Category: Family

Insulin and carbohydrate metabolism

Insulin and carbohydrate metabolism

analysed Insulin and carbohydrate metabolism omics meabolism. When on the institution site, please use the metabollism provided by your institution. More from Oxford Academic. Search for:. Article ADS PubMed PubMed Central Google Scholar. Pentose phosphate pathway Fructolysis Polyol pathway Galactolysis Leloir pathway.

Ever since Insilin metabolic fate of carbohydrates in the body has carvohydrate associated with the mstabolism of carbohydrtae, biochemists have Insulin and carbohydrate metabolism concerned carrbohydrate the question as Dextrose Workout Recovery how the pancreatic hormone affects Insulin and carbohydrate metabolism so as to render it utilizable.

Macleod 1 carbohydtate realized that Insulun very carbohydrste fall in carbohydfate sugar that sets in almost immediately after injection of insulin suggests that some carbohyrrate occurring in the blood itself must be responsible for it—an increased mehabolism.

There Insulin and carbohydrate metabolism widespread agreement among investigators, however, Insulin and carbohydrate metabolism insulin Forskolin and immune system not directly influence the rate of glycolysis or sugar decomposition.

A second explanation, which also was suggested early in the investigation of insulin, involves the possibility that it might have some influence on the properties of sugars so as to render them more labile in metabolism.

The stereochemical character of glucose lends tenability to such an hypothesis. Several investigators, in fact, believed that they. Artificial Intelligence Resource Center. Featured Clinical Reviews Screening for Atrial Fibrillation: US Preventive Services Task Force Recommendation Statement JAMA.

X Facebook LinkedIn. This Issue. Share X Facebook Email LinkedIn. January 16, visual abstract icon Visual Abstract. Access through your institution. Add or change institution. Select Your Interests Customize your JAMA Network experience by selecting one or more topics from the list below.

Save Preferences. Privacy Policy Terms of Use. Access your subscriptions. Free access to newly published articles. Purchase access. Rent article Rent this article from DeepDyve. Sign in to access free PDF. Save your search. Customize your interests. Create a personal account or sign in to:.

Privacy Policy. Make a comment.

: Insulin and carbohydrate metabolism

Carbohydrates and Blood Sugar | The Nutrition Source | Harvard T.H. Chan School of Public Health indistinctus JCMA. Plasma cytokines Insulin and carbohydrate metabolism snd 10 -transformed and scaled Insuljn the Carbohydrae analyses. Glycero- phospholipids. Skip to main content Thank you for visiting nature. Maegawa H, Hasegawa M, Sugai S, Obata T, Ugi S, Morino K, Egawa K, Fujita T, Sakamoto T, Nishio Y, Kojima H, Haneda M, Yasuda H, Kikkawa R, Kashiwagi A Expression of a dominant negative SHP-2 in transgenic mice induces insulin resistance.
HISTORICAL PERSPECTIVE Kelleher JK Estimating gluconeogenesis with [UC]glucose: molecular condensation requires a molecular approach. This Feature Is Available To Subscribers Only Sign In or Create an Account. Notably, different genera and species correlated with other clinical markers, suggesting that the individual association between microbial taxa and clinical manifestation is not as robust as in the co-abundance analysis. Never disregard professional medical advice or delay in seeking it because of something you have read on this website. Tayek JA, Katz J Glucose production, recycling, and gluconeogenesis in normals and diabetics: a mass isotopomer [UC]glucose study.
MeSH terms

Save Preferences. Privacy Policy Terms of Use. Access your subscriptions. Free access to newly published articles. Purchase access. Rent article Rent this article from DeepDyve. Sign in to access free PDF. Save your search. Customize your interests.

Growth hormone decreases glucose uptake in muscle and adipose tissue gluconeogenesis in liver. In the presence of insulin, growth hormone stimulates protein synthesis. The net metabolic effects of a single hormone are directly related to the activity of other synergistic or antagonistic hormones.

Abstract Insulin is the key hormone of carbohydrate metabolism, it also influences the metabolism of fat and proteins. den Besten, G. Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids.

Liver Physiol. Article Google Scholar. Zierer, J. The fecal metabolome as a functional readout of the gut microbiome. Lloyd-Price, J. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Article ADS CAS PubMed PubMed Central Google Scholar.

Hui, D. Intestinal phospholipid and lysophospholipid metabolism in cardiometabolic disease. Tsugawa, H. A lipidome atlas in MS-DIAL 4. Yasuda, S. iScience 23 , Erion, D. Diacylglycerol-mediated insulin resistance. An, D.

Sphingolipids from a symbiotic microbe regulate homeostasis of host intestinal natural killer T cells. Cell , — Claesson, M. Gut microbiota composition correlates with diet and health in the elderly. Liu, R. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention.

Piening, B. Integrative personal omics profiles during periods of weight gain and loss. Cell Syst. Ridaura, V. Gut microbiota from twins discordant for obesity modulate metabolism in mice.

Science , Flint, H. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 3 , — Article PubMed PubMed Central Google Scholar. Vacca, M. The controversial role of human gut Lachnospiraceae. Microorganisms 8 , David, L. Diet rapidly and reproducibly alters the human gut microbiome.

Deutscher, J. How phosphotransferase system-related protein phosphorylation regulates carbohydrate metabolism in bacteria. Flores, R. Association of fecal microbial diversity and taxonomy with selected enzymatic functions. PLoS ONE 7 , e Cani, P. Metabolic endotoxemia initiates obesity and insulin resistance.

Diabetes 56 , — Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet—induced obesity and diabetes in mice.

Diabetes 57 , — Rajbhandari, P. IL signaling remodels adipose chromatin architecture to limit thermogenesis and energy expenditure. Beppu, L. JCI Insight 6 , e Acosta, J. Human-specific function of IL in adipose tissue linked to insulin resistance.

Tingley, D. mediation: R package for causal mediation analysis. Dekker, M. Fructose: a highly lipogenic nutrient implicated in insulin resistance, hepatic steatosis, and the metabolic syndrome. Baig, S. Genes involved in oxidative stress pathways are differentially expressed in circulating mononuclear cells derived from obese insulin-resistant and lean insulin-sensitive individuals following a single mixed-meal challenge.

Dasu, M. High glucose induces toll-like receptor expression in human monocytes: mechanism of activation. Hannou, S. Fructose metabolism and metabolic disease.

Chang, C. Posttranscriptional control of T cell effector function by aerobic glycolysis. Matsuzawa, Y. Metabolic syndrome—definition and diagnostic criteria in Japan. Vidigal, F. Prevalence of metabolic syndrome and pre-metabolic syndrome in health professionals: LATINMETS Brazil study. Sato, K. Obesity-related gut microbiota aggravates alveolar bone destruction in experimental periodontitis through elevation of uric acid.

mBio 12 , e Takeuchi, T. Acetate differentially regulates IgA reactivity to commensal bacteria. Methods 12 , — Langfelder, P. WGCNA: an R package for weighted correlation network analysis. BMC Bioinform. Xia, J. MetaboAnalyst: a web server for metabolomic data analysis and interpretation.

Nucleic Acids Res. Milanese, A. Microbial abundance, activity and population genomic profiling with mOTUs2. Article ADS PubMed PubMed Central Google Scholar. Nishijima, S. The gut microbiome of healthy Japanese and its microbial and functional uniqueness.

DNA Res. Li, J. An integrated catalog of reference genes in the human gut microbiome. Cantarel, B. The carbohydrate-active EnZymes database CAZy : an expert resource for glycogenomics.

Kouno, T. C1 CAGE detects transcription start sites and enhancer activity at single-cell resolution. Salimullah, M. NanoCAGE: a high-resolution technique to discover and interrogate cell transcriptomes. Cold Spring Harb. prot Hasegawa, A. MOIRAI: a compact workflow system for CAGE analysis.

Frankish, A. GENCODE reference annotation for the human and mouse genomes. Article PubMed Central Google Scholar. Forrest, A. A promoter-level mammalian expression atlas. Chen, E. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. Kuleshov, M.

Enrichr: a comprehensive gene set enrichment analysis web server update. Kubota, T. Downregulation of macrophage Irs2 by hyperinsulinemia impairs ILindeuced M2a-subtype macrophage activation in obesity.

Impaired insulin signaling in endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. Kubota, N. Dynamic functional relay between insulin receptor substrate 1 and 2 in hepatic insulin signaling during fasting and feeding. Kloke, J. Rfit: rank-based estimation for linear models.

Gevers, D. Cell Host Microbe 15 , — Shannon, P. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Wang, D. Characterization of gut microbial structural variations as determinants of human bile acid metabolism.

Cell Host Microbe 29 , — Download references. We thank E. Miyauchi, T. Kanaya and T. Kato for advice; A. Ito, N. Tachibana, A. Hori and the staff at the RIKEN Yokohama animal facility for technical support; H. Koseki, M. Furuno and H. Iwano for data discussion; and the staff at the RIKEN BioResource Research Center for providing essential materials.

Kubota, 21K to H. and 22H to H. and M. Kubota and the RIKEN Junior Research Associate Program to T. Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan. Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.

Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. Division of Diabetes and Metabolism, The Institute for Medical Science Asahi Life Foundation, Tokyo, Japan. Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition NIBIOHN , Tokyo, Japan.

Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science CSRS , Yokohama, Japan. Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan.

Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan. Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan.

Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan. Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan. Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan.

Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan. Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan. Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan.

Institute for Advanced Biosciences, Keio University, Fujisawa, Japan. Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.

Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan. Center for Epidemiology and Preventive Medicine, The University of Tokyo Hospital, Tokyo, Japan. International University of Health and Welfare, Tokyo, Japan. Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, Ami Town, Japan.

Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan. Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan.

Human Biology-Microbiome-Quantum Research Center WPI-Bio2Q , Keio University, Tokyo, Japan. Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences IMS , Yokohama, Japan.

You can also search for this author in PubMed Google Scholar. Kadowaki and H. conceived the project. Kubota, Y. Mizuno, N. and T. Kadowaki contributed to the enrolment of study participants and clinical data collection.

and Y. processed faecal samples for metagenomics and metabolomic analyses. performed 16S rRNA gene sequencing and metagenomic analysis. performed metabolomic analyses for hydrophilic metabolites. performed lipidomics analyses.

and P. performed CAGE analysis. and O. performed cytokine measurement and RNA extraction from PBMCs. Mochizuki prepared fundamental information tools for the analysis. Kubota and S. performed animal experiments and analysed the data. Kitami and K.

analysed the omics data. Kubota, P. and H. provided essential materials and raised funding. Kubota and H. wrote the paper together with A. Kitami and P. Correspondence to Tetsuya Kubota or Hiroshi Ohno.

are listed as the inventors on a patent regarding the metabolic effects of gut bacteria identified by a human cohort. The other authors declare no competing interests.

Nature thanks Gregory Steinberg and the other, anonymous, reviewer s for their contribution to the peer review of this work. Insulin resistance IR and metabolic syndrome MetS were the main clinical phenotypes.

To evaluate the host-microbe relationship, we collected 1 host factors: clinical, plasma metabolome, peripheral blood mononuclear cells PBMC transcriptome, and cytokine data, and 2 microbial factors: 16S rRNA pyrosequencing, shotgun metagenome, and faecal metabolome.

The numbers of elements after quality filtering are shown for each data set. b , The multi-omics analysis workflow. To identify the microbes that affect metabolic phenotypes, we first analysed the phenotype-associated metabolomic signatures by binning metabolites into co-abundance groups CAGs.

Microbial signatures were determined using the 16S and metagenomic datasets, and their associations with metabolites were analysed. We also assessed the mediation effects of plasma cytokines on the relationships between faecal metabolites and metabolic markers.

The associations between clinical phenotypes and omics markers were adjusted by age and sex wherever appropriate.

a , The KEGG pathway enrichment analysis of the metabolites in hydrophilic CAGs 5, 8, 12, 15, and 18, which were associated with IR in Fig. The size of disks shows the enrichment i.

b , Partial correlations between HOMA-IR and faecal levels of short-chain fatty acids SCFA such as acetate, propionate, and butyrate left panel , and disaccharides such as maltose and sucrose right panel.

Density plots indicate median and distribution. The detailed statistics are reported in Supplementary Table 5 , 6.

The size and colour of the disks represent the estimate and the direction of the associations. c , The associations between faecal glucose and arabinose and HOMA-IR as analysed in Fig. The estimates of metabolites and their P values are described. The data were analysed with a generalized linear mixed-effect model with consent age and sex as fixed effects, and the sample collection site as a random effect.

The estimate and P value are described. The first faecal sampling for metabolomics was used to avoid redundancy. The detailed statistics are reported in Supplementary Table 9. Dots represent individual data summarized into PCo1 and PCo2.

Dots represent individual data summarized into PC1 and PC2. f , Co-abundance groups of genus-level microbes and their abundance in the participant clusters defined in Fig. The disk size represents the median abundance in the participants. g , The co-abundance groups of genus-level microbes and their abundance in the participant clusters.

The size of the disks represents overabundance to the mean in four clusters of participants determined in Fig. The far-left column shows the genera that exhibit significant differences among the four clusters.

The genera forming distinct groups in f , i. The participants were clustered into three mOTU clusters A to C based on the heatmap clustering. The proportion of individuals with IS, intermediate, and IR are shown in the pie charts above the heatmap as Fig.

Only those with significant associations with metabolic markers are depicted. The disk size and colour represent absolute values of standardized coefficient and the direction of associations.

The detailed statistics are reported in Supplementary Table j , Microbe-metabolite networks of IR- or and IS-associated co-abundance microbial groups from Fig. All faecal hydrophilic metabolites and faecal microbe-related lipid metabolites were included in the analysis.

The metabolites in CAGs relating to carbohydrates shown in Fig. k , The relative abundance of IR-associated faecal carbohydrates in the participant clusters. The metabolites significantly different among these four clusters are coloured grey in the top row.

a , b , Box plots indicate the median, upper and lower quartiles, and upper and lower extremes except for outliers. Kruskal-Wallis test g , k.

See the Source Data g for exact P values. a , b , The associations between the KEGG pathways relating to amino acid metabolism a and lipid metabolism b , faecal carbohydrates, top three genera positively or negatively correlated with faecal carbohydrates in Fig.

c , The associations between representative metabolic markers and the KEGG pathways relating to carbohydrate metabolism, amino acid metabolism, lipid metabolism, and membrane transport defined in the KEGG orthology database. The pathways with significant associations with metabolic markers are included in the plots.

The far-left column shows the type of carbohydrate metabolites that each PTS gene is involved in. The far-left column shows whether the genes were predicted to function as extracellular enzymes. g , Representative pathways in starch and sucrose metabolism KEGG pathway relating to glycosidase activities to degrade poly- and oligosaccharides into monosaccharides.

i , The presence and absence of KEGG orthologues predicted to function as extracellular enzymes in 45 strains. The strains from the top three genera positively or negatively correlated with faecal carbohydrates shown in Fig. Density plots indicate median and distribution e , h. a , Cell-type gene set enrichment analysis based on the Human Gene Atlas database using Enrichr.

Red and blue colour scales represent IR and IS-associated cell types, respectively please refer to Methods for details. b , The cross-omics network shown in Fig.

c , The number of correlations between faecal carbohydrates and other omics elements shown in Fig. The proportion to all possible correlations is shown. d , Representative causal mediation models analysing the effects of IL and adiponectin mediating in silico relationships between faecal carbohydrates and HOMA-IR.

Causal mediation analysis with multiple test corrections were used to test significance. Estimates β and P adj values of average causal mediation effects ACME , which are the indirect effects between the metabolites and host markers mediated by cytokines, and average direct effects ADE , which are the direct effects controlling for cytokines, are described.

Age and sex were adjusted in the models. The detailed information is reported in Supplementary Table a , b , PCA plots of metabolites in cell-free supernatants of 22 bacterial strains listed in a.

These strains were selected based on the findings from the genus-level co-occurrence Fig. The strains from genera and species relating to IR-related markers shown in Extended Data Fig. The top 10 metabolites contributing to the PCA separation left panel and 13 out of 15 IR-related carbohydrates identified in Fig.

c , d , The levels of carbohydrate fermentation products c and carbohydrates relating to IR in the human cohort d in the cell-free supernatants. e , Pie charts summarizing the consumption and production of carbohydrates shown in d.

Those significantly decreased or increased compared with the vehicle control group were considered as consumption or production. f , The top consumers of carbohydrates, which summarizes the results shown in e.

Representative data of two independent experiments. c , d , Data are mean and s. a , Body mass change from the baseline. indistinctus AI groups, respectively. Pooled data of three independent experiments. Pooled data of two independent experiments.

k , l , Representative images of phosphorylated Akt p-Akt at S and total Akt in the liver and epidydimal fat eWAT in mice administered Alistipes indistinctus AI , Alistipes finegoldii AF , and PBS as vehicle control k. The raw images of blotting membranes are shown in Supplementary Fig.

P values for interactions between time and group are described in m. Other metabolic measures are reported in Supplementary Table Representative data of two independent experiments c—g , k—o.

a , Density plots indicate median and distribution. a , PCA plots of metabolites in caecal contents of AI-administered mice. The top 10 metabolites contributing to the PCA separation left panel and 12 out of 15 IR-related carbohydrates identified in Fig. b , The PC1 of PCA plots in Fig.

The detailed statistics of all caecal metabolites are reported in Supplementary Table e , A schematic summary. In this study, we combined faecal metabolome, 16S rRNA gene sequencing, and metagenome data with host metabolome, transcriptome, and cytokine data to comprehensively delineate the involvement of gut microbiota in IR upper panel.

Carbohydrate degradation products such as monosaccharides are prominently increased in IR middle panel. Metagenomic findings show that the degradation and utilization of poly- and disaccharides are facilitated in IR and that these microbial functions are strongly associated with faecal monosaccharides.

Further analysis also suggests that the effects of these metabolites on host metabolic parameters such as BMI are in part mediated by specific cytokines.

Hormonal interactions in carbohydrate metabolism Metabolusm glucoregulatory hormones of Insulin and carbohydrate metabolism body are designed to maintain circulating glucose Insulin and carbohydrate metabolism Healthy habits for athletic development a relatively narrow range. Books Insulim. As for the remaining participants, they collected their faeces in the Insuulin between 2 days Insulin and carbohydrate metabolism and 7 days metaholism their hospital carrbohydrate, with carbohyddrate exception of 5 individuals who collected their carbohydrte in the morning more than 7 days after their hospital visit, 2 individuals who reported collecting their faeces in the evening 1 day before their hospital visit, and 5 individuals who did not provide faecal samples. To construct and visualize a correlation-based network of omics data, we first analysed IR-associated host signatures using plasma cytokines, plasma metabolites and CAGE promoter expression data. Thus, individuals who underwent physical examination, laboratory tests, faecal sampling for faecal 16S rRNA pyrosequencing and metabolomic analyses, and plasma sampling for plasma metabolomic analyses were included for the analysis. Glycosidases, which catalyse the breakdown of oligo- and disaccharides 32were also associated with faecal monosaccharides Extended Data Fig.
Insulin and carbohydrate metabolism

Insulin and carbohydrate metabolism -

It is secreted from pancreatic α-cells. Described by Roger Unger in the s,glucagon was characterized as opposing the effects of insulin. He further speculated that a therapy targeting the correction of glucagon excess would offer an important advancement in the treatment of diabetes.

Hepatic glucose production, which is primarily regulated by glucagon,maintains basal blood glucose concentrations within a normal range during the fasting state.

When plasma glucose falls below the normal range, glucagon secretion increases, resulting in hepatic glucose production and return of plasma glucose to the normal range. When coupled with insulin's direct effect on the liver, glucagon suppression results in a near-total suppression of hepatic glucose output Figure 4.

Insulin and glucagon secretion: nondiabetic and diabetic subjects. In nondiabetic subjects left panel , glucose-stimulated insulin and amylin release from the β -cells results in suppression of postprandial glucagon secretion.

In a subject with type 1 diabetes, infused insulin does not suppress α -cell production of glucagon. Adapted from Ref. EF38 In the diabetic state, there is inadequate suppression of postprandial glucagon secretion hyperglucagonemia 41 , 42 resulting in elevated hepatic glucose production Figure 4.

Importantly,exogenously administered insulin is unable both to restore normal postprandial insulin concentrations in the portal vein and to suppress glucagon secretion through a paracrine effect. This results in an abnormally high glucagon-to-insulin ratio that favors the release of hepatic glucose.

The intricacies of glucose homeostasis become clearer when considering the role of gut peptides. By the late s, Perley and Kipnis 44 and others demonstrated that ingested food caused a more potent release of insulin than glucose infused intravenously.

Additionally, these hormonal signals from the proximal gut seemed to help regulate gastric emptying and gut motility. Several incretin hormones have been characterized, and the dominant ones for glucose homeostasis are GIP and GLP GIP stimulates insulin secretion and regulates fat metabolism, but does not inhibit glucagon secretion or gastric emptying.

GLP-1 also stimulates glucose-dependent insulin secretion but is significantly reduced postprandially in people with type 2 diabetes or impaired glucose tolerance. Derived from the proglucagon molecule in the intestine, GLP-1 is synthesized and secreted by the L-cells found mainly in the ileum and colon.

Circulating GLP-1 concentrations are low in the fasting state. However, both GIP and GLP-1 are effectively stimulated by ingestion of a mixed meal or meals enriched with fats and carbohydrates. GLP-1 has many glucoregulatory effects Table 1 and Figure 3. In the pancreas,GLP-1 stimulates insulin secretion in a glucose-dependent manner while inhibiting glucagon secretion.

Infusion of GLP-1 lowers postprandial glucose as well as overnight fasting blood glucose concentrations. Yet while GLP-1 inhibits glucagon secretion in the fed state, it does not appear to blunt glucagon's response to hypoglycemia.

Administration of GLP-1 has been associated with the regulation of feeding behavior and body weight. Of significant and increasing interest is the role GLP-1 may have in preservation of β-cell function and β-cell proliferation. Our understanding of the pathophysiology of diabetes is evolving.

Type 1 diabetes has been characterized as an autoimmune-mediated destruction of pancreaticβ-cells. Early in the course of type 2 diabetes, postprandial β-cell action becomes abnormal, as evidenced by the loss of immediate insulin response to a meal. Abnormal gastric emptying is common to both type 1 and type 2 diabetes.

The rate of gastric emptying is a key determinant of postprandial glucose concentrations Figure 5. In individuals with diabetes, the absent or delayed secretion of insulin further exacerbates postprandial hyperglycemia.

Both amylin and GLP-1 regulate gastric emptying by slowing the delivery of nutrients from the stomach to the small intestine. Gastric emptying rate is an important determinant of postprandial glycemia.

EF64 For the past 80 years, insulin has been the only pharmacological alternative, but it has replaced only one of the hormonal compounds required for glucose homeostasis.

Newer formulations of insulin and insulin secretagogues, such as sulfonylureas and meglitinides, have facilitated improvements in glycemic control. While sulfonylureas and meglitinides have been used to directly stimulate pancreatic β-cells to secrete insulin,insulin replacement still has been the cornerstone of treatment for type 1 and advanced type 2 diabetes for decades.

Advances in insulin therapy have included not only improving the source and purity of the hormone, but also developing more physiological means of delivery.

Clearly, there are limitations that hinder normalizing blood glucose using insulin alone. First, exogenously administered insulin does not mimic endogenous insulin secretion. In normal physiology, the liver is exposed to a two- to fourfold increase in insulin concentration compared to the peripheral circulation.

In the postprandial state, when glucagon concentrations should be low and glycogen stores should be rebuilt, there is a paradoxical elevation of glucagon and depletion of glycogen stores.

As demonstrated in the Diabetes Control and Complications Trial and the United Kingdom Prospective Diabetes Study,intensified care is not without risk.

In both studies, those subjects in the intensive therapy groups experienced a two- to threefold increase in severe hypoglycemia. Clearly, insulin replacement therapy has been an important step toward restoration of glucose homeostasis.

But it is only part of the ultimate solution. The vital relationship between insulin and glucagon has suggested additional areas for treatment. With inadequate concentrations of insulin and elevated concentrations of glucagon in the portal vein, glucagon's actions are excessive, contributing to an endogenous and unnecessary supply of glucose in the fed state.

To date, no pharmacological means of regulating glucagon exist and the need to decrease postprandial glucagon secretion remains a clinical target for future therapies. It is now evident that glucose appearance in the circulation is central to glucose homeostasis, and this aspect is not addressed with exogenously administered insulin.

Amylin works with insulin and suppresses glucagon secretion. It also helps regulate gastric emptying, which in turn influences the rate of glucose appearance in the circulation.

A synthetic analog of human amylin that binds to the amylin receptor, an amylinomimetic agent, is in development. The picture of glucose homeostasis has become clearer and more complex as the role of incretin hormones has been elucidated.

Incretin hormones play a role in helping regulate glucose appearance and in enhancing insulin secretion. Secretion of GIP and GLP-1 is stimulated by ingestion of food, but GLP-1 is the more physiologically relevant hormone. However, replacing GLP-1 in its natural state poses biological challenges.

In clinical trials, continuous subcutaneous or intravenous infusion was superior to single or repeated injections of GLP-1 because of the rapid degradation of GLP-1 by DPP-IV. To circumvent this intensive and expensive mode of treatment, clinical development of compounds that elicit similar glucoregulatory effects to those of GLP-1 are being investigated.

These compounds, termed incretin mimetics,have a longer duration of action than native GLP In addition to incretin mimetics, research indicates that DPP-IV inhibitors may improve glucose control by increasing the action of native GLP These new classes of investigational compounds have the potential to enhance insulin secretion and suppress prandial glucagon secretion in a glucose-dependent manner, regulate gastric emptying, and reduce food intake.

Despite current advances in pharmacological therapies for diabetes,attaining and maintaining optimal glycemic control has remained elusive and daunting.

Intensified management clearly has been associated with decreased risk of complications. Glucose regulation is an exquisite orchestration of many hormones, both pancreatic and gut, that exert effect on multiple target tissues, such as muscle, brain, liver, and adipocyte.

While health care practitioners and patients have had multiple therapeutic options for the past 10 years, both continue to struggle to achieve and maintain good glycemic control.

There remains a need for new interventions that complement our current therapeutic armamentarium without some of their clinical short-comings such as the risk of hypoglycemia and weight gain.

These evolving therapies offer the potential for more effective management of diabetes from a multi-hormonal perspective Figure 3 and are now under clinical development. Aronoff, MD, FACP, FACE, is a partner and clinical endocrinologist at Endocrine Associates of Dallas and director at the Research Institute of Dallas in Dallas, Tex.

Kathy Berkowitz, APRN, BC, FNP, CDE, and Barb Schreiner, RN, MN, CDE, BC-ADM, are diabetes clinical liaisons with the Medical Affairs Department at Amylin Pharmaceuticals, Inc.

Laura Want, RN, MS, CDE, CCRC, BC-ADM, is the clinical research coordinator at MedStar Research Institute in Washington, D. Note of disclosure: Dr. Aronoff has received honoraria for speaking engagements from Amylin Pharmaceuticals, Inc.

Berkowitz and Ms. Schreiner are employed by Amylin. Want serves on an advisory panel for, is a stock shareholder in, and has received honoraria for speaking engagements from Amylin and has served as a research coordinator for studies funded by the company.

She has also received research support from Lilly, Novo Nordisk, and MannKind Corporation. Amylin Pharmaceuticals, Inc. Sign In or Create an Account. Search Dropdown Menu. header search search input Search input auto suggest.

filter your search All Content All Journals Diabetes Spectrum. Advanced Search. User Tools Dropdown. Sign In. Skip Nav Destination Close navigation menu Article navigation.

Volume 17, Issue 3. Previous Article. β-CELL HORMONES. α-CELL HORMONE: GLUCAGON. INCRETIN HORMONES GLP-1 AND GIP. AMYLIN ACTIONS. GLP-1 ACTIONS. Article Navigation. Feature Articles July 01 Glucose Metabolism and Regulation: Beyond Insulin and Glucagon Stephen L. Aronoff, MD, FACP, FACE ; Stephen L.

Aronoff, MD, FACP, FACE. This Site. Google Scholar. Kathy Berkowitz, APRN, BC, FNP, CDE ; Kathy Berkowitz, APRN, BC, FNP, CDE. Barb Shreiner, RN, MN, CDE, BC-ADM ; Barb Shreiner, RN, MN, CDE, BC-ADM.

Laura Want, RN, MS, CDE, CCRC, BC-ADM Laura Want, RN, MS, CDE, CCRC, BC-ADM. Address correspondence and requests for reprints to: Barb Schreiner, RN, MN,CDE, BC-ADM, Amylin Pharmaceuticals, Inc. Diabetes Spectr ;17 3 — Get Permissions. toolbar search Search Dropdown Menu.

toolbar search search input Search input auto suggest. Figure 1. View large Download slide. Table 1. Effects of Primary Glucoregulatory Hormones.

View large. View Large. Figure 2. Figure 3. Figure 4. Figure 5. American Diabetes Association: Clinical Practice Recommendations Diabetes Care. Am Fam Physician. DCCT Research Group: Hypoglycemia in the Diabetes Control and Complications Trial.

DCCT Research Group: Weight gain associated with intensive therapy in the Diabetes Control and Complications Trial. UKPDS Study Group: Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes.

Clinical Diabetes. Biochem Biophys Res Commun. Am J Physiol. Proc Natl Acad Sci U S A. In International Textbook of Diabetes Mellitus. In William's Textbook of Endocrinology. Baillieres Best Pract Res Clin Endocrinol Metab. Chinkes DL, Aarsland A, Rosenblatt J, Wolfe RR Comparison of mass isotopomer dilution methods used to compute VLDL production in vivo.

Landau BR, Fernandez CA, Previs SF, Ekberg K, Chandramouli V, Wahren J, Kalhan SC, Brunengraber H A limitation in the use of mass isotopomer distributions to measure gluconeogenesis in fasting humans.

Am J Physiol E18—E Previs SF, Fernandez CA, Yang D, Soloviev MV, David F, Brunengraber H Limitations of the mass isotopomer distribution analysis of glucose to study gluconeogenesis. Substrate cycling between glycerol and triose phosphates in liver. J Biol Chem — Previs SF, Cline GW, Shulman GI A critical evaluation of mass isotopomer distribution analysis of gluconeogenesis in vivo.

Peroni O, Large V, Beylot M Measuring gluconeogenesis with [C]glycerol and mass isotopomer distribution analysis of glucose.

Tayek JA, Katz J Glucose production, recycling, and gluconeogenesis in normals and diabetics: a mass isotopomer [UC]glucose study. Kelleher JK Estimating gluconeogenesis with [UC]glucose: molecular condensation requires a molecular approach.

Tayek JA, Katz J Glucose production, recycling, Cori cycle, and gluconeogenesis in humans: relationship to serum cortisol. Landau BR, Wahren J, Ekberg K, Previs SF, Yang D, Brunengraber H Limitations in estimating gluconeogenesis and Cori cycling from mass isotopomer distributions using [UC6]glucose.

Katz J, Tayek JA Recycling of glucose and determination of the Cori Cycle and gluconeogenesis. Radziuk J, Lee WP Measurement of gluconeogenesis and mass isotopomer analysis based on [U- 13 C]glucose. Katz J, Tayek JA Gluconeogenesis and the Cori cycle in , , and h-fasted humans. Mao CS, Bassilian S, Lim SK, Lee WN Underestimation of gluconeogenesis by the [U- 13 C 6 ]glucose method: effect of lack of isotope equilibrium.

Am J Physiol Endocrinol Metab E—E Haymond MW, Sunehag AL The reciprocal pool model for the measurement of gluconeogenesis by use of [U- 13 C]glucose.

Allick G, van der Crabben SN, Ackermans MT, Endert E, Sauerwein HP Measurement of gluconeogenesis by deuterated water: the effect of equilibration time and fasting period. Jin ES, Jones JG, Merritt M, Burgess SC, Malloy CR, Sherry AD Glucose production, gluconeogenesis, and hepatic tricarboxylic acid cycle fluxes measured by nuclear magnetic resonance analysis of a single glucose derivative.

Anal Biochem — Jones JG, Solomon MA, Cole SM, Sherry AD, Malloy CR An integrated 2 H and 13 C NMR study of gluconeogenesis and TCA cycle flux in humans. Landau BR, Wahren J, Chandramouli V, Schumann WC, Ekberg K, Kalhan SC Use of 2H 2 O for estimating rates of gluconeogenesis.

Application to the fasted state. Jones JG, Carvalho RA, Sherry AD, Malloy CR Quantitation of gluconeogenesis by 2 H nuclear magnetic resonance analysis of plasma glucose following ingestion of 2 H 2 O.

Basu R, Chandramouli V, Schumann W, Basu A, Landau BR, Rizza RA Additional evidence that transaldolase exchange, isotope discrimination during the triose-isomerase reaction, or both occur in humans: effects of type 2 diabetes.

Article CAS Google Scholar. Giaccari A, Rossetti L Isocratic high-performance liquid chromatographic determination of the concentration and specific radioactivity of phosphoenolpyruvate and uridine diphosphate glucose in tissue extracts. J Chromatogr B Biomed Appl — Rossetti L, Massillon D, Barzilai N, Vuguin P, Chen W, Hawkins M, Wu J, Wang J Short term effects of leptin on hepatic gluconeogenesis and in vivo insulin action.

Rossetti L, Giaccari A, Barzilai N, Howard K, Sebel G, Hu M Mechanism by which hyperglycemia inhibits hepatic glucose production in conscious rats. Implications for the pathophysiology of fasting hyperglycemia in diabetes.

Massillon D, Chen W, Hawkins M, Liu R, Barzilai N, Rossetti L Quantitation of hepatic glucose fluxes and pathways of hepatic glycogen synthesis in conscious mice.

Goldstein RE, Rossetti L, Palmer BA, Liu R, Massillon D, Scott M, Neal D, Williams P, Peeler B, Cherrington AD Effects of fasting and glucocorticoids on hepatic gluconeogenesis assessed using two independent methods in vivo. Goldstein RE, Wasserman DH, McGuinness OP, Lacy DB, Cherrington AD, Abumrad NN Effects of chronic elevation in plasma cortisol on hepatic carbohydrate metabolism.

Muniyappa R, Lee S, Chen H, Quon MJ Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage.

Am J Physiol Endocrinol Metab E15—E Choukem SP, Gautier JF How to measure hepatic insulin resistance? Diab Metab E—E Yokoyama H, Emoto M, Fujiwara S, Motoyama K, Morioka T, Komatsu M, Tahara H, Shoji T, Okuno Y, Nishizawa Y Quantitative insulin sensitivity check index and the reciprocal index of homeostasis model assessment in normal range weight and moderately obese type 2 diabetic patients.

Laakso M How good a marker is insulin level for insulin resistance? Am J Epidemiol — Tran TT, Gupta N, Goh T, Naigamwalla D, Chia MC, Koohestani N, Mehrotra S, Keown-Eyssen G, Giacca A, Bruce WR Direct measure of insulin sensitivity with the hyperinsulinemic-euglycemic clamp and surrogate measures of insulin sensitivity with the oral glucose tolerance test: correlations with aberrant crypt foci promotion in rats.

Cancer Epidemiol Biomark Prev — Lee S, Muniyappa R, Yan X, Chen H, Yue LQ, Hong EG, Kim JK, Quon MJ Comparison between surrogate indexes of insulin sensitivity and resistance and hyperinsulinemic euglycemic clamp estimates in mice. Wallace TM, Levy JC, Matthews DR Use and abuse of HOMA modeling.

Article PubMed Google Scholar. Turner RC, Holman RR, Matthews D, Hockaday TD, Peto J Insulin deficiency and insulin resistance interaction in diabetes: estimation of their relative contribution by feedback analysis from basal plasma insulin and glucose concentrations. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man.

Diabetologia — Hosker JP, Matthews DR, Rudenski AS, Burnett MA, Darling P, Bown EG, Turner RC Continuous infusion of glucose with model assessment: measurement of insulin resistance and beta-cell function in man. Bonora E, Targher G, Alberiche M, Bonadonna RC, Saggiani F, Zenere MB, Monauni T, Muggeo M Homeostasis model assessment closely mirrors the glucose clamp technique in the assessment of insulin sensitivity: studies in subjects with various degrees of glucose tolerance and insulin sensitivity.

Tripathy D, Almgren P, Tuomi T, Groop L Contribution of insulin-stimulated glucose uptake and basal hepatic insulin sensitivity to surrogate measures of insulin sensitivity.

Matsuda M, DeFronzo RA Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Abdul-Ghani MA, Matsuda M, DeFronzo RA Strong association between insulin resistance in liver and skeletal muscle in non-diabetic subjects.

Diabet Med — Abdul-Ghani MA, Matsuda M, Balas B, DeFronzo RA Muscle and liver insulin resistance indexes derived from the oral glucose tolerance test.

Katz A, Nambi SS, Mather K, Baron AD, Follmann DA, Sullivan G, Quon MJ Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. Rabasa-Lhoret R, Bastard JP, Jan V, Ducluzeau PH, Andreelli F, Guebre F, Bruzeau J, Louche-Pellissier C, MaItrepierre C, Peyrat J, Chagne J, Vidal H, Laville M Modified quantitative insulin sensitivity check index is better correlated to hyperinsulinemic glucose clamp than other fasting-based index of insulin sensitivity in different insulin-resistant states.

Yokoyama H, Emoto M, Fujiwara S, Motoyama K, Morioka T, Komatsu M, Tahara H, Koyama H, Shoji T, Inaba M, Nishizawa Y Quantitative insulin sensitivity check index and the reciprocal index of homeostasis model assessment are useful indexes of insulin resistance in type 2 diabetic patients with wide range of fasting plasma glucose.

Uwaifo GI, Fallon EM, Chin J, Elberg J, Parikh SJ, Yanovski JA Indices of insulin action, disposal, and secretion derived from fasting samples and clamps in normal glucose-tolerant black and white children. euglycaemic-hyperinsulinemic clamp as an indicator of insulin resistance and cardiovascular risk factors in overweight and obese postmenopausal women.

Diabetes Metab — Cacho J, Sevillano J, de Castro J, Herrera E, Ramos MP Validation of simple indexes to assess insulin sensitivity during pregnancy in Wistar and Sprague-Dawley rats.

Bonora E, Moghetti P, Zancanaro C, Cigolini M, Querena M, Cacciatori V, Corgnati A, Muggeo M Estimates of in vivo insulin action in man: comparison of insulin tolerance tests with euglycemic and hyperglycemic glucose clamp studies. Hirst S, Phillips DI, Vines SK, Clark PM, Hales CN Reproducibility of the short insulin tolerance test.

Chen CC, Wang TY, Hsu SY, Chen RH, Chang CT, Chen SJ Is the short insulin tolerance test safe and reproducible? Reaven GM Insulin resistance in noninsulin-dependent diabetes mellitus.

Does it exist and can it be measured? Am J Med — Gelding SV, Robinson S, Lowe S, Niththyananthan R, Johnston DG Validation of the low dose short insulin tolerance test for evaluation of insulin sensitivity.

Clin Endocrinol — Akinmokun A, Selby PL, Ramaiya K, Alberti KG The short insulin tolerance test for determination of insulin sensitivity: a comparison with the euglycaemic clamp.

Young RP, Critchley JA, Anderson PJ, Lau MS, Lee KK, Chan JC The short insulin tolerance test: feasibility study using venous sampling. Phillips DI, Clark PM, Hales CN, Osmond C Understanding oral glucose tolerance: comparison of glucose or insulin measurements during the oral glucose tolerance test with specific measurements of insulin resistance and insulin secretion.

Xu E, Dubois MJ, Leung N, Charbonneau A, Turbide C, Avramoglu RK, DeMarte L, Elchebly M, Streichert T, Levy E, Beauchemin N, Marette A Targeted disruption of carcinoembryonic antigen-related cell adhesion molecule 1 promotes diet-induced hepatic steatosis and insulin resistance.

Endocrinology — Minami A, Iseki M, Kishi K, Wang M, Ogura M, Furukawa N, Hayashi S, Yamada M, Obata T, Takeshita Y, Nakaya Y, Bando Y, Izumi K, Moodie SA, Kajiura F, Matsumoto M, Takatsu K, Takaki S, Ebina Y Increased insulin sensitivity and hypoinsulinemia in APS knockout mice.

Cariou B, van Harmelen K, Duran-Sandoval D, van Dijk TH, Grefhorst A, Abdelkarim M, Caron S, Torpier G, Fruchart JC, Gonzalez FJ, Kuipers F, Staels B The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. Carvalho E, Kotani K, Peroni OD, Kahn BB Adipose-specific overexpression of GLUT4 reverses insulin resistance and diabetes in mice lacking GLUT4 selectively in muscle.

Araujo EP, De Souza CT, Ueno M, Cintra DE, Bertolo MB, Carvalheira JB, Saad MJ, Velloso LA Infliximab restores glucose homeostasis in an animal model of diet-induced obesity and diabetes.

Mulder H, Sorhede-Winzell M, Contreras JA, Fex M, Strom K, Ploug T, Galbo H, Arner P, Lundberg C, Sundler F, Ahren B, Holm C Hormone-sensitive lipase null mice exhibit signs of impaired insulin sensitivity whereas insulin secretion is intact.

Ahren B, Pacini G A novel approach to assess insulin sensitivity reveals no increased insulin sensitivity in mice with a dominant-negative mutant hepatocyte nuclear factor-1alpha.

Am J Physiol Regul Integr Comp Physiol R—R Kahn SE The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of Type 2 diabetes.

American Diabetes Association Executive summary: standards of medical care in diabetes — Diabetes Care 32 Suppl 1 :S6—S Canadian Diabetes Association Clinical Practice Guidelines Expert Committee Canadian Diabetes Association clinical practice guidelines for the prevention and management of diabetes in Canada.

Can J Diabetes S1—S Soonthornpun S, Setasuban W, Thamprasit A, Chayanunnukul W, Rattarasarn C, Geater A Novel insulin sensitivity index derived from oral glucose tolerance test. Kazama Y, Takamura T, Sakurai M, Shindo H, Ohkubo E, Aida K, Harii N, Taki K, Kaneshige M, Tanaka S, Shimura H, Endo T, Kobayashi T New insulin sensitivity index from the oral glucose tolerance test.

Diab Res Clin Pract — Avignon A, Boegner C, Mariano-Goulart D, Colette C, Monnier L Assessment of insulin sensitivity from plasma insulin and glucose in the fasting or post oral glucose-load state.

Int J Obes Relat Metab Disord — Tao R, Ye F, He Y, Tian J, Liu G, Ji T, Su Y Improvement of high-fat-diet-induced metabolic syndrome by a compound from Balanophora polyandra Griff in mice. Eur J Pharmacol — Nakaya Y, Minami A, Harada N, Sakamoto S, Niwa Y, Ohnaka M Taurine improves insulin sensitivity in the Otsuka Long-Evans Tokushima Fatty rat, a model of spontaneous type 2 diabetes.

Am J Clin Nutr — Yao XH, Gregoire Nyomba BL Abnormal glucose homeostasis in adult female rat offspring after intrauterine ethanol exposure. Parlevliet ET, Heijboer AC, Schroder-van der Elst JP, Havekes LM, Romijn JA, Pijl H, Corssmit EP Oxyntomodulin ameliorates glucose intolerance in mice fed a high-fat diet.

Pacini G, Bergman RN MINMOD: a computer program to calculate insulin sensitivity and pancreatic responsivity from the frequently sampled intravenous glucose tolerance test. Comput Meth Programs Biomed — Pacini G, Tonolo G, Sambataro M, Maioli M, Ciccarese M, Brocco E, Avogaro A, Nosadini R Insulin sensitivity and glucose effectiveness: minimal model analysis of regular and insulin-modified FSIGT.

Mehring GH, Coates PA, Brunel PC, Luzio SD, Owens DR Insulin sensitivity in type 2 diabetes: univariate and multivariate techniques to derive estimates of insulin sensitivity from the insulin modified intravenous glucose tolerance test FSIGT.

Bergman RN, Ider YZ, Bowden CR, Cobelli C Quantitative estimation of insulin sensitivity. Bergman RN, Phillips LS, Cobelli C Physiologic evaluation of factors controlling glucose tolerance in man: measurement of insulin sensitivity and beta-cell glucose sensitivity from the response to intravenous glucose.

Yang YJ, Youn JH, Bergman RN Modified protocols improve insulin sensitivity estimation using the minimal model. Coates PA, Luzio SD, Brunel P, Owens DR Comparison of estimates of insulin sensitivity from minimal model analysis of the insulin-modified frequently sampled intravenous glucose tolerance test and the isoglycemic hyperinsulinemic clamp in subjects with NIDDM.

Brehm A, Thomaseth K, Bernroider E, Nowotny P, Waldhausl W, Pacini G, Roden M The role of endocrine counterregulation for estimating insulin sensitivity from intravenous glucose tolerance tests.

Saad MF, Steil GM, Kades WW, Ayad MF, Elsewafy WA, Boyadjian R, Jinagouda SD, Bergman RN Differences between the tolbutamide-boosted and the insulin-modified minimal model protocols.

Sumner AE, Luercio MF, Frempong BA, Ricks M, Sen S, Kushner H, Tulloch-Reid MK Validity of the reduced-sample insulin modified frequently-sampled intravenous glucose tolerance test using the nonlinear regression approach. Caumo A, Vicini P, Zachwieja JJ, Avogaro A, Yarasheski K, Bier DM, Cobelli C Undermodeling affects minimal model indexes: insights from a two-compartment model.

Hoffman RP, Vicini P, Cobelli C Comparison of insulin sensitivity and glucose effectiveness determined by the one- and two-compartment-labeled minimal model in late prepubertal children and early adolescents. Krudys KM, Dodds MG, Nissen SM, Vicini P Integrated model of hepatic and peripheral glucose regulation for estimation of endogenous glucose production during the hot IVGTT.

Tokuyama K, Suzuki M Intravenous glucose tolerance test-derived glucose effectiveness in endurance-trained rats. Pawlak DB, Bryson JM, Denyer GS, Brand-Miller JC High glycemic index starch promotes hypersecretion of insulin and higher body fat in rats without affecting insulin sensitivity.

J Nutr — Pacini G, Thomaseth K, Ahren B Contribution to glucose tolerance of insulin-independent vs. insulin-dependent mechanisms in mice.

Pacini G, Ahren M, Ahren B Reappraisal of the intravenous glucose tolerance index for a simple assessment of insulin sensitivity in mice. Pei D, Jones CN, Bhargava R, Chen YD, Reaven GM Evaluation of octreotide to assess insulin-mediated glucose disposal by the insulin suppression test.

Kim SH, Reaven GM Isolated impaired fasting glucose and peripheral insulin sensitivity: not a simple relationship. Shen SW, Reaven GM, Farquhar JW Comparison of impedance to insulin-mediated glucose uptake in normal subjects and in subjects with latent diabetes.

Hwu CM, Kwok CF, Chiang SC, Wang PY, Hsiao LC, Lee SH, Lin SH, Ho LT A comparison of insulin suppression tests performed with somatostatin and octreotide with particular reference to tolerability.

Ikebuchi M, Suzuki M, Kageyama A, Hirose J, Yokota C, Ikeda K, Shinozaki K, Todo R, Harano Y Modified method using a somatostatin analogue, octreotide acetate Sandostatin to assess in vivo insulin sensitivity.

Endocr J — Weir GC, Bonner-Weir S Islets of Langerhans: the puzzle of intraislet interactions and their relevance to diabetes. Maheux P, Azhar S, Kern PA, Chen YD, Reuven GM Relationship between insulin-mediated glucose disposal and regulation of plasma and adipose tissue lipoprotein lipase.

Facchini F, Humphreys MH, Jeppesen J, Reaven GM Measurements of insulin-­mediated glucose disposal are stable over time. Greenfield MS, Doberne L, Kraemer F, Tobey T, Reaven G Assessment of insulin resistance with the insulin suppression test and the euglycemic clamp.

Rodnick KJ, Mondon CE, Haskell WL, Azhar S, Reaven GM Differences in insulin-induced glucose uptake and enzyme activity in running rats. J Appl Physiol — Maegawa H, Kobayashi M, Ishibashi O, Takata Y, Shigeta Y Effect of diet change on insulin action: difference between muscles and adipocytes.

Maegawa H, Hasegawa M, Sugai S, Obata T, Ugi S, Morino K, Egawa K, Fujita T, Sakamoto T, Nishio Y, Kojima H, Haneda M, Yasuda H, Kikkawa R, Kashiwagi A Expression of a dominant negative SHP-2 in transgenic mice induces insulin resistance.

Uwaifo GI, Parikh SJ, Keil M, Elberg J, Chin J, Yanovski JA Comparison of insulin sensitivity, clearance, and secretion estimates using euglycemic and hyperglycemic clamps in children.

Carpentier A, Zinman B, Leung N, Giacca A, Hanley AJ, Harris SB, Hegele RA, Lewis GF Free fatty acid-mediated impairment of glucose-stimulated insulin secretion in nondiabetic Oji-Cree individuals from the Sandy Lake community of Ontario, Canada: a population at very high risk for developing type 2 diabetes.

Mari A, Ahren B, Pacini G Assessment of insulin secretion in relation to insulin resistance. Goh TT, Mason TM, Gupta N, So A, Lam TK, Lam L, Lewis GF, Mari A, Giacca A Lipid-induced beta-cell dysfunction in vivo in models of progressive beta-cell failure.

Mitrakou A, Vuorinen-Markkola H, Raptis G, Toft I, Mokan M, Strumph P, Pimenta W, Veneman T, Jenssen T, Bolli G Simultaneous assessment of insulin secretion and insulin sensitivity using a hyperglycemia clamp.

Elahi D, Meneilly GS, Minaker KL, Andersen DK, Rowe JW Escape of hepatic ­glucose production during hyperglycemic clamp. Ayala JE, Bracy DP, McGuinness OP, Wasserman DH Considerations in the design of hyperinsulinemic-euglycemic clamps in the conscious mouse. Xu B, Bird VG, Miller WT Substrate specificities of the insulin and insulin-like growth factor 1 receptor tyrosine kinase catalytic domains.

Araki E, Lipes MA, Patti ME, Bruning JC, Haag B III, Johnson RS, Kahn CR Alternative pathway of insulin signalling in mice with targeted disruption of the IRS-1 gene. Nature — Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, Zhang Y, Bernal D, Pons S, Shulman GI, Bonner-Weir S, White MF Disruption of IRS-2 causes type 2 diabetes in mice.

Liu SC, Wang Q, Lienhard GE, Keller SR Insulin receptor substrate 3 is not essential for growth or glucose homeostasis. Fantin VR, Wang Q, Lienhard GE, Keller SR Mice lacking insulin receptor substrate 4 exhibit mild defects in growth, reproduction, and glucose homeostasis.

Sciacchitano S, Taylor SI Cloning, tissue expression, and chromosomal localization of the mouse IRS-3 gene. Fantin VR, Lavan BE, Wang Q, Jenkins NA, Gilbert DJ, Copeland NG, Keller SR, Lienhard GE Cloning, tissue expression, and chromosomal location of the mouse insulin receptor substrate 4 gene.

Genes Dev — Thirone AC, Huang C, Klip A Tissue-specific roles of IRS proteins in insulin signaling and glucose transport. Trends Endocrinol Metab — Article PubMed CAS Google Scholar. Yamauchi T, Tobe K, Tamemoto H, Ueki K, Kaburagi Y, Yamamoto-Honda R, Takahashi Y, Yoshizawa F, Aizawa S, Akanuma Y, Sonenberg N, Yazaki Y, Kadowaki T Insulin signalling and insulin actions in the muscles and livers of insulin-resistant, insulin receptor substrate 1-deficient mice.

Mol Cell Biol — Kubota N, Tobe K, Terauchi Y, Eto K, Yamauchi T, Suzuki R, Tsubamoto Y, Komeda K, Nakano R, Miki H, Satoh S, Sekihara H, Sciacchitano S, Lesniak M, Aizawa S, Nagai R, Kimura S, Akanuma Y, Taylor SI, Kadowaki T Disruption of insulin receptor substrate 2 causes type 2 diabetes because of liver insulin resistance and lack of compensatory beta-cell hyperplasia.

Kido Y, Burks DJ, Withers D, Bruning JC, Kahn CR, White MF, Accili D Tissue-specific insulin resistance in mice with mutations in the insulin receptor, IRS-1, and IRS Taniguchi CM, Ueki K, Kahn R Complementary roles of IRS-1 and IRS-2 in the hepatic regulation of metabolism.

J Clin Invest — Simmgen M, Knauf C, Lopez M, Choudhury AI, Charalambous M, Cantley J, Bedford DC, Claret M, Iglesias MA, Heffron H, Cani PD, Vidal-Puig A, Burcelin R, Withers DJ Liver-specific deletion of insulin receptor substrate 2 does not impair hepatic glucose and lipid metabolism in mice.

Dong X, Park S, Lin X, Copps K, Yi X, White MF Irs1 and Irs2 signaling is essential for hepatic glucose homeostasis and systemic growth. Razzini G, Ingrosso A, Brancaccio A, Sciacchitano S, Esposito DL, Falasca M Different subcellular localization and phosphoinositides binding of insulin receptor substrate protein pleckstrin homology domains.

Mol Endocrinol — Sawka-Verhelle D, Tartare-Deckert S, White MF, Van OE Insulin receptor substrate-2 binds to the insulin receptor through its phosphotyrosine-binding domain and through a newly identified domain comprising amino acids Evidence for two distinct phosphotyrosine-dependent interaction domains within IRS Wolf G, Trub T, Ottinger E, Groninga L, Lynch A, White MF, Miyazaki M, Lee J, Shoelson SE PTB domains of IRS-1 and Shc have distinct but overlapping binding specificities.

Cheatham B, Vlahos CJ, Cheatham L, Wang L, Blenis J, Kahn CR Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp 70 S6 kinase, DNA synthesis, and glucose transporter translocation.

CAS PubMed PubMed Central Google Scholar. Cell Metab — Geering B, Cutillas PR, Nock G, Gharbi SI, Vanhaesebroeck B Class IA phosphoinositide 3-kinases are obligate pp heterodimers.

Vanhaesebroeck B, Leevers SJ, Panayotou G, Waterfield MD Phosphoinositide 3-kinases: a conserved family of signal transducers. Trends Biochem Sci — Shepherd PR, Withers DJ, Siddle K Phosphoinositide 3-kinase: the key switch mechanism in insulin signalling.

Biochem J — Terauchi Y, Tsuji Y, Satoh S, Minoura H, Murakami K, Okuno A, Inukai K, Asano T, Kaburagi Y, Ueki K, Nakajima H, Hanafusa T, Matsuzawa Y, Sekihara H, Yin Y, Barrett JC, Oda H, Ishikawa T, Akanuma Y, Komuro I, Suzuki M, Yamamura K, Kodama T, Suzuki H, Yamamura K, Kodama T, Suzuki H, Koyasu S, Aizawa S, Tobe K, Fukui Y, Yazaki Y, Kadowaki T Increased insulin sensitivity and hypoglycaemia in mice lacking the p85 alpha subunit of phosphoinositide 3-kinase.

Nat Genet — Ueki K, Yballe CM, Brachmann SM, Vicent D, Watt JM, Kahn CR, Cantley LC Increased insulin sensitivity in mice lacking p85beta subunit of phosphoinositide 3-kinase. Aoki K, Matsui J, Kubota N, Nakajima H, Iwamoto K, Takamoto I, Tsuji Y, Ohno A, Mori S, Tokuyama K, Murakami K, Asano T, Aizawa S, Tobe K, Kadowaki T, Terauchi Y Role of the liver in glucose homeostasis in PI 3-kinase p85alpha-deficient mice.

Brachmann SM, Ueki K, Engelman JA, Kahn RC, Cantley LC Phosphoinositide 3-kinase catalytic subunit deletion and regulatory subunit deletion have opposite effects on insulin sensitivity in mice. Taniguchi CM, Tran TT, Kondo T, Luo J, Ueki K, Cantley LC, Kahn CR Phosphoinositide 3-kinase regulatory subunit p85alpha suppresses insulin action via positive regulation of PTEN.

Taniguchi CM, Aleman JO, Ueki K, Luo J, Asano T, Kaneto H, Stephanopoulos G, Cantley LC, Kahn CR The p85alpha regulatory subunit of phosphoinositide 3-kinase potentiates c-Jun N-terminal kinase-mediated insulin resistance. Logie L, Ruiz-Alcaraz AJ, Keane M, Woods YL, Bain J, Marquez R, Alessi DR, Sutherland C Characterization of a protein kinase B inhibitor in vitro and in insulin-treated liver cells.

Tanti JF, Grillo S, Gremeaux T, Coffer PJ, Van OE, Le Marchand-Brustel Y Potential role of protein kinase B in glucose transporter 4 translocation in adipocytes. Scheid MP, Marignani PA, Woodgett JR Multiple phosphoinositide 3-kinase-dependent steps in activation of protein kinase B. Stokoe D, Stephens LR, Copeland T, Gaffney PR, Reese CB, Painter GF, Holmes AB, McCormick F, Hawkins PT Dual role of phosphatidylinositol-3,4,5-trisphosphate in the activation of protein kinase B.

Cell — Liu P, Heng H, Roberts TM, Hao JJ Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov — Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw EB III, Kaestner KH, Bartolomei MS, Shulman GI, Birnbaum MJ Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 PKB beta.

Development — Dummler B, Tschopp O, Hynx D, Yang ZZ, Dirnhofer S, Hemmings BA Life with a single isoform of Akt: mice lacking Akt2 and Akt3 are viable but display impaired glucose homeostasis and growth deficiencies.

Brozinick JT Jr, Roberts BR, Dohm GL Defective signaling through Akt-2 and -3 but not Akt-1 in insulin-resistant human skeletal muscle: potential role in insulin resistance. Kim YB, Peroni OD, Franke TF, Kahn BB Divergent regulation of Akt1 and Akt2 isoforms in insulin target tissues of obese Zucker rats.

Kahn CR Insulin resistance, insulin insensitivity, and insulin unresponsiveness: a necessary distinction. Metabolism 27 12 Suppl 2 — Paz K, Hemi R, LeRoith D, Karasik A, Elhanany E, Kanety H, Zick Y A molecular basis for insulin resistance.

Aguirre V, Uchida T, Yenush L, Davis R, White MF The c-Jun NH 2 -terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser Greene MW, Sakaue H, Wang L, Alessi DR, Roth RA Modulation of insulin-­stimulated degradation of human insulin receptor substrate-1 by Serine phosphorylation.

Zhang J, Gao Z, Yin J, Quon MJ, Ye J S6K directly phosphorylates IRS-1 on Ser to promote insulin resistance in response to TNF- alpha signaling through IKK2. Emanuelli B, Peraldi P, Filloux C, Chavey C, Freidinger K, Hilton DJ, Hotamisligil GS, Van OE SOCS-3 inhibits insulin signaling and is up-regulated in response to tumor necrosis factor-alpha in the adipose tissue of obese mice.

Ueki K, Kondo T, Kahn CR Suppressor of cytokine signaling 1 SOCS-1 and SOCS-3 cause insulin resistance through inhibition of tyrosine phosphorylation of insulin receptor substrate proteins by discrete mechanisms.

Rui L, Yuan M, Frantz D, Shoelson S, White MF SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2. Liberman Z, Plotkin B, Tennenbaum T, Eldar-Finkelman H Coordinated phosphorylation of insulin receptor substrate-1 by glycogen synthase kinase-3 and protein kinase C betaII in the diabetic fat tissue.

Liberman Z, Eldar-Finkelman H Serine phosphorylation of insulin receptor substrate-1 by glycogen synthase kinase-3 attenuates insulin signaling. Kovacs P, Hanson RL, Lee YH, Yang X, Kobes S, Permana PA, Bogardus C, Baier LJ The role of insulin receptor substrate-1 gene IRS1 in type 2 diabetes in Pima Indians.

Carlson CJ, Koterski S, Sciotti RJ, Poccard GB, Rondinone CM Enhanced basal activation of mitogen-activated protein kinases in adipocytes from type 2 diabetes: potential role of p38 in the downregulation of GLUT4 expression. Danielsson A, Ost A, Lystedt E, Kjolhede P, Gustavsson J, Nystrom FH, Stralfors P Insulin resistance in human adipocytes occurs downstream of IRS1 after surgical cell isolation but at the level of phosphorylation of IRS1 in type 2 diabetes.

FEBS J — Goodyear LJ, Giorgino F, Sherman LA, Carey J, Smith RJ, Dohm GL Insulin receptor phosphorylation, insulin receptor substrate-1 phosphorylation, and phosphatidylinositol 3-kinase activity are decreased in intact skeletal muscle strips from obese subjects.

Bjornholm M, Kawano Y, Lehtihet M, Zierath JR Insulin receptor substrate-1 phosphorylation and phosphatidylinositol 3-kinase activity in skeletal muscle from NIDDM subjects after in vivo insulin stimulation.

Krook A, Bjornholm M, Galuska D, Jiang XJ, Fahlman R, Myers MG Jr, Wallberg-Henriksson H, Zierath JR Characterization of signal transduction and glucose transport in skeletal muscle from type 2 diabetic patients.

Beeson M, Sajan MP, Dizon M, Grebenev D, Gomez-Daspet J, Miura A, Kanoh Y, Powe J, Bandyopadhyay G, Standaert ML, Farese RV Activation of protein kinase C-zeta by insulin and phosphatidylinositol-3,4,5- PO4 3 is defective in muscle in type 2 diabetes and impaired glucose tolerance: amelioration by rosiglitazone and exercise.

Rondinone CM, Carvalho E, Wesslau C, Smith UP Impaired glucose transport and protein kinase B activation by insulin, but not okadaic acid, in adipocytes from subjects with Type II diabetes mellitus.

Rondinone CM, Wang LM, Lonnroth P, Wesslau C, Pierce JH, Smith U Insulin receptor substrate IRS 1 is reduced and IRS-2 is the main docking protein for phosphatidylinositol 3-kinase in adipocytes from subjects with non-insulin-dependent diabetes mellitus.

Rieusset J, Bouzakri K, Chevillotte E, Ricard N, Jacquet D, Bastard JP, Laville M, Vidal H Suppressor of cytokine signaling 3 expression and insulin resistance in skeletal muscle of obese and type 2 diabetic patients.

Avruch J Insulin signal transduction through protein kinase cascades. Mol Cell Biochem — Matozaki T, Murata Y, Saito Y, Okazawa H, Ohnishi H Protein tyrosine phosphatase SHP a proto-oncogene product that promotes Ras activation.

Cancer Sci 10 — Li N, Batzer A, Daly R, Yajnik V, Skolnik E, Chardin P, Bar-Sagi D, Margolis B, Schlessinger J Guanine-nucleotide-releasing factor hSos1 binds to Grb2 and links receptor tyrosine kinases to Ras signalling.

Ravichandran KS, Lorenz U, Shoelson SE, Burakoff SJ Interaction of Shc with Grb2 regulates association of Grb2 with mSOS. Holgado-Madruga M, Emlet DR, Moscatello DK, Godwin AK, Wong AJ A Grb2-associated docking protein in EGF- and insulin-receptor signalling.

Baltensperger K, Kozma LM, Cherniack AD, Klarlund JK, Chawla A, Banerjee U, Czech MP Binding of the Ras activator son of sevenless to insulin receptor substrate-1 ­signaling complexes. Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms.

Dunn KL, Espino PS, Drobic B, He S, Davie JR The Ras-MAPK signal transduction pathway, cancer and chromatin remodeling.

Biochem Cell Biol — Roberts PJ, Der CJ Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer.

Oncogene — Ma XM, Blenis J Molecular mechanisms of mTOR-mediated translational control. Hay N, Sonenberg N Upstream and downstream of mTOR. Avruch J, Long X, Ortiz-Vega S, Rapley J, Papageorgiou A, Dai N Amino acid regulation of TOR complex 1.

Anjum R, Blenis J The RSK family of kinases: emerging roles in cellular signalling. Memmott RM, Dennis PA Akt-dependent and -independent mechanisms of mTOR regulation in cancer. Cell Signal — Bouzakri K, Roques M, Gual P, Espinosa S, Guebre-Egziabher F, Riou JP, Laville M, Le Marchand-Brustel Y, Tanti JF, Vidal H Reduced activation of phosphatidylinositol-3 kinase and increased serine phosphorylation of insulin receptor substrate-1 in primary culture of skeletal muscle cells from patients with type 2 diabetes.

Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, Fumagalli S, Allegrini PR, Kozma SC, Auwerx J, Thomas G Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity.

Tremblay F, Gagnon A, Veilleux A, Sorisky A, Marette A Activation of the mammalian target of rapamycin pathway acutely inhibits insulin signaling to Akt and glucose transport in 3T3-L1 and human adipocytes.

Katz H, Butler P, Homan M, Zerman A, Caumo A, Cobelli C, Rizza R Hepatic and extrahepatic insulin action in humans: measurement in the absence of non-steady-state error. Olson AL, Pessin JE Structure, function, and regulation of the mammalian facilitative glucose transporter gene family.

Annu Rev Nutr — Dentin R, Pegorier JP, Benhamed F, Foufelle F, Ferre P, Fauveau V, Magnuson MA, Girard J, Postic C Hepatic glucokinase is required for the synergistic action of ChREBP and SREBP-1c on glycolytic and lipogenic gene expression.

Sibrowski W, Seitz HJ Rapid action of insulin and cyclic AMP in the regulation of functional messenger RNA coding for glucokinase in rat liver. Kim SY, Kim HI, Kim TH, Im SS, Park SK, Lee IK, Kim KS, Ahn YH SREBP-1c mediates the insulin-dependent hepatic glucokinase expression.

Postic C, Shiota M, Niswender KD, Jetton TL, Chen Y, Moates JM, Shelton KD, Lindner J, Cherrington AD, Magnuson MA Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell-specific gene knock-outs using Cre recombinase.

Frame S, Cohen P GSK3 takes centre stage more than 20 years after its discovery. Imazu M, Strickland WG, Chrisman TD, Exton JH Phosphorylation and inactivation of liver glycogen synthase by liver protein kinases.

Sharfi H, Eldar-Finkelman H Sequential phosphorylation of insulin receptor ­substrate-2 by glycogen synthase kinase-3 and c-Jun NH 2 -terminal kinase plays a role in hepatic insulin signaling. MacAulay K, Doble BW, Patel S, Hansotia T, Sinclair EM, Drucker DJ, Nagy A, Woodgett JR Glycogen synthase kinase 3alpha-specific regulation of murine hepatic glycogen metabolism.

Patel S, Doble BW, MacAulay K, Sinclair EM, Drucker DJ, Woodgett JR Tissue-specific role of glycogen synthase kinase 3beta in glucose homeostasis and insulin action.

Aiston S, Coghlan MP, Agius L Inactivation of phosphorylase is a major component of the mechanism by which insulin stimulates hepatic glycogen synthesis.

Eur J Biochem — Cohen P The twentieth century struggle to decipher insulin signalling. Munro S, Ceulemans H, Bollen M, Diplexcito J, Cohen PT A novel glycogen-­targeting subunit of protein phosphatase 1 that is regulated by insulin and shows differential tissue distribution in humans and rodents.

Alemany S, Cohen P Phosphorylase a is an allosteric inhibitor of the glycogen and microsomal forms of rat hepatic protein phosphatase FEBS Lett — Moorhead G, MacKintosh C, Morrice N, Cohen P Purification of the hepatic glycogen-associated form of protein phosphatase-1 by microcystin-Sepharose affinity chromatography.

Bollen M, Keppens S, Stalmans W Specific features of glycogen metabolism in the liver. Doherty MJ, Cadefau J, Stalmans W, Bollen M, Cohen PT Loss of the hepatic glycogen-binding subunit GL of protein phosphatase 1 underlies deficient glycogen synthesis in insulin-dependent diabetic rats and in adrenalectomized starved rats.

Lam TK, Carpentier A, Lewis GF, van de Werve G, Fantus IG, Giacca A Mechanisms of the free fatty acid-induced increase in hepatic glucose production. Physiol Rev — Schmoll D, Walker KS, Alessi DR, Grempler R, Burchell A, Guo S, Walther R, Unterman TG Regulation of glucosephosphatase gene expression by protein kinase Balpha and the forkhead transcription factor FKHR.

Evidence for insulin response unit-dependent and -independent effects of insulin on promoter activity. Accili D Lilly lecture the struggle for mastery in insulin action: from triumvirate to republic.

Puigserver P, Spiegelman BM Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha PGC-1 alpha : transcriptional coactivator and metabolic regulator. Endocr Rev — Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, Oriente F, Kitamura Y, Altomonte J, Dong H, Accili D, Spiegelman BM Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction.

Zhang W, Patil S, Chauhan B, Guo S, Powell DR, Le J, Klotsas A, Matika R, Xiao X, Franks R, Heidenreich KA, Sajan MP, Farese RV, Stolz DB, Tso P, Koo SH, Montminy M, Unterman TG FoxO1 regulates multiple metabolic pathways in the liver: effects on gluconeogenic, glycolytic, and lipogenic gene expression.

Rena G, Guo S, Cichy SC, Unterman TG, Cohen P Phosphorylation of the transcription factor forkhead family member FKHR by protein kinase B. Guo S, Rena G, Cichy S, He X, Cohen P, Unterman T Phosphorylation of serine by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin-like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence.

Zhang X, Gan L, Pan H, Guo S, He X, Olson ST, Mesecar A, Adam S, Unterman TG Phosphorylation of serine suppresses transactivation by FKHR FOXO1 by multiple mechanisms. Greer EL, Brunet A FOXO transcription factors at the interface between longevity and tumor suppression.

Matsuzaki H, Daitoku H, Hatta M, Tanaka K, Fukamizu A Insulin-induced phosphorylation of FKHR Foxo1 targets to proteasomal degradation.

Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, Rhee J, Adelmant G, Stafford J, Kahn CR, Granner DK, Newgard CB, Spiegelman BM Control of hepatic gluconeogenesis through the transcriptional coactivator PGC Daitoku H, Yamagata K, Matsuzaki H, Hatta M, Fukamizu A Regulation of PGC-1 promoter activity by protein kinase B and the forkhead transcription factor FKHR.

Hirota K, Sakamaki J, Ishida J, Shimamoto Y, Nishihara S, Kodama N, Ohta K, Yamamoto M, Tanimoto K, Fukamizu A A combination of HNF-4 and Foxo1 is required for reciprocal transcriptional regulation of glucokinase and glucosephosphatase genes in response to fasting and feeding. Dentin R, Liu Y, Koo SH, Hedrick S, Vargas T, Heredia J, Yates J III, Montminy M Insulin modulates gluconeogenesis by inhibition of the coactivator TORC2.

Koo SH, Flechner L, Qi L, Zhang X, Screaton RA, Jeffries S, Hedrick S, Xu W, Boussouar F, Brindle P, Takemori H, Montminy M The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Lewis GF, Zinman B, Groenewoud Y, Vranic M, Giacca A Hepatic glucose production is regulated both by direct hepatic and extrahepatic effects of insulin in humans.

Lewis GF, Vranic M, Harley P, Giacca A Fatty acids mediate the acute extrahepatic effects of insulin on hepatic glucose production in humans. Lewis GF, Vranic M, Giacca A Glucagon enhances the direct suppressive effect of insulin on hepatic glucose production in humans.

Lewis GF, Vranic M, Giacca A Role of free fatty acids and glucagon in the peripheral effect of insulin on glucose production in humans. McCall RH, Wiesenthal SR, Shi ZQ, Polonsky K, Giacca A Insulin acutely suppresses glucose production by both peripheral and hepatic effects in normal dogs.

Gupta N, Sandhu H, Goh T, Shah K, Wiesenthal SR, Yoshii H, Chong V, Lam TK, Haber CA, Williams W, Tchipashvili V, Giacca A Insulin inhibits glucose production by a direct effect in diabetic depancreatized dogs during euglycemia. Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, Guilar-Bryan L, Rossetti L Hypothalamic K ATP channels control hepatic glucose production.

Obici S, Zhang BB, Karkanias G, Rossetti L Hypothalamic insulin signaling is required for inhibition of glucose production. Nat Med — Nat Neurosci — Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats.

Buettner C, Camacho RC Hypothalamic control of hepatic glucose production and its potential role in insulin resistance. Endocrinol Metab Clin North Am — Inoue H, Ogawa W, Asakawa A, Okamoto Y, Nishizawa A, Matsumoto M, Teshigawara K, Matsuki Y, Watanabe E, Hiramatsu R, Notohara K, Katayose K, Okamura H, Kahn CR, Noda T, Takeda K, Akira S, Inui A, Kasuga M Role of hepatic STAT3 in brain-insulin action on hepatic glucose production.

Inoue H, Ogawa W, Ozaki M, Haga S, Matsumoto M, Furukawa K, Hashimoto N, Kido Y, Mori T, Sakaue H, Teshigawara K, Jin S, Iguchi H, Hiramatsu R, LeRoith D, Takeda K, Akira S, Kasuga M Role of STAT-3 in regulation of hepatic gluconeogenic genes and carbohydrate metabolism in vivo.

Am J Physiol Endocrinol Metab E29—E Isakoff SJ, Taha C, Rose E, Marcusohn J, Klip A, Skolnik EY The inability of phosphatidylinositol 3-kinase activation to stimulate GLUT4 translocation indicates additional signaling pathways are required for insulin-stimulated glucose uptake.

Sano H, Kane S, Sano E, Miinea CP, Asara JM, Lane WS, Garner CW, Lienhard GE Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. Randhawa VK, Ishikura S, Talior-Volodarsky I, Cheng AW, Patel N, Hartwig JH, Klip A GLUT4 vesicle recruitment and fusion are differentially regulated by Rac, AS, and Rab8A in muscle cells.

Bandyopadhyay G, Standaert ML, Sajan MP, Karnitz LM, Cong L, Quon MJ, Farese RV Dependence of insulin-stimulated glucose transporter 4 translocation on 3-phosphoinositide-dependent protein kinase-1 and its target threonine in the activation loop of protein kinase C-zeta.

Bandyopadhyay G, Kanoh Y, Sajan MP, Standaert ML, Farese RV Effects of adenoviral gene transfer of wild-type, constitutively active, and kinase-defective protein kinase C-lambda on insulin-stimulated glucose transport in L6 myotubes. Garvey WT, Maianu L, Zhu JH, Brechtel-Hook G, Wallace P, Baron AD Evidence for defects in the trafficking and translocation of GLUT4 glucose transporters in skeletal muscle as a cause of human insulin resistance.

Cline GW, Petersen KF, Krssak M, Shen J, Hundal RS, Trajanoski Z, Inzucchi S, Dresner A, Rothman DL, Shulman GI Impaired glucose transport as a cause of decreased insulin-stimulated muscle glycogen synthesis in type 2 diabetes.

N Engl J Med — Ren JM, Marshall BA, Mueckler MM, McCaleb M, Amatruda JM, Shulman GI Overexpression of Glut4 protein in muscle increases basal and insulin-stimulated whole body glucose disposal in conscious mice.

Kelley DE, Mokan M, Mandarino LJ Intracellular defects in glucose metabolism in obese patients with NIDDM. Kim YI, Lee FN, Choi WS, Lee S, Youn JH Insulin regulation of skeletal muscle PDK4 mRNA expression is impaired in acute insulin-resistant states. Bouskila M, Hirshman MF, Jensen J, Goodyear LJ, Sakamoto K Insulin promotes glycogen synthesis in the absence of GSK3 phosphorylation in skeletal muscle.

Am J Physiol Endocrinol Metab E28—E Vaag A, Alford F, Henriksen FL, Christopher M, Beck-Nielsen H Multiple defects of both hepatic and peripheral intracellular glucose processing contribute to the hyperglycaemia of NIDDM.

Nikoulina SE, Ciaraldi TP, Mudaliar S, Mohideen P, Carter L, Henry RR Potential role of glycogen synthase kinase-3 in skeletal muscle insulin resistance of type 2 diabetes.

Dokken BB, Henriksen EJ Chronic selective glycogen synthase kinase-3 inhibition enhances glucose disposal and muscle insulin action in prediabetic obese Zucker rats. Krssak M, Falk PK, Dresner A, DiPietro L, Vogel SM, Rothman DL, Roden M, Shulman GI Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study.

Dobbins RL, Szczepaniak LS, Bentley B, Esser V, Myhill J, McGarry JD Prolonged inhibition of muscle carnitine palmitoyltransferase-1 promotes intramyocellular lipid accumulation and insulin resistance in rats.

Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Rothman DL, DiPietro L, Cline GW, Shulman GI Mitochondrial dysfunction in the elderly: possible role in insulin resistance.

Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. Rasmussen BB, Holmback UC, Volpi E, Morio-Liondore B, Paddon-Jones D, Wolfe RR Malonyl coenzyme A and the regulation of functional carnitine palmitoyltransferase-1 activity and fat oxidation in human skeletal muscle.

Gaster M, Beck-Nielsen H Triacylglycerol accumulation is not primarily affected in myotubes established from type 2 diabetic subjects.

Biochim Biophys Acta — Morino K, Petersen KF, Shulman GI Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction.

Diabetes 55 Suppl 2 :S9—S Samuel VT, Liu ZX, Wang A, Beddow SA, Geisler JG, Kahn M, Zhang XM, Monia BP, Bhanot S, Shulman GI Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease.

Befroy DE, Petersen KF, Dufour S, Mason GF, de Graaf RA, Rothman DL, Shulman GI Impaired mitochondrial substrate oxidation in muscle of insulin-resistant offspring of type 2 diabetic patients.

Turner N, Heilbronn LK Is mitochondrial dysfunction a cause of insulin resistance? Zhang D, Liu ZX, Choi CS, Tian L, Kibbey R, Dong J, Cline GW, Wood PA, Shulman GI Mitochondrial dysfunction due to long-chain Acyl-CoA dehydrogenase deficiency causes hepatic steatosis and hepatic insulin resistance.

Galgani JE, Moro C, Ravussin E Metabolic flexibility and insulin resistance. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB, Lopaschuk GD, Muoio DM Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance.

Bandyopadhyay G, Standaert ML, Kikkawa U, Ono Y, Moscat J, Farese RV Effects of transiently expressed atypical zeta, lambda , conventional alpha, beta and novel delta, epsilon protein kinase C isoforms on insulin-stimulated translocation of epitope-tagged GLUT4 glucose transporters in rat adipocytes: specific interchangeable effects of protein kinases C-zeta and C-lambda.

Ducluzeau PH, Fletcher LM, Vidal H, Laville M, Tavare JM Molecular mechanisms of insulin-stimulated glucose uptake in adipocytes. Diab Metab — Watson RT, Kanzaki M, Pessin JE Regulated membrane trafficking of the insulin-responsive glucose transporter 4 in adipocytes.

Mol Biol Cell — Chiang SH, Baumann CA, Kanzaki M, Thurmond DC, Watson RT, Neudauer CL, Macara IG, Pessin JE, Saltiel AR Insulin-stimulated GLUT4 translocation requires the CAP-dependent activation of TC Abel ED, Peroni O, Kim JK, Kim YB, Boss O, Hadro E, Minnemann T, Shulman GI, Kahn BB Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver.

Shoelson SE, Lee J, Goldfine AB Inflammation and insulin resistance. Lewis GF, Carpentier A, Adeli K, Giacca A Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Kershaw EE, Flier JS Adipose tissue as an endocrine organ.

Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance.

Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia.

Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity.

Biochem Biophys Res Commun — Nieto-Vazquez I, Fernandez-Veledo S, de Alvero C, Lorenzo M Dual role of interleukin-6 in regulating insulin sensitivity in murine skeletal muscle.

Nguyen MT, Satoh H, Favelyukis S, Babendure JL, Imamura T, Sbodio JI, Zalevsky J, Dahiyat BI, Chi NW, Olefsky JM JNK and tumor necrosis factor-alpha mediate free fatty acid-induced insulin resistance in 3T3-L1 adipocytes.

Schmitz-Peiffer C, Browne CL, Oakes ND, Watkinson A, Chisholm DJ, Kraegen EW, Biden TJ Alterations in the expression and cellular localization of protein kinase C isozymes epsilon and theta are associated with insulin resistance in skeletal muscle of the high-fat-fed rat.

Perseghin G, Petersen K, Shulman GI Cellular mechanism of insulin resistance: potential links with inflammation.

Thank you for Insulin and carbohydrate metabolism nature. You are using metabolisk browser Insulin and carbohydrate metabolism with limited support for CSS. Insulin and carbohydrate metabolism obtain the best Weight gain advice, we cafbohydrate you Improve exercise flexibility a more carbohydratf to date browser or qnd off compatibility mode in Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. Insulin resistance is the primary pathophysiology underlying metabolic syndrome and type 2 diabetes 12. Previous metagenomic studies have described the characteristics of gut microbiota and their roles in metabolizing major nutrients in insulin resistance 3456789.

Author: Vir

0 thoughts on “Insulin and carbohydrate metabolism

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com