruption might also be an important factor to IR development via various factors including clock gene mutations, disturbed sleep cycles, shift work and jet lag.

Different investigations suggest that vitamin D supplementation might reduce IR in some people due to increasing insulin receptor genes transcription and anti-inflammatory properties, 95 while some researchers found that Vitamin D has no effect on IR. Both experimental animals and clinical studies have shown that many hormones can induce IR including glucocorticoids GCs , 97 cortisol, 98 growth hormone, 99 and human placental lactogen, which may decrease the insulin-suppressive effects on glucose production and reduce the insulin-stimulated glucose uptake.

Several other clinical medications including anti-adrenergic such as salbutamol, salmeterol, and formoterol , HIV protease inhibitors, , atypical antipsychotics and some exogenous insulin that may improve IR because of the disordered insulin signaling.

All together, there may have synergistic effects of different risk factors on insulin resistance, scientific researchers should cooperate with medical experts to reduce the chances of becoming insulin resistant. As discribed above, insulin signaling calibrates glucose homeostasis by limiting hepatic glucose output via decreased gluconeogenesis and glycogenolysis activities.

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

Insulin induces SREBP-1c maturation via a proteolytic mechanism started in the endoplasmic reticulum ER , wherein hepatic IR is highly associated with hepatic steatosis. Accordingly, restoration of nuclear SREBP-1c expression in liver-specific Chrebp defective mice normalized expression of some lipogenic genes, while not affecting glycolytic genes expressing.

In contrast, ChREBP overexpression alone failed to promote the expression of lipogenic genes in the livers of mice lacking active SREBPs. Together, these data demonstrate that SREBP-1c mediates the induction of insulin lipogenic genes, but that SREBP-1c and ChREBP are both necessary for harmonious induction of glycolytic and lipogenic genes.

Altogether, these above pathways and components can be used to clarify the popular pathophysiology of hepatic IR. The lipid metabolisms including increased de novo lipogenesis and attenuation of lipolysis in the adipose tissue largely coordinate with glucose homeostasis response to insulin stimulation.

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

The lipogenesis stimulation of insulin is also reduced in larger, more insulin-resistant cells. Insulin suppression of lipolysis includes the hydrolytic cleavage of triglycerides, resulting in the generation of fatty acids and glycerol.

The best understood effectors for this process are PDE3B and ABHD15 that operated by the suppression of cAMP to attenuate pro-lipolytic PKA signaling toward adipose triglyceride lipase ATGL , hormone-sensitive lipase HSL , and perilipin PLIN. Further, inhibition of PDE3B inhibits insulin-induced glucose uptake and antilipolysis.

Insulin stimulated protein synthesis is mediated by activation of the protein kinases Akt and mTOR specifically mTORC1 and mTORC2 in numerous insulin-responsive cell types, such as hepatocytes, adipocytes, and myocytes. Inhibition of mTOR by rapamycin obviously impairs insulin-activated protein synthesis.

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

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

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

Thus, chemerin may increase insulin sensitivity in adipose tissue. Leptin is a cytokine encoded by ob gene and produced by the adipocytes. In summary, adipose tissue is a central node for distinct adipokines and bioactive mediators in IR pathophysiology.

Consequently, identifying the effects of new adipokines will help in the development of new therapeutic strategies for obesity-induced diseases. The specific insulin actions in adipose tissue include activation of glucose uptake and triglyceride synthesis, suppression of triglyceride hydrolysis and free fatty acids FFA and glycerol release into the blood circulation.

Once the adipose tissue expandability exceeded limit under overnutrition, excess lipids and toxic lipid metabolites FFA, diacylglycerol, ceramide accumulated in non-adipose tissues, thus leading to lipid-induced toxicity lipotoxicity and developed IR in liver and muscle.

This process would in turn induce increased intracellular citrate levels, thereby inhibiting glucosephosphate G6P accumulation. Increased G6P levels then result in decreased hexokinase activity, increased glucose accumulation, and reduced glucose uptake.

Other studies have demonstrated the relevance of the glucose-fatty acid cycle to lipid-induced IR. For example, lipid infusions combined with heparin can be used to activate lipoprotein lipase, thereby increasing plasma concentrations of fatty acids.

Further, these infusions promote muscle lipid accumulation and effectively induce IR. Consistent with the above studies, elevated plasma fatty acid concentrations can result in increased intracellular diacylglycerol DAG levels, leading to the activation of protein kinase C isoform PKC-θ and PKC-ε isoforms in skeletal muscles and liver respectively.

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

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

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

Some human genetic studies indicated that different genomic loci were associated with fasting insulin levels, higher triglyceride and lower HDL cholesterol levels, , which are different hallmarks of IR.

The peroxisome proliferator-activated receptor gamma PPARγ variant Pro12Ala was one of the first genetic variants identified that is involved in fatty acid and energy metabolism and that is associated with a low risk of developing T2DM. Nevertheless, additional studies are needed to assess the functional relationships between the genetic variants and IR, that are also influenced by various lifestyle and environmental factors.

Recent studies have suggested that epigenetic modifications such as DNA methylation DNAm and histone post-translational modifications PTM are implicated in the development of systemic IR.

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

For example, increased INS promoter methylation levels and INS mRNA suppression were observed under over-nutrition conditions and obese T2DM patients. Another study demonstrated that increased IGFBP2 DNA methylation levels were are associated with lower mRNA expression levels in Visceral Adipose tissue VAT of abdominal obesity.

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

For example, peroxisome proliferator-activated receptor-α and -γ PPAR-α and PPAR-γ, respectively are encoded by PPARA and PPARG , respectively, and they are the two primary nuclear peroxisome proliferator-activated receptors involved in lipid metabolism.

Higher PPARA and PPARG methylation levels were observed in association with obesity, consistent with decreased PPAR-α and PPAR-γ protein expression levels, that lead to dyslipidemia and IR.

SLC19A1, a gene encoding a membrane folate carrier, was reduced in obese WAT and induced global DNA hypermethylation of chemokine C-C motif chemokine ligand 2 CCL2 that is a key factor in WAT inflammation, resulting in increased CCL2 protein secretion and the development of IR in obese.

In addition, several genes methylation involved in hypoxia stress and endoplasmic reticulum stress were regulated in obesity related metabolic diseases. Recent epigenetic genome-wide analysis identified low HIF3A methylation levels upregulates HIF3A expression in adipose tissue, thereby leading to adipose tissue dysfunction and adiposity.

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

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

Increasing evidence indicates , that IGFR, InsR, IRS1, Akt, GLUT4, and PPAR are more deacetylated in association with IR than in normal physiological conditions. In contrast, IRS2, FoxO, JNK, and AMPK are usually acetylated in association with IR. Castellano-Castillo, D. Further, global proteomic analyses have revealed 15 histone modifications that are differentially abundant in hepatic IR.

MicroRNAs miRNAs are small ncRNAs nucleotides incorporated into Argonaute Ago protein to form miRISCs, which can inhibit the expression of partially or completely complementary target mRMAs.

Several miRNAs are involved in β cell differentiation and mature β cell functioning. For example, islet-specific miR overexpression represses glucose-stimulated insulin secretion GSIS and insulin gene transcription, that is then reversed upon miR inhibition.

Thus, these markers may improve disease prediction and prevention in individuals at high risk for T2DM. Furthermore, pdx1, neurogenin-3 ngn3 , and a transcriptional factor essential for insulin transcription MafA are essential transcription factors for β-cell differentiation.

Thus, miRa2 can directly modulate insulin expression through foxA2 and then pdx1. miR expression is induced by the cellular redox regulator thioredoxin-interacting protein TXNIP that then represses MafA, thereby inhibiting insulin production.

Numerous studies suggest that miRNAs have pivotal roles in glucose and lipid metabolism. miR was first reported to directly regulate GLUT4 expression in adipocytes.

In addition, the anti-diabetic drug metformin can up-regulate miRp expression to suppress G6Pase and inhibit hepatic gluconeogenesis.

The balance of low-density lipoprotein LDL and high-density lipoprotein HDL molecules that are synthesized in hepatocytes is critical for lipid homeostasis. Many miRNAs have been identified as critical regulators of HDL and LDL biogenesis.

For example, miR, miR, miR, and miRa repress expression of the ATP-binding cassette transporter ABCA1 that mediates hepatic HDL generation. In addition, miRc targets the gene encoding microsomal triglyceride transfer protein MTP that is required for the lipidation of newly synthesized APOB in the liver for LD lipoprotein production.

miRc overexpression reduces the assembly and secretion of these APOB-containing lipoproteins, resulting in decreased plasma LDL levels.

miRa and miR repress LDLR expression and inhibition of these miRNAs results in enhanced LDLR expression and clearance of circulating LDL. Further, miR and miRd target the LDLR chaperonin PCSK9 and IDOL in addition to the rate-limiting enzyme in cholesterol biosynthesis, HMGCR.

Chronic inflammation in insulin-reactive tissues is one of the most important causes of IR and increasing evidence suggests that miRNAs has a pivotal role in the inflammatory process. Obesity inhibited miR expression in adipose tissue macrophages ATMs , and miR was shown to target Delta-like-4 DLL4 , a Notch1 ligand is associated with ATM inflammation.

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

Therefore, it might be that metabolism-regulating miRNAs play a vital role in the dynamics of metabolic homeostasis. Long non-coding RNAs lncRNAs are non-coding transcripts more than nucleotides, and the subcellular localization of lncRNAs determines their function. LncRNAs located in the nucleus could affect chromosomal biology or interact with transcription factors to regulate gene transcription; lncRNAs located in cytosol could modulate mRNA stability and translational efficiency by acting as sponges for miRNAs or direct pairing with mRNA.

Recent advances have shown that lncRNAs play crucial roles in the pathologys of IR and diabetes. Glucose and lipid metabolism disorders are the primary causes for the pathophysiological development of IR.

The lncRNA SRA promotes insulin-stimulated glucose uptake by co-activating PPARγ, leading to increased phosphorylation of the downstream targets Akt and FOXO1 in adipocytes. These processes are closely related to the genes PGC1a and CPT1b that reverse FFA-induced lipid accumulation and improve IR.

In addition the insulin target tissues, transcriptome profiling and different studies have identified several β-cell specific lncRNAs that contribute to obesity-mediated β-cell dysfunction and apoptosis. LncRNA MALAT1 downregulation may lead to pancreatic β-cell dysfunction and T2DM development by direct interaction and regulation of polypyrimidine bundle binding protein 1 PTBP1.

Further, lncRNA-p overexpression can decrease the β cell apoptosis ratio and partially reverse the glucotoxicity effects on GSIS function. Contrary to conventional linear RNA, circRNAs are noncoding RNAs that generated from precursor mRNAs by back-splicing circularization, which is derived from exonic circRNAs, intronic circRNAs, exonic-intronic circRNAs and ntergenic circRNAs.

Recent studies have suggested that newly identified circRNAs are novel factors in the initiation and development of IR.

CircHIPK3 is one of the most abundant circRNAs in β-cells and regulates hyperglycemia and IR by sequestering miRp and miRp, thereby increasing mRNA expression of key β-cell genes e.

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

Another circRNA deep sequencing analysis of sera from patients with metabolic syndrome MetS identified the presence of a novel circRNA, circRNF, involved in MetS progression. AMPK is a critical factor in energy homeostasis including glycolysis, lipolysis, and fatty acid oxidation FAO.

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

circMAP3K4 is another potentially important circRNA involved in glucose metabolism that is highly expressed in the placentas of patients with gestational diabetes mellitus GDM and the IR model. Nevertheless, the exact roles and regulatory mechanisms of circRNAs in IR require additional clarity.

The microbes living in the human gut are key contributors to host metabolism and immune function through mediating the interaction between the host and environment, or releasing metabolites and cytokines.

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

For example, microbiome genome-wide association studies mGWAS have identified that variants of different genes for example, VDR , LCT , NOD2 , FUT2 , and APOA5 that are associated with distinct gut microbiome compositions.

Growing evidence in the last two decades has suggested that gut microbial dysbiosis contributes to increased risks of metabolic defects like obesity, IR, and diabetes. LPS circulation then contributes to the chronic inflammation of liver and adipose tissue that is associated with the development of IR, in addition to other conditions associated with metabolic syndromes.

As we all know, IR is a state in which higher than normal concentrations of insulin are needed for a normal response, leading directly to hyperinsulinaemia and impaired glucose tolerance. Non-alcoholic fatty liver disease NAFLD is one of the most common liver diseases worldwide. Adipose tissue is a physiologic reservoir of fatty acids, when the storage capacity is exceeded, the accumulation of heterotopic lipids leads to lipotoxicity, thereby promoting low-grade inflammation and IR in the liver.

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

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

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

Current researches have demonstrated that the mechanisms of systemic IR and brain-specific IR have close links with AD pathogenesis.

Pioglitazone acts similarly as Rosiglitazone by reducing tau and Aβ deposits in the hippocampus, and improving neuronal plasticity and learning in AD. Moreover, overlapping pathological features exist for diabetes, IR, and AD. Chronic kidney disease CKD involves a gradual loss of kidney function and inability to filter blood , and is a major risk factor for end-stage kidney failure ESKF and CVDs.

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

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

Actually, IR precedes the occurrence of T2DM, so how to increase the accurate assessment of insulin sensitivity is very important to predict the risk and evaluate the management of impaired insulin sensitivity and metabolic syndrome in research and clinical practice.

HOMA2 updated HOMA model which took account of variations in hepatic and peripheral glucose resistance , homeostatic Model Assessment for IR HOMA-IR , the oral glucose insulin sensitivity index OGSI , fasting Insulin FINS , and fasting plasma glucose FPG based on fasting glucose and insulin levels , , , , are widely utilized IR measurements in clinical research.

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

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

No medications exist currently that are specifically approved to treat IR, but IR management 91 , , is possible through lifestyle changes like dietary, increased exercise, and disease prevention in addition to alternative medications Fig.

Among these treatments, lifestyle changes should be the main focus for IR treatment, with nutritional intervention to decrease calories, avoidance of carbohydrates, and focusing on aliments with low glycemic index including vegetables, fruits, whole-grain products, nuts, lean meats or beans to provide higher fiber, vitamins, healthy fats and protein are particularly helpful for people trying to improve insulin sensitivity.

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

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

gov to reduce IR and summarized them mainly include: 1 Diet intervention, such as Low-fat vegetarian Food, high-protein food, calorie restriction, vitamin D supplementation to reduce the IR in human obesity. We present some clinical trials of IR intervention in Table 2.

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

In recent years, accumulating preclinical studies on the intervention of IR have been reported, which have important reference significance for the development of new drugs. We present the related studies on IR reported in recent years in Table 3 , including animal models, treatment methods and results.

Pre-clinical IR intervention mainly includes drug intervention, probiotic therapy and exercise supplement. Drug therapy to improve IR is the main research direction at present. Researchers found that Valdecoxib VAL can inhibit inflammation and endoplasmic reticulum ER stress through AMPK-regulated HSPB1 pathway, thus improving skeletal muscle IR under hyperlipidemia.

The researchers found that the mixed nasal administration of GLP-1 receptor agonist and L-form of peneracin can effectively alleviate the cognitive dysfunction of SAMP8 mice.

Natividad et al. Regular exercise is an alternative intervention measure to maintain the blood sugar level in the normal range and reduce the risk factors. Hsu and colleagues found that exercise combined with probiotics intervention can have a positive effect on blood sugar and increase insulin sensitivity in mice.

The above results show that drug intervention, probiotic supplementation and intensive exercise can improve IR but more clinical data are still needed. Overall, the increased incidence of IR and the key roles of IR plays in many diseases, urgently require a better understanding of IR pathogenesis in addition to how IR interacts with genetics and different environments.

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

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

Kung, Y. Total synthesis of crystalline bovine insulin. Goeddel, D. et al. Expression in Escherichia coli of chemically synthesized genes for human insulin. USA 76 , — Vecchio, I. The discovery of insulin: an important milestone in the history of medicine.

Front Endocrinol. Article Google Scholar. Cheatham, B. Insulin action and the insulin signaling network. Root, H. Insulin resistance and bronze diabetes. Laakso, M. Insulin resistance and hyperglycaemia in cardiovascular disease development.

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

Diamanti-Kandarakis, E. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications. Stenvers, D. Circadian clocks and insulin resistance. Article PubMed CAS Google Scholar. Freeman AM, Pennings N. Insulin Resistance. In: StatPearls Internet.

Treasure Island FL : StatPearls Publishing. PMID: American Diabetes Association. Prevention or delay of type 2 diabetes: standards of medical care in diabetes Diabetes Care 44 , S34—S39 Pharmacologic approaches to glycemic treatment: Standards of Medical Care in Diabetes Diabetes Care 44 , S—S Weiss, M.

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

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

White, M. Mechanisms of insulin action. In Atlas of diabetes pp. Springer, Boston, MA Newgard, C. Organizing glucose disposal: emerging roles of the glycogen targeting subunits of protein phosphatase Diabetes 49 , — Beurel, E. Glycogen synthase kinase-3 GSK3 : regulation, actions, and diseases.

Article CAS Google Scholar. Dong, X. Inactivation of hepatic Foxo1 by insulin signaling is required for adaptive nutrient homeostasis and endocrine growth regulation. Cell Metab. Puigserver, P. Insulin-regulated hepatic gluconeogenesis through FOXO1—PGC-1α interaction.

Nature , — Vander Haar, E. Garami, A. Cell 11 , — Laplante, M. mTORC1 activates SREBP-1c and uncouples lipogenesis from gluconeogenesis. USA , — Han, Y. Post-translational regulation of lipogenesis via AMPK-dependent phosphorylation of insulin-induced gene.

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

Cell Rep. James, D. The aetiology and molecular landscape of insulin resistance. Tam, C. Defining insulin resistance from hyperinsulinemic-euglycemic clamps.

Diabetes care 35 , — Samuel, V. Mechanisms for insulin resistance: common threads and missing links. Cell , — Ye, J. Mechanisms of insulin resistance in obesity.

Front Med. Article PubMed PubMed Central Google Scholar. Yaribeygi, H. Insulin resistance: Review of the underlying molecular mechanisms. Cell Physiol. De Meyts, P. The insulin receptor: a prototype for dimeric, allosteric membrane receptors?

Trends Biochem Sci. Caro, J. Insulin receptor kinase in human skeletal muscle from obese subjects with and without noninsulin dependent diabetes. Invest 79 , — Fröjdö, S. Alterations of insulin signaling in type 2 diabetes: a review of the current evidence from humans.

Biochim Biophys. Acta , 83—92 Fisher, S. Michael, M. Loss of insulin signaling in hepatocytes leads to severe insulin resistance and progressive hepatic dysfunction.

Cell 6 , 87—97 Davis, R. The c-Jun NH2-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser Article PubMed Google Scholar.

Carvalho-Filho, M. Diabetes 54 , — Taniguchi, C. Critical nodes in signalling pathways: insights into insulin action. Brachmann, S. Phosphoinositide 3-kinase catalytic subunit deletion and regulatory subunit deletion have opposite effects on insulin sensitivity in mice.

Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp70 S6 kinase, DNA synthesis, and glucose transporter translocation.

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

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

Insulin receptor tyrosine kinase is defective in skeletal muscle of insulin-resistant obese mice. Brozinick, J. Defective signaling through Akt-2 and-3 but not Akt-1 in insulin-resistant human skeletal muscle: potential role in insulin resistance. Learn how much fruit you should eat per day and what to know about fruit as part of a diabetes eating plan.

Reducing your carb intake could help increase insulin sensitivity because high carb eating patterns tend to lead to spikes in blood sugar Eating regularly benefits insulin sensitivity, and eating low glycemic index GI carbs, in particular, is better because they slow the release of sugar into the blood, giving insulin more time to work efficiently 20 , Learn more about the types of carbs.

Added sugars , which are found mostly in highly processed foods, include primarily high fructose corn syrup and table sugar sucrose. Many studies have found that higher intakes of fructose can increase insulin resistance among people with diabetes 22 , The effects of fructose on insulin resistance also appear to affect people who do not have diabetes, as reported in a review of literature showing that consuming a lot of fructose over less than 60 days increased liver insulin resistance, independent of total calorie intake Learn which foods contain lots of added sugar.

Herbs and spices, including fenugreek , turmeric , ginger , cinnamon , and garlic , have shown promising results in increasing insulin sensitivity 25 , Other herbs that may have this effect include basil, dill, fennel, parsley, cumin, nutmeg, oregano, and rosemary. Research has identified at least compounds contained in a variety of herbs and spices that may contribute to reducing insulin resistance Several studies have found that drinking green tea can help increase insulin sensitivity and reduce blood sugar 27 , These beneficial effects of green tea could be due to its powerful antioxidant epigallocatechin gallate EGCG , which helps increase insulin sensitivity Vinegar could help increase insulin sensitivity by reducing blood sugar and improving the effectiveness of insulin It also appears to delay the stomach from releasing food into the intestines, giving the body more time to absorb sugar into the bloodstream Unlike other fats, trans fats provide no health benefits and increase the risk of many diseases Evidence on the effects of high trans-fat intake on insulin resistance appears to be mixed.

Some human studies have found it harmful, while others have not 33 , Many different supplements can help increase insulin sensitivity, including vitamin C , probiotics , and magnesium.

That said, many other supplements, such as zinc, folate, and vitamin D, do not appear to have this effect, according to research As with all supplements, there is a risk they may interact with any current medication you may be taking.

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

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

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

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

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

A resistance to its effects, called insulin resistance, is a leading driver of many health conditions. If not treated, high insulin levels can lead to serious health problems. For example, pair an apple with peanut butter, whole grain crackers with cheese, or a banana with almonds.

While you do not need to eliminate any foods from your diet completely, the key is to be aware of how certain foods affect your blood sugar levels and how to balance or offset those with other food choices, says Hoskins.

Hoskins recommends the following foods to provide a more stable energy source and support insulin sensitivity. These types of food are high in fiber and nutrients. To know if it is a whole grain, read the label, says Hoskins.

All fruits are packed with fiber and nutrients, but some are higher in sugar than others, like grapes and bananas. So if you want to consume a larger portion, keep in mind that you can eat a cup of berries compared to half a banana for about the same sugar content, Hoskins says.

With this in mind, some lower carbohydrate fruits include:. While vegetables are always a good choice, keep in mind that some vegetables, like potatoes and sweet potatoes, are starchier than others and provide more carbohydrates.

Vegetables with little to no carbohydrate include:. Trying to make the changes in your diet needed to adhere to these food choices may seem overwhelming at first.

To make it easier, Hoskins recommends the following tips:. Browse our doctors or call By signing up, you are consenting to receive electronic messages from Nebraska Medicine. Find a Doctor Find a Location Find a Service. Advancing Health Homepage.

Get health information you can use, fact-checked by Nebraska Medicine experts. Breadcrumb Home Advancing Health Conditions and Services Body Systems Diabetes 5 best foods to improve insulin resistance. Conditions and Services Body Systems Diabetes 5 best foods to improve insulin resistance.

March 2, Complex carbohydrates These types of food are high in fiber and nutrients.

Fat metabolism and metabolic syndrome resistance IR plays Insulon crucial role in the development and progression of metabolism-related diseases insuoin as diabetes, Hypertension management strategies, tumors, and nonalcoholic fatty liver disease, and provides the basis for a common Promoting insulin function of these chronic diseases. Insuln Promoting insulin function study, we provide Promotinb systematic review of the causes, mechanisms, and treatments of IR. The pathogenesis of IR depends on genetics, obesity, age, disease, and drug effects. Mechanistically, any factor leading to abnormalities in the insulin signaling pathway leads to the development of IR in the host, including insulin receptor abnormalities, disturbances in the internal environment regarding inflammation, hypoxia, lipotoxicity, and immunitymetabolic function of the liver and organelles, and other abnormalities. The available therapeutic strategies for IR are mainly exercise and dietary habit improvement, and chemotherapy based on biguanides and glucagon-like peptide-1, and traditional Chinese medicine treatments e. Invisible changes in the body begin long funftion a insuoin is diagnosed Promoting insulin function type Promotjng Fat metabolism and metabolic syndrome. One of the most important unseen changes? Insulin resistance. Insulin is a key player in developing type 2 diabetes. Here are the high points:. But this finely tuned system can quickly get out of whack, as follows:.

Promoting insulin function -

Middelbeek RJW , Chambers MA , Tantiwong P , et al. Insulin stimulation regulates AS and TBC1D1 phosphorylation sites in human skeletal muscle. Nutr Diabetes. Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation.

Dumm M. Glucose utilization and lactate production by leucocytes of patients with diabetes mellitus. Proc Soc Exp Biol Med. Esmann V. Effect of insulin on human leucocytes. Kalant N , Schucher R. Glucose utilization and insulin responsiveness of leukocytes in diabetics.

Can J Biochem Physiol. Martin SP , McKinney GR , Green R , Becker C. The influence of glucose, fructose, and insulin on the metabolism of leukocytes of healthy and diabetic subjects.

J Clin Invest. Englhardt A , Metz T. Investigations on glucose uptake in isolated human leucocytes from normal and diabetic subjects. Chaudhry R , Varacallo M. Biochemistry Glycolysis. In: StatPearls [Internet] , ed. Treasure Island FL. StatPearls Publishing , Burgner JW , Ray WJ.

On the origin of the lactate dehydrogenase induced rate effect. Alabduladhem T , Bordoni B. Physiology, Krebs Cycle. In: StatPearls , ed. Berg J , Tymoczko J , Stryer L.

The metabolism of glucose 6-phosphate by the pentose phosphate pathway is coordinated with glycolysis. In: Freeman WH , ed, Biochemestry. New York. WH Freeman and Company , : Section Le A , Lane AN , Hamaker M , et al.

Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. Yang C , Ko B , Hensley CT , et al. Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport. Mol Cell. Kramer PA , Ravi S , Chacko B , Johnson MS , Darley-Usmar VM.

A review of the mitochondrial and glycolytic metabolism in human platelets and leukocytes: implications for their use as bioenergetic biomarkers. Redox Biol. Borregaard N , Herlin T.

Energy metabolism of human neutrophils during phagocytosis. Hayashi Y , Yokota A , Harada H , Huang G. Cancer Sci. Jantsch J , Wiese M , Schödel J , et al. Toll-like receptor activation and hypoxia use distinct signaling pathways to stabilize hypoxia-inducible factor 1α HIF1A and result in differential HIF1A-dependent gene expression.

J Leukoc Biol. Azevedo EP , Rochael NC , Guimarães-Costa AB , et al. A metabolic shift toward pentose phosphate pathway is necessary for amyloid fibril- and phorbol myristate acetate-induced neutrophil extracellular trap NET formation. van Raam BJ , Sluiter W , de Wit E , Roos D , Verhoeven AJ , Kuijpers TW.

Mitochondrial membrane potential in human neutrophils is maintained by complex III activity in the absence of supercomplex organisation. Porter L , Toepfner N , Bashant KR , et al. Metabolic profiling of human eosinophils. Front Immunol. Domblides C , Lartigue L , Faustin B. Metabolic stress in the immune function of T cells, macrophages and dendritic cells.

West AP , Brodsky IE , Rahner C , et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Pearce EL , Pearce EJ. Metabolic pathways in immune cell activation and quiescence. Vats D , Mukundan L , Odegaard JI , et al. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation.

Macintyre AN , Rathmell JC. Activated lymphocytes as a metabolic model for carcinogenesis. Cancer Metab. Sp Walrand , Guillet C , Boirie Y , Vasson M-P. Insulin differentially regulates monocyte and polymorphonuclear neutrophil functions in healthy young and elderly humans.

J Clin Endocrinol Metab. Helderman JH. Acute regulation of human lymphocyte insulin receptors. Analysis by the glucose clamp. Brown TJ , Ercolani L , Ginsberg BH.

Properties and regulation of the T lymphocyte insulin receptor. Helderman JH , Ayuso R , Rosenstock J , Raskin P. Monocyte-T lymphocyte interaction for regulation of insulin receptors of the activated T lymphocyte. Helderman JH , Raskin P. The T lymphocyte insulin receptor in diabetes and obesity: an intrinsic binding defect.

Bieger W , Weicker H , Michl J. Transport and utilization of amino acids and glucose in human monocytes: activation of glucose metabolism by insulin.

Daneman D , Zinman B , Elliott ME , Bilan PJ , Klip A. Insulin-stimulated glucose transport in circulating mononuclear cells from nondiabetic and IDDM subjects. Dimitriadis G , Maratou E , Boutati E , Psarra K , Papasteriades C , Raptis SA.

Evaluation of glucose transport and its regulation by insulin in human monocytes using flow cytometry. Cytometry A.

The diabetic leukocyte. The polymorphonuclear leukocyte in diabetes mellitus. J Clin Chem Clin Biochem. Korgun ET , Demir R , Sedlmayr P , et al.

Sustained hypoglycemia affects glucose transporter expression of human blood leukocytes. Blood Cells Mol Dis. Maratou E , Dimitriadis G , Kollias A , et al. Glucose transporter expression on the plasma membrane of resting and activated white blood cells.

Eur J Clin Invest. Fu Y , Maianu L , Melbert BR , Garvey WT. Facilitative glucose transporter gene expression in human lymphocytes, monocytes, and macrophages: a role for GLUT isoforms.

Ahmed N , Kansara M , Berridge MV. Acute regulation of glucose transport in a monocyte-macrophage cell line: glut-3 affinity for glucose is enhanced during the respiratory burst. Biochem J. Mavros Y , Simar D , Singh MAF. Glucose tranporter-4 expression in monocytes: a systematic review.

Diabetes Res Clin Pract. Kumar S , Dikshit M. Metabolic insight of neutrophils in health and disease. Schuster DP , Brody SL , Zhou Z , et al. Regulation of lipopolysaccharide-induced increases in neutrophil glucose uptake.

Am J Physiol Lung Cell Mol Physiol. Venge P , Moberg L , Björnsson E , Bergström M , Långström B , Håkansson L. Mechanisms of basal and cytokine-induced uptake of glucose in normal human eosinophils: relation to apoptosis.

Respir Med. Sanderson CJ. Interleukin-5, eosinophils, and disease. Viardot A , Grey ST , Mackay F , Chisholm D. Potential antiinflammatory role of insulin via the preferential polarization of effector T cells toward a T helper 2 phenotype.

Piątkiewicz P , Czech A , Tatoń J. Glucose transport in human peripheral blood lymphocytes influenced by type 2 diabetes mellitus. Arch Immunol Ther Exp Warsz. Maclver NJ , Jacobs SR , Wieman HL , Wofford JA , Coloff JL , Rathmell JC.

Glucose metabolism in lymphocytes is a regulated process with significant effects on immune cell function and survival. Fischer HJ , Sie C , Schumann E , et al. The insulin receptor plays a critical role in T cell function and adaptive immunity. J Immunol. Wang R , Dillon CP , Shi LZ , et al.

The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Swords RT , Schenk T , Stengel S , et al.

Tsai S , Clemente-Casares X , Zhou AC , et al. Insulin receptor-mediated stimulation boosts T cell immunity during inflammation and infection.

Brinkmann V , Reichard U , Goosmann C , et al. Neutrophil extracellular traps kill bacteria. Segal AW. How neutrophils kill microbes. Annu Rev Immunol. Cavalot F , Anfossi G , Russo I , et al. Insulin stimulates the polymorphonuclear leukocyte chemokinesis.

Horm Metab Res. Insulin, at physiological concentrations, enhances the polymorphonuclear leukocyte chemotactic properties. Oldenborg P-A , Sehlin J. Insulin-stimulated chemokinesis in normal human neutrophils is dependent on D-glucose concentration and sensitive to inhibitors of tyrosine kinase and phosphatidylinositol 3-kinase.

Pillinger MH , Feoktistov AS , Capodici C , et al. Mitogen-activated protein kinase in neutrophils and enucleate neutrophil cytoplasts: evidence for regulation of cell-cell adhesion.

Zhang Z , Amorosa LF , Coyle SM , et al. Insulin-dependent regulation of mTORC2-Akt-FoxO suppresses TLR4 signaling in human leukocytes: relevance to Type 2 diabetes.

Walrand S , Guillet C , Boirie Y , Vasson MP. In vivo evidences that insulin regulates human polymorphonuclear neutrophil functions. Kappert K , Meyborg H , Clemenz M , et al. Insulin facilitates monocyte migration: a possible link to tissue inflammation in insulin-resistance.

Biochem Biophys Res Commun. Insulin regulates monocyte trans-endothelial migration through surface expression of macrophage-1 antigen.

Liu Y , Dhall S , Castro A , Chan A , Alamat R , Martins-Green M. Biol Open. Bunn RC , Cockrell GE , Ou Y , Thrailkill KM , Lumpkin CK , Fowlkes JL.

Palmitate and insulin synergistically induce IL-6 expression in human monocytes. Cardiovasc Diabetol. Alberts B , Johnson A , Lewis J.

Limphocytes and the cellular basis of adaptative immunity. In: Alberts B , Bray D , Lewis J , Raff M , Roberts K , Watson JD , eds. Molecular Biology of Cell. Garland Science , Eosinophil inversely associates with type 2 diabetes and insulin resistance in Chinese adults. Hübner MP , Larson D , Torrero MN , et al.

Anti-FcεR1 antibody injections activate basophils and mast cells and delay Type 1 diabetes onset in NOD mice. Clin Immunol. Hirai K , Miyamasu M , Yamaguchi M , et al. Modulation of human basophil histamine release by insulin-like growth factors.

Kettner A , Di Matteo M , Santoni A. Insulin potentiates FcɛRI-mediated signaling in mouse bone marrow-derived mast cells. Mol Immunol. Lessmann E , Grochowy G , Weingarten L , et al. Insulin and insulin-like growth factor-1 promote mast cell survival via activation of the phosphatidylinositolkinase pathway.

Exp Hematol. Walrand Sp , Guillet C , Gachon P , et al. Insulin regulates protein synthesis rate in leukocytes from young and elderly healthy humans.

Freeman A , Pennings N. Insulin Resistance. Moller DE , Kaufman KD. Metabolic syndrome: a clinical and molecular perspective. Annu Rev Med. Wilcox G. Insulin and insulin resistance.

Clin Biochem Rev. Cerf ME. Beta cell dysfunction and insulin resistance. Front Endocrinol Lausanne. Dandona P , Aljada A , Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes.

Trends Immunol. Pinto-Junior DC , Silva KS , Michalani ML , et al. Advanced glycation end products-induced insulin resistance involves repression of skeletal muscle GLUT4 expression. Sci Rep. Unoki H , Yamagishi S.

Advanced glycation end products and insulin resistance. Curr Pharm Des. Vazquez-Jimenez G , Chavez-Reyes J , Romero-Garcia T , et al. Palmitic acid but not palmitoleic acid induces insulin resistance in a human endothelial cell line by decreasing SERCA pump expression.

Cell Signal. Fazakerley DJ , Minard AY , Krycer JR , et al. Mitochondrial oxidative stress causes insulin resistance without disrupting oxidative phosphorylation. Hurrle S , Hsu WH. The etiology of oxidative stress in insulin resistance. Biomed J. Ohta Y , Kinugawa S , Matsushima S , et al.

Oxidative stress impairs insulin signal in skeletal muscle and causes insulin resistance in postinfarct heart failure. Am J Physiol Heart Circ Physiol. Boden G , Duan X , Homko C , et al. Increase in endoplasmic reticulum stress-related proteins and genes in adipose tissue of obese, insulin-resistant individuals.

Chan SMH , Sun R-Q , Zeng X-Y , et al. Activation of PPARα ameliorates hepatic insulin resistance and steatosis in high fructose-fed mice despite increased endoplasmic reticulum stress.

Villalobos-Labra R , Subiabre M , Toledo F , Pardo F , Sobrevia L. Endoplasmic reticulum stress and development of insulin resistance in adipose, skeletal, liver, and foetoplacental tissue in diabesity.

Mol Aspects Med. Lasram MM , Dhouib IB , Annabi A , El Fazaa S , Gharbi N. A review on the molecular mechanisms involved in insulin resistance induced by organophosphorus pesticides. Organophosphorus pesticide chlorpyrifos intake promotes obesity and insulin resistance through impacting gut and gut microbiota.

Andersen CJ , Murphy KE , Fernandez ML. Impact of obesity and metabolic syndrome on immunity. Adv Nutr. Geerlings SE , Hoepelman AIM.

Immune dysfunction in patients with diabetes mellitus DM. FEMS Immunol Med Microbiol. Kunjathoor VV , Febbraio M , Podrez EA , et al.

Zingg JM , Ricciarelli R , A A. Scavenger receptors and modified lipoproteins: fatal attractions? IUBMB Life. Kashyap SR , Ioachimescu AG , Gornik HL , et al. Lipid-induced insulin resistance is associated with increased monocyte expression of scavenger receptor CD36 and internalization of oxidized LDL.

Sarigianni M , Bekiari E , Tsapas A , et al. Effect of leptin and insulin resistance on properties of human monocytes in lean and obese healthy participants. de Matos MA , Duarte TC , Ottone V , et al.

Cell Biochem Funct. Szegezdi E , Logue SE , Gorman AM , Samali A. Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep. Senokuchi T , Liang C-P , Seimon TA , et al. Forkhead transcription factors FoxOs promote apoptosis of insulin-resistant macrophages during cholesterol-induced endoplasmic reticulum stress.

Han S , Liang C-P , DeVries-Seimon T , et al. Macrophage insulin receptor deficiency increases ER stress-induced apoptosis and necrotic core formation in advanced atherosclerotic lesions. Liang C-P , Han S , Li G , Tabas I , Tall AR. Impaired MEK signaling and SERCA expression promote ER stress and apoptosis in insulin-resistant macrophages and are reversed by exenatide treatment.

Tabas I , Tall A , Accili D. The impact of macrophage insulin resistance on advanced atherosclerotic plaque progression. Circ Res. Ieronymaki E , Theodorakis EM , Lyroni K , et al. Insulin resistance in macrophages alters their metabolism and promotes an M2-Like phenotype.

Ieronymaki E , Daskalaki MG , Lyroni K , Tsatsanis C. Insulin signaling and insulin resistance facilitate trained immunity in macrophages through metabolic and epigenetic changes.

Naghibi M , Smith RP , Baltch AL , et al. The effect of diabetes mellitus on chemotactic and bactericidal activity of human polymorphonuclear leukocytes. Shetty N , Thomas B , Ramesh A.

Comparison of neutrophil functions in diabetic and healthy subjects with chronic generalized periodontitis. J Indian Soc Periodontol. Brotfain E , Hadad N , Shapira Y , et al. Neutrophil functions in morbidly obese subjects. Clin Exp Immunol. Trottier MD , Naaz A , Kacynski K , Yenumula PR , Fraker PJ.

Functional capacity of neutrophils from class III obese patients. Kim SH , Reaven GM. Insulin resistance and hyperinsulinemia: you can't have one without the other. Diabetes Care. Neethi Raj P , Shaji BV , Haritha VH , Anie Y.

J Cell Immunother. Stegenga ME , van der Crabben SN , Dessing MC , et al. Diabet Med. Berbudi A , Rahmadika N , Tjahjadi AI , Ruslami R. Type 2 diabetes and its impact on the immune system. Casqueiro J , Casqueiro J , Alves C. Infections in patients with diabetes mellitus: a review of pathogenesis.

Indian J Endocrinol Metab. Dronge AS , Perkal MF , Kancir S , Concato J , Aslan M , Rosenthal RA. Long-term glycemic control and postoperative infectious complications.

Arch Surg. Tsalamandris S , Antonopoulos AS , Oikonomou E , et al. The role of inflammation in diabetes: current concepts and future perspectives. Eur Cardiol. Van den Berghe G , Wilmer A , Milants I , et al. Vatankhah N , Jahangiri Y , Landry GJ , Moneta GL , Azarbal AF.

Effect of systemic insulin treatment on diabetic wound healing. Wound Repair Regen. UK Prospective Diabetes Study UKPDS Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes UKPDS Mowat AG , Baum J.

Chemotaxis of polymorphonuclear leukocytes from patients with diabetes mellitus. N Engl J Med. Yano H , Kinoshita M , Fujino K , et al. Insulin treatment directly restores neutrophil phagocytosis and bactericidal activity in diabetic mice and thereby improves surgical site Staphylococcus aureus infection.

Infect Immun. Ebtehaj S , Gruppen EG , Parvizi M , Tietge UJ , Dullaart RP. The anti-inflammatory function of HDL is impaired in type 2 diabetes: role of hyperglycemia, paraoxonase-1 and low grade inflammation. Kengne AP , Batty GD , Hamer M , Stamatakis E , Sb Czernichow.

Association of C-reactive protein with cardiovascular disease mortality according to diabetes status: pooled analyses of 25, participants from four U.

prospective cohort studies. Mirza S , Hossain M , Mathews C , et al. Type 2-diabetes is associated with elevated levels of TNF-alpha, IL-6 and adiponectin and low levels of leptin in a population of Mexican Americans: a cross-sectional study.

Biochem Biophys. Res Commun. Etgen, G. Jr et al. Exercise training reverses insulin resistance in muscle by enhanced recruitment of GLUT-4 to the cell surface. Ryder, J. Use of a novel impermeable biotinylated photolabeling reagent to assess insulin-and hypoxia-stimulated cell surface GLUT4 content in skeletal muscle from type 2 diabetic patients.

Garvey, W. Multiple defects in the adipocyte glucose transport system cause cellular insulin resistance in gestational diabetes: heterogeneity in the number and a novel abnormality in subcellular localization of GLUT4 glucose transporters. Diabetes 42 , — Chadt, A. Deletion of both Rab-GTPase—activating proteins TBC14KO and TBC1D4 in mice eliminates insulin-and AICAR-stimulated glucose transport.

Diabetes 64 , — Chen, S. Tramunt, B. Sex differences in metabolic regulation and diabetes susceptibility. Diabetologia 63 , — Greenhill, C. Sex differences in insulin resistance. Qiu, J. Estradiol protects proopiomelanocortin neurons against insulin resistance.

Endocrinology , — Zidon, T. Effects of ERβ and ERα on OVX-induced changes in adiposity and insulin resistance. Ikeda, K. Functions of estrogen and estrogen receptor signaling on skeletal muscle.

Steroid Biochem Mol. Gerdts, E. Sex differences in cardiometabolic disorders. Chia, C. Age-related changes in glucose metabolism, hyperglycemia, and cardiovascular risk.

Shi, H. Sex differences in obesity-related glucose intolerance and insulin resistance. Glucose Tolerance 4 , 37—66 Google Scholar. Geer, E. Gender differences in insulin resistance, body composition, and energy balance.

Christen, T. Sex differences in body fat distribution are related to sex differences in serum leptin and adiponectin. Peptides , 25—31 Palmisano, B. Sex differences in lipid and lipoprotein metabolism. Kodama, K. Ethnic differences in the relationship between insulin sensitivity and insulin response: a systematic review and meta-analysis.

Diabetes Care 36 , — Raygor, V. Diab Vasc. Sumner, A. Ethnic differences in the ability of triglyceride levels to identify insulin resistance. Atherosclerosis , — Tan, V. Ethnic differences in insulin sensitivity and beta-cell function among Asian men.

Diabetes 5 , e—e Ministry of Health Singapore MOHS. Potts, J. Sex and ethnic group differences in fat distribution in young United Kingdom South Asians and Europids. Ehtisham, S. Ethnic differences in insulin resistance and body composition in United Kingdom adolescents.

Lear, S. Ethnic variation in fat and lean body mass and the association with insulin resistance. Mason, C. Dietary weight loss and exercise effects on insulin resistance in postmenopausal women. Med 41 , — Mikusova, V. Insulin resistance and need for a lifestyle change to eliminate it.

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

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

Diabetes 61 , — Kan, C. A systematic review and meta-analysis of the association between depression and insulin resistance. Diabetes care 36 , — Sung, C. Role of vitamin D in insulin resistance. Ardabili, H. Vitamin D supplementation has no effect on insulin resistance assessment in women with polycystic ovary syndrome and vitamin D deficiency.

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

Effects of growth hormone on insulin action in man: mechanisms of insulin resistance, impaired suppression of glucose production, and impaired stimulation of glucose utilization. Diabetes 31 , — Barbour, L.

A Cellular mechanisms for insulin resistance in normal pregnancy and gestational diabetes. Diabetes Care 30 , S—S Parichatikanond, W. Prolonged stimulation of β2-adrenergic receptor with β2-agonists impairs insulin actions in H9c2 cells.

Walli, R. Treatment with protease inhibitors associated with peripheral insulin resistance and impaired oral glucose tolerance in HIVinfected patients.

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

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

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

Matsumoto, M. Dual role of transcription factor FoxO1 in controlling hepatic insulin sensitivity and lipid metabolism. Shimomura, I. Cell 6 , 77—86 Petersen, K.

Mechanism by which glucose and insulin inhibit net hepatic glycogenolysis in humans. Henriksen, E. Role of glycogen synthase kinase-3 in insulin resistance and type 2 diabetes. Drug Targets 7 , — Karim, S. Hepatic expression and cellular distribution of the glucose transporter family.

World J. Rencurel, F. Requirement of glucose metabolism for regulation of glucose transporter type 2 GLUT2 gene expression in liver.

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

Ferré, P. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes. Tobe, K. Dentin, R. Carbohydrate responsive element binding protein ChREBP and sterol regulatory element binding protein-1c SREBP-1c : two key regulators of glucose metabolism and lipid synthesis in liver.

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

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

Regulation of triglyceride metabolism. Hormonal regulation of lipolysis in adipose tissue. Liver Physiol. Vaughan, M.

Hormone-sensitive lipase and monoglyceride lipase activities in adipose tissue. Zmuda-Trzebiatowska, E. Role of PDE3B in insulin-induced glucose uptake, GLUT-4 translocation and lipogenesis in primary rat adipocytes.

Cell Signal 18 , — Choi, Y. Alterations in regulation of energy homeostasis in cyclic nucleotide phosphodiesterase 3B—null mice.

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

USA 98 , — Mechanisms of Insulin Action and Insulin Resistance. Kimball, S. Regulation of protein synthesis by insulin.

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

Rudrappa, S. Human skeletal muscle disuse atrophy: effects on muscle protein synthesis, breakdown, and insulin resistance-a qualitative review. Front Physiol. Medeiros, C. Antuna-Puente, B. Adipokines: the missing link between insulin resistance and obesity. Diabetes Metab. Rabe, K.

Adipokines and insulin resistance. Adipokines mediate inflammation and insulin resistance. Lausanne 4 , 71 Li, S. Adiponectin levels and risk of type 2 diabetes: a systematic review and meta-analysis. JAMA , — Hotta, K.

Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 50 , — Takahashi, M. Chemerin enhances insulin signaling and potentiates insulin-stimulated glucose uptake in 3T3-L1 adipocytes.

FEBS Lett. Yamauchi, T. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Li, L. Changes and relations of circulating visfatin, apelin, and resistin levels in normal, impaired glucose tolerance, and type 2 diabetic subjects.

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

Endocrinology , 59—68 Segal, K. Relationship between insulin sensitivity and plasma leptin concentration in lean and obese men.

Amitani, M. The role of leptin in the control of insulin-glucose axis. Front Neurosci. Article PubMed PubMed Central CAS Google Scholar. Ceddia, R. Analysis of paradoxical observations on the association between leptin and insulin resistance. Covey, S. The pancreatic beta cell is a key site for mediating the effects of leptin on glucose homeostasis.

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

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

Diabetes 55 , — Saponaro, C. The subtle balance between lipolysis and lipogenesis: a critical point in metabolic homeostasis. Nutrients 7 , — Frayn, K. Adipose tissue and the insulin resistance syndrome. Roden, M.

Mechanism of free fatty acid-induced insulin resistance in humans. Invest 97 , — Goossens, G. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance.

RANDLE, P. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1 , — Randle, P. The glucose fatty acid cycle in obesity and maturity onset diabetes mellitus. Y Acad. Brechtel, K.

Fast elevation of the intramyocellular lipid content in the presence of circulating free fatty acids and hyperinsulinemia: a dynamic 1H-MRS study.

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

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

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

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

Muscular diacylglycerol metabolism and insulin resistance. Liu, L. Upregulation of myocellular DGAT1 augments triglyceride synthesis in skeletal muscle and protects against fat-induced insulin resistance.

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

Med Biol. Stöckli, J. Metabolomic analysis of insulin resistance across different mouse strains and diets. Blachnio-Zabielska, A.

The crucial role of CCer in fat-induced skeletal muscle insulin resistance. Biochem 40 , — Raichur, S. CerS2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance.

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

PLoS One 8 , e Brown, A. Genetics of Insulin Resistance and the Metabolic Syndrome. Chen, Z. Functional Screening of Candidate Causal Genes for Insulin Resistance in Human Preadipocytes and Adipocytes.

Shakhanova, A. Association of polymorphism genes LPL , ADRB2 , AGT and AGTR1 with risk of hyperinsulinism and insulin resistance in the Kazakh population. Henkin, L. Genetic epidemiology of insulin resistance and visceral adiposity. The IRAS Family Study design and methods.

Parks, B. Genetic architecture of insulin resistance in the mouse. Xue, A. Genome-wide association analyses identify risk variants and putative regulatory mechanisms for type 2 diabetes. Yousef, A. IRS-1 genetic polymorphism r. Appl Clin. Genet 11 , 99— Hashemian, L.

The role of the PPARG Pro12Ala common genetic variant on type 2 diabetes mellitus risk. Zeng, Q. Association Between Insulin-like Growth Factor-1 rs Polymorphism and Type 2 Diabetes Mellitus Susceptibility: A Meta-Analysis.

Front Genet 12 , Knowles, J. Identification and validation of N-acetyltransferase 2 as an insulin sensitivity gene. Invest , Richards, J. A genome-wide association study reveals variants in ARL15 that influence adiponectin levels. PLoS Genet 5 , e Eichler, E. Missing heritability and strategies for finding the underlying causes of complex disease.

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

Metz, S. The Arg82Cys polymorphism of the protein nepmucin implies a role in HDL metabolism. Moltke, I. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Huypens, P. Epigenetic germline inheritance of diet-induced obesity and insulin resistance.

Genet 48 , — Ling, C. Epigenetics in human obesity and type 2 diabetes. Ahmed, S. The role of DNA methylation in the pathogenesis of type 2 diabetes mellitus.

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

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

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

Wittenbecher, C. Insulin-like growth factor binding protein 2 IGFBP-2 and the risk of developing type 2 diabetes.

Diabetes 68 , — Haywood, N. Crujeiras, A. Genome-wide DNA methylation pattern in visceral adipose tissue differentiates insulin-resistant from insulin-sensitive obese subjects. Res , 13—24 Ma, X. Functional implications of DNA methylation in adipose biology. Gao, H. Long non-coding RNAs associated with metabolic traits in human white adipose tissue.

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

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

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

Goedeke, L. MicroRNAa regulates LDL receptor and ABCA1 expression to control circulating lipoprotein levels. Soh, J. MicroRNAc reduces hyperlipidemia and atherosclerosis in mice by decreasing lipid synthesis and lipoprotein secretion. Salerno, A. LDL Receptor Pathway Regulation by miR and miRd.

Front Cardiovasc Med. Miranda, K. MicroRNA modulates metabolic inflammation by regulating Notch signaling in adipose tissue macrophages. Wang, Y.

Natural killer cell-derived exosomal miRp attenuates insulin resistance and inflammation in mouse models of type 2 diabetes.

Signal Transduct. Target Ther. Quinn, J. Unique features of long non-coding RNA biogenesis and function. Genet 17 , 47—62 Squillaro, T.

Long non-coding RNAs in regulation of adipogenesis and adipose tissue function. Elife 9 , e Zhang, T.

Regulation of glucose and lipid metabolism by long non-coding RNAs: facts and research progress. Lausanne 11 , Yang, S. LncRNA SRA deregulation contributes to the development of atherosclerosis by causing dysfunction of endothelial cells through repressing the expression of adipose triglyceride lipase.

Med Rep. Zhang, N. Elevated hepatic expression of H19 long noncoding RNA contributes to diabetic hyperglycemia. JCI insight 3 , e Article PubMed Central Google Scholar. Gui, W. LncRNAH19 improves insulin resistance in skeletal muscle by regulating heterogeneous nuclear ribonucleoprotein A1.

Cell Commun. Akerman, I. Human pancreatic β cell lncRNAs control cell-specific regulatory networks. Xiong, L. LncRNA-Malat1 is involved in lipotoxicity-induced ß-cell dysfunction and the therapeutic effect of exendin-4 via Ptbp1. Ruan, Y. Circulating LncRNAs analysis in patients with type 2 diabetes reveals novel genes influencing glucose metabolism and Islet β-cell function.

Elizabeth Mann 1Promotinv Sunni 2and Melena Skinfold measurement accuracy. Performance nutrition for triathletes 2. The pancreas is a complex gland active in functlon and metabolism through secretion functino digestive enzymes Peomoting the exocrine portion and hormones from the funcfion portion. The Flavonoids and cardiovascular health Skinfold measurement accuracy islets was Skinfold measurement accuracy by Paul Langerhans inand the functional role of islets in glucose homeostasis was first demonstrated in when Joseph von Mering and colleagues showed that dogs developed diabetes mellitus following pancreatectomy Though islet mass may vary between individuals—an example is the increase in the setting of adult obesity 83 — the average adult human pancreas is estimated to contain one to two million islets 33, In humans, the concentration of islets is up to two times higher in the tail compared to the head and neck.

Author: Meztikora

1 thoughts on “Promoting insulin function

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com