Category: Family

Insulin sensitivity and insulin signaling

Insulin sensitivity and insulin signaling

Sibnaling of Berry Infused Water on intracellular GLUT4 vesicles in adipocytes: Muscle recovery foods for a Insullin mode of regulation. When the insulin is introduced to the liver, it connects to the insulin receptors already present, that is tyrosine kinase receptor. One of these pathways, involves the PI 3 K enzyme Phosphoinositide 3-kinase. Insulin sensitivity and insulin signaling

Insulin sensitivity and insulin signaling -

The α-subunits act as insulin receptors and the insulin molecule acts as a ligand. Together, they form a receptor-ligand complex. Binding of insulin to the α-subunit results in a conformational change of the protein, which activates tyrosine kinase domains on each β-subunit.

The tyrosine kinase activity causes an autophosphorylation of several tyrosine residues in the β-subunit. The phosphorylation of 3 residues of tyrosine is necessary for the amplification of the kinase activity.

This autophosphorylation triggers the activation of the docking proteins, in this case IRS on which PhosphatidylinositolKinase PI-3K can be attached or GRB2 where the ras Guanine nucleotide exchange factor GEF also known as SOS can be attached.

PI-3K causes the phosphorylation of PIP2 to PIP3. This protein acts as a docking site for PDPK1 and Protein kinase B also known as AKT , which is then phosphorylated by the latter and PK2 to be activated. This leads to crucial metabolic functions such as synthesis of lipids, proteins and glycogen.

It also leads to cell survival and cell proliferation. Most importantly, the PI-3K pathway is responsible for the distribution of glucose for important cell functions. For example, the suppression of hepatic glucose synthesis and the activation of glycogen synthesis.

Hence, AKT possesses a crucial role in the linkage of the glucose transporter GLUT4 to the insulin signaling pathway.

The activated GLUT4 will translocate to the cell membrane and promotes the transportation of glucose into the intracellular medium. The Ras-GEF stimulates the exchange of GDP to GTP in the RAS protein, causing it to activate.

Ras then activates the Mitogen-activated protein kinase MAP-Kinase route, which ultimately results in changes in protein activity and gene expression. Thus, insulin's role is more of a promoter for the usage of glucose in the cells rather than neutralizing or counteracting it.

PI-3K is one of the important components in the regulation of the insulin signaling pathway. It maintains the insulin sensitivity in the liver. PI-3K is composed of a regulatory subunit P85 and a catalytic subunit P P85 regulates the activation of PI-3K enzyme.

It was noted that an increase of P85 a isoform of P85 results in a competition between the later and the PP complex to the IRS binding site, reducing the PI-3k activity and leading to insulin resistance.

Insulin resistance refers also to Type 2 diabetes. It was also noted that increased serine phosphorylation of IRS is involved in the insulin resistance by reducing their ability to attract PI3K.

The serine phosphorylation can also lead to degradation of IRS Signal transduction is a mechanism in which the cell responds to a signal from the environment by activating several proteins and enzymes that will give a response to the signal.

Feedback mechanism might involve negative and positive feedbacks. In the negative feedback, the pathway is inhibited and the result of the transduction pathway is reduced or limited. In positive feedback, the transduction pathway is promoted and stimulated to produce more products.

Insulin secretion results in positive feedback in different ways. Firstly, insulin increases the uptake of glucose from blood by the translocation and exocytosis of GLUT4 storage vesicles in the muscle and fat cells.

Secondly, it promotes the conversion of glucose into triglyceride in the liver, fat, and muscle cells. Finally, the cell will increase the rate of glycolysis within itself to break glucose in the cell into other components for tissue growth purposes. An example of positive feedback mechanism in the insulin transduction pathway is the activation of some enzymes that inhibit other enzymes from slowing or stopping the insulin transduction pathway which results in improved intake of the glucose.

One of these pathways, involves the PI 3 K enzyme Phosphoinositide 3-kinase. This pathway is responsible for activating glycogen, lipid-protein synthesis, and specific gene expression of some proteins which will help in the intake of glucose.

Different enzymes control this pathway. Some of these enzymes constrict the pathway causing a negative feedback like the GSK-3 pathway. Other enzymes will push the pathway forward causing a positive feedback like the AKT and P70 enzymes.

When insulin binds to its receptor, it activates the glycogen synthesis by inhibiting the enzymes that slow down the PI 3 K pathway such as PKA enzyme. At the same time, it will promote the function of the enzymes that provide a positive feedback for the pathway like the AKT and P70 enzymes.

Image to help explain the function of the proteins mentioned above in the positive feedback. When insulin binds to the cell's receptor, it results in negative feedback by limiting or stopping some other actions in the cell.

It inhibits the release and production of glucose from the cells which is an important part in reducing the glucose blood level. Insulin will also inhibit the breakdown of glycogen into glucose by inhibiting the expression of the enzymes that catalyzes the degradation of Glycogen.

An example of negative feedback is slowing or stopping the intake of glucose after the pathway was activated. Negative feedback is shown in the insulin signal transduction pathway by constricting the phosphorylation of the insulin-stimulated tyrosine.

When activated, this enzyme provides a negative feedback by catalyzing the dephosphorylation of the insulin receptors. Insulin is synthesized and secreted in the beta cells of the islets of Langerhans. The pleiotropic effects of insulin action on cell growth and metabolism result from a complex interaction between rapid phosphorylation-dependent signalling [ 35 , 36 ] and slower changes in gene expression [ 37 ].

For example, the effect of insulin on glucose transport in skeletal muscle and adipocytes is dependent on the movement of pre-existing vesicles containing GLUT4 glucose transporters to the plasma membrane [ 38 ] and is dependent on AS phosphorylation by Akt [ 39 ], while glycogen synthesis and glycolytic and oxidative glucose metabolism are supported by increased mRNA expression of glycogen synthase 1 [ 40 ], hexokinase 2 [ 41 ] and many components of the mitochondrial electron transport chain [ 42 ].

Insulin also regulates several key mechanisms involved in gene expression, with the regulation of mRNA transcription being the best studied [ 43 ]. This important aspect of insulin action is accomplished by insulin-induced changes in phosphorylation, expression, processing and translocation of a variety of transcription factors, leading to stimulation or inhibition of gene transcription.

FOX proteins represent a large family of transcription factors, of which FOXOs FOXO1, FOXO3, FOXO4 and FOXO6 are the most well-characterised regulators of downstream insulin signalling. Here, the effect of insulin is one of negative regulation Fig. This creates interaction sites for FOXOs with phosphoserine-binding proteins, resulting in their retention in the cytoplasm and decreased transcriptional activity in the nucleus [ 44 , 45 ].

Thus, insulin-induced phosphorylation of FOXOs results in reduced hepatic gluconeogenesis [ 46 ], inhibition of muscle autophagy and protein degradation [ 30 , 47 ] and regulation of adipocyte differentiation [ 48 ].

Reciprocal regulation of FOX transcription factors by insulin. a Under feeding or other conditions where insulin action is high, FOXOs are phosphorylated by Akt on serine residues, creating interaction sites for proteins, leading to cytoplasmic retention and inhibited transcriptional activity.

Under these conditions, increased Akt and mTORC1 activity inhibits GSK3 signalling and relieves FOXKs from inhibitory GSK3-mediated phosphorylation, leading to increased nuclear translocation and FOXK transcriptional activity.

Under these conditions, increased GSK3 activity leads to increased FOXK phosphorylation and interaction with phosphoserine-binding proteins, resulting in cytoplasmic retention and decreased transcriptional activity.

Line thickness indicates strength of signalling activity, with thicker lines indicating stronger signalling activity. Another emerging class of FOX proteins that act in insulin signalling are the FOXK1 and FOXK2 transcription factors [ 21 , 52 ].

In contrast to FOXOs, which are turned off by insulin, FOXKs display increased nuclear localisation and transcriptional activity following insulin stimulation Fig. In the basal state, GSK3 phosphorylates FOXKs leading to increased interaction with proteins and nuclear exclusion Fig.

In hepatocytes, FOXKs regulate genes involved in the cell cycle, apoptosis and lipid metabolism [ 21 ], while in adipocytes and muscle, FOXKs promote glucose transport and lactate production by stimulation of glycolytic metabolism and inhibition of mitochondrial pyruvate oxidation [ 53 ].

In addition to phosphorylation, insulin also regulates the expression and processing of transcription factors. For example, sterol regulatory element binding proteins SREBP 1 and 2 are important regulators of triacylglycerol and cholesterol synthesis and are synthesised as precursors that reside in the endoplasmic reticulum ER.

A re-emerging concept in insulin control of gene expression is the possibility of direct effects of the insulin receptor itself. Studies from over 40 years ago showed binding of insulin to nuclear preparations [ 55 ]. The significance of such findings has only come to light by recent studies demonstrating interactions between the insulin receptor and FOXK1 [ 21 ] and interactions of the insulin receptor with RNA polymerase II Pol II on DNA in the nucleus [ 56 ].

Indeed, chromatin immunoprecipitation followed by sequencing ChIP-seq analysis of HepG2 hepatocytes revealed ~ peaks bound by the insulin receptor, many overlapping with Pol II sites on promoters.

These occur in genes involved in a variety of cellular functions including lipid metabolism, translation and immunity, as well as genes involved in pathophysiological states, such as diabetes. Type 2 diabetes affects more than million adults worldwide and its prevalence continues to increase at epidemic rates, thus posing one of the greatest public health challenges to society [ 57 ].

This is the result of both genetic and environmental factors. While it remains debated whether insulin resistance and relative beta cell failure constitute the primary defect in type 2 diabetes [ 58 , 59 ], a 25 year prospective longitudinal study of people at high genetic risk of developing type 2 diabetes has demonstrated that insulin resistance precedes and predicts disease development [ 60 ].

Likewise, family studies have shown that glucose tolerant offspring of parents with type 2 diabetes show insulin resistance, while loss of first-phase insulin secretion was observed in those that developed impaired glucose tolerance [ 61 ]. Clamp and MRI studies have revealed skeletal muscle as a primary site of insulin resistance in the offspring of parents with type 2 diabetes, with the muscle of these individuals exhibiting reduced glucose uptake and reduced glycogen synthesis before hyperglycaemia develops [ 62 ].

This impaired glucose metabolism has been attributed to a number of defects, including decreased glucose transport [ 63 ], lower rates of insulin-induced ATP production [ 42 ] and reduced expression of genes involved in mitochondrial function [ 64 , 65 ].

The major question that remains is what are the fundamental defects leading to insulin resistance and how do cell-intrinsic vs cell-extrinsic factors contribute to these defects? Conversely, cell-intrinsic factors are those that persist after removal or normalisation of all extrinsic factors.

These are most likely due to genetic or epigenetic effects, but may or may not be in the insulin signalling pathway itself. How each of these might contribute to insulin resistance in type 2 diabetes is discussed in the following sections. In type 2 diabetes, most attention has focused on extrinsic factors contributing to insulin resistance, including the role of adipose tissue, circulating metabolites, inflammatory signals and the gut microbiome [ 66 , 67 , 68 ] Fig.

Accumulation of ceramides can also activate protein phosphatase 2A PP2A and PKCζ, inhibiting Akt2. Adipose tissue expansion is also associated with increased adipose tissue inflammation and hypoxia [ 77 ], promoting recruitment of proinflammatory macrophages [ 78 ] that secrete cytokines, such as TNF-α and IL-6, which further worsen insulin resistance by activation of the TNF-α receptor TNFR and other cytokine receptors [ 79 ].

Extrinsic factors contributing to insulin resistance. Several environmental factors may lead to systemic changes affecting multiple tissues and contributing to impaired insulin signalling.

Obesity negatively correlates with circulating levels of adiponectin [ ] and signalling lipids with beneficial properties, such as 12,dihydroxy-9Z-octadecenoic acid 12,diHOME [ ] and branched fatty acid esters of hydroxy fatty acids FAHFAs [ ].

Overnutrition leads to adipose tissue expansion and increased release of cytokines and other inflammatory mediators e. JNK, IKK and novel PKCs [nPKCs] and increased IRS serine phosphorylation, and due to increased transcription of SOCS proteins, which interfere with IRS tyrosine phosphorylation.

Adipose tissue insulin resistance is associated with ectopic lipid accumulation, mitochondrial dysfunction and reactive oxygen species ROS generation, and ER stress in insulin-sensitive tissues.

Adipose tissue expansion in obesity may also have an impact on systemic metabolism through altered release of exosomal miRNAs. Insulin signalling proteins are shown in blue and intracellular mediators of cytokine receptors and other stress signals are shown in green.

DAG, diacylglycerol; IRE1, inositol-requiring enzyme 1; JAK, Janus kinase; STAT, signal transducer and activator of transcription; TLR4, Toll-like receptor 4; TNFR, TNF-α receptor; UPR, unfolded protein response; XBP1, X-box binding protein 1. Circulating branched-chain amino acids BCAAs and aromatic amino acids isoleucine, leucine, valine, phenylalanine and tyrosine are also associated with insulin resistance [ 67 ], and lowering BCAA levels can improve insulin sensitivity, at least in mice [ 83 ].

Gut microbiota may also play a role in regulating BCAA supply, as well as the production of short-chain fatty acids and other metabolites, which, in turn, have an impact on systemic insulin sensitivity [ 85 ].

Recently, we and others have shown that adipose tissue can also crosstalk with other tissues through secretion of exosomal microRNAs miRNAs [ 88 , 89 ]; however, how this fits in the regulation of insulin sensitivity at a signalling level remains to be determined.

In vitro approaches, where cells are cultured under controlled conditions, provides an opportunity to minimise the influence of extrinsic factors and isolate cell-autonomous determinants of insulin resistance, which are more closely linked to the genetic and epigenetic alterations underlying type 2 diabetes.

Skeletal muscle biopsies and primary cultured myoblasts derived from people with type 2 diabetes show insulin resistance and several metabolic defects. However, primary cell models have limited usefulness for the definition of molecular mechanisms underlying insulin resistance due to limits in expandability and ability for screening using RNA interference RNAi , chemical genetics or CRISPR.

Such iPSC modelling has been applied to severe insulin resistance caused by insulin receptor mutations [ 95 , 96 , 97 ] and other forms of genetically determined type 2 diabetes and obesity [ 98 , 99 ].

Recently, we have applied the iPSC technology to study signalling defects that underlie skeletal muscle insulin resistance in type 2 diabetes [ ]. Only a small proportion of these abnormalities are in classical insulin-regulated phosphorylations that define critical nodes in insulin action [ ].

More importantly, type 2 diabetic iMyos show a large degree of perturbations in pathways outside of the canonical insulin signalling pathway and not regulated by insulin Fig.

These findings clearly open our view to a wider definition of mechanisms of insulin resistance at the molecular and cellular level that needs to be taken into account in understanding the pathogenesis of type 2 diabetes.

Intrinsic factors contributing to insulin resistance. Cell-autonomous insulin resistance is associated with defects in glucose transport, mitochondrial metabolism and insulin signalling. Global phosphoproteomics of iMyos from individuals with type 2 diabetes reveal a network of signalling defects that underlie skeletal muscle insulin resistance [ ].

Proteins linked to insulin action and metabolism are indicated in blue and site-specific effects of type 2 diabetes evidenced by increased and decreased basal phosphorylation are shown in orange and green, respectively.

Groups of multiple proteins of the same category are shown in purple and non-labelled circles indicate groups of up- or downregulated phosphosites. Faded shading of text boxes indicates lower cytoplasm abundance. Ac, acetyl group; ARHGAP, Rho GTPase activating protein; ARHGEF, Rho guanine nucleotide exchange factor; HDAC, histone deacetylase; KAT, lysine acetyltransferase; KDM, lysine demethylase; Me, methyl group; MEF2C, myocyte enhancer factor 2C; PDHA1, pyruvate dehydrogenase E1 subunit alpha 1; SETD, SET domain containing histone lysine methyltransferase; SR, serine- and arginine-rich splicing factor; T2D, type 2 diabetes; TBC1D1, TBC1 domain family member 1; TSC2, tuberous sclerosis 2; U1, U1 small nuclear ribonucleoprotein complex; U2, U2 small nuclear ribonucleoprotein complex.

A major challenge going forward will be to identify the molecular defect s that drive these signalling changes.

Possibilities include kinases and phosphatases, a wide range of co-regulators of the activity of kinases and phosphatases, redox balance, ionic milieu, scaffolding proteins and other factors.

While these alterations could represent some form of metabolic memory or epigenetic regulation due to altered DNA methylation [ , ], this seems unlikely since genetic reprogramming of iPSCs is known to erase most epigenetic marks [ ]. Likewise, while genome-wide association studies GWAS have collectively identified over independent SNPs associated with type 2 diabetes [ ], few of these are in proteins active in insulin signalling.

Furthermore, although some SNPs may fall into regulatory regions acting on adjacent or even distant genes, most of these GWAS variants occur in non-coding regions of the genome [ , ]. While there has been some progress in linking GWAS variants to alterations in beta cell function, insulin sensitivity and energy balance [ , ], even taken together, all GWAS loci account for only a small fraction of the strong familial clustering of type 2 diabetes, leaving understanding the primary defect a major challenge.

Advances in profiling technologies have led to a greater appreciation of the potential role of non-coding RNAs, especially miRNAs and long non-coding RNAs lncRNAs , in the control of cellular metabolism.

While some miRNA changes in type 2 diabetes may result from tissue crosstalk through exosomal delivery [ ], miRNA profiling of cultured myoblasts from donors with type 2 diabetes also revealed some modest, but significant, changes compared with control donors [ ].

Similar to miRNAs, some lncRNAs are also regulated by insulin and other physiological cues [ 37 , ], and are dysregulated in type 2 diabetes [ ], resulting in abnormal insulin signalling [ ].

The finding of altered phosphorylation and gene expression of factors involved in mRNA splicing in iMyos [ ] and skeletal muscle biopsies from individuals with type 2 diabetes [ ] could provide another link between genetic regulation and the insulin resistance of type 2 diabetes.

Insulin and IGF-1 signalling is present in virtually every cell of the body and plays a central role in the control of metabolism, growth and differentiation. In spite of significant progress, understanding the primary driver of altered insulin receptor signalling in type 2 diabetes, obesity and the metabolic syndrome represents a continuing challenge.

Nicole A. Yahui Kong, Rohit B. Sharma, … Laura C. Macleod JJR Treatment of diabetes mellitus by pancreatic extracts. Can Med Assoc J 12 6 — Google Scholar.

Himsworth HP Diabetes mellitus: its differentiation into insulin-sensitive and insulin-insensitive types. Int J Epidemiol 42 6 — Article CAS PubMed Google Scholar.

Freychet P, Roth J, Neville DM Jr Insulin receptors in the liver: specific binding of I insulin to the plasma membrane and its relation to insulin bioactivity. Proc Natl Acad Sci U S A 68 8 — Article CAS PubMed PubMed Central Google Scholar. Kasuga M, Zick Y, Blithe DL, Crettaz M, Kahn CR Insulin stimulates tyrosine phosphorylation of the insulin receptor in a cell-free system.

Nature — Chevalier S, Burgess SC, Malloy CR, Gougeon R, Marliss EB, Morais JA The greater contribution of gluconeogenesis to glucose production in obesity is related to increased whole-body protein catabolism. Diabetes 55 3 — Perry RJ, Camporez JP, Kursawe R et al Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes.

Cell 4 — Seino S, Seino M, Nishi S, Bell GI Structure of the human insulin receptor gene and characterization of its promoter.

Proc Natl Acad Sci U S A 86 1 — Belfiore A, Malaguarnera R, Vella V et al Insulin receptor isoforms in physiology and disease: an updated view. Endocr Rev 38 5 — Article PubMed PubMed Central Google Scholar.

Cai W, Sakaguchi M, Kleinridders A et al Domain-dependent effects of insulin and IGF-1 receptors on signalling and gene expression. Nat Commun Urso B, Cope DL, Kalloo-Hosein HE et al Differences in signaling properties of the cytoplasmic domains of the insulin receptor and insulin-like growth factor receptor in 3T3-L1 adipocytes.

J Biol Chem 43 — Scapin G, Dandey VP, Zhang Z et al Structure of the insulin receptor-insulin complex by single-particle cryo-EM analysis. Uchikawa E, Choi E, Shang G, Yu H, Bai XC Activation mechanism of the insulin receptor revealed by cryo-EM structure of the fully liganded receptor-ligand complex.

eLife 8. Thirone AC, Huang C, Klip A Tissue-specific roles of IRS proteins in insulin signaling and glucose transport. Trends Endocrinol Metab 17 2 — Araki E, Lipes MA, Patti ME et al Alternative pathway of insulin signalling in mice with targeted disruption of the IRS-1 gene.

Kaburagi Y, Satoh S, Tamemoto H et al Role of insulin receptor substrate-1 and pp60 in the regulation of insulin-induced glucose transport and GLUT4 translocation in primary adipocytes. J Biol Chem 41 — Withers DJ, Gutierrez JS, Towery H et al Disruption of IRS-2 causes type 2 diabetes in mice.

Cell Signal — Ussar S, Bezy O, Bluher M, Kahn CR Glypican-4 enhances insulin signaling via interaction with the insulin receptor and serves as a novel adipokine.

Diabetes 61 9 — Maddux BA, Goldfine ID Membrane glycoprotein PC-1 inhibition of insulin receptor function occurs via direct interaction with the receptor alpha-subunit. Diabetes 49 1 — Batista TM, Dagdeviren S, Carroll SH et al Arrestin domain-containing 3 Arrdc3 modulates insulin action and glucose metabolism in liver.

Proc Natl Acad Sci U S A 12 — Sakaguchi M, Cai W, Wang CH et al FoxK1 and FoxK2 in insulin regulation of cellular and mitochondrial metabolism. Nat Commun 10 1 Choi E, Zhang X, Xing C, Yu H Mitotic checkpoint regulators control insulin signaling and metabolic homeostasis. Cell 3 — Mora A, Komander D, van Aalten DM, Alessi DR PDK1, the master regulator of AGC kinase signal transduction.

Semin Cell Dev Biol 15 2 — Bilanges B, Posor Y, Vanhaesebroeck B PI3K isoforms in cell signalling and vesicle trafficking.

Nat Rev Mol Cell Biol 20 9 — J Biol Chem 39 — Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Matsumoto M, Pocai A, Rossetti L, Depinho RA, Accili D Impaired regulation of hepatic glucose production in mice lacking the forkhead transcription factor Foxo1 in liver.

Cell Metab 6 3 — Nakae J, Barr V, Accili D Differential regulation of gene expression by insulin and IGF-1 receptors correlates with phosphorylation of a single amino acid residue in the forkhead transcription factor FKHR. EMBO J 19 5 — J Clin Invest 9 — Sancak Y, Thoreen CC, Peterson TR et al PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase.

Mol Cell 25 6 — Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC, Blenis J Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin mTOR -mediated downstream signaling.

A second molecular mechanism that can potentially lead to insulin resistance is a disruption in the balance between the amounts of the PI 3-kinase subunits PI 3-kinase belongs to the class 1a 3-kinases 82 , which exist as heterodimers, consisting of a regulatory subunit p85 , which is tightly associated with a catalytic subunit, p The regulatory subunit, p85, is encoded by at least three genes that generate highly homologous products.

Two isoforms are termed p85α PIK3R1 and p85β products of the two genes. Three splice variants of p85α have been reported, including p85α itself, p55α, and p50α. The third gene product is p55γ. p85α, however, appears to be the most abundant isoform Normally, the regulatory subunit exists in stoichiometric excess to the catalytic one, resulting in a pool of free p85 monomers not associated with the p catalytic subunit.

Thus, there exists a balance between the free p85 monomer and the pp heterodimer, with the latter being responsible for the PI 3-kinase activity.

Increases or decreases in expression of p85 shift this balance in favor of either free p85 or pp complexes 83 — Because the p85 monomer and the pp heterodimer compete for the same binding sites on the tyrosine-phosphorylated IRS proteins, an imbalance could cause either increased or decreased PI 3-kinase activity Fig.

This possibility has been recently supported by studies in insulin-resistant states induced by human placental growth hormone 87 , obesity, and type 2 diabetes 58 and by short-term overfeeding of lean nondiabetic women One of the first indications that an imbalance between the abundance of p85 and p can alter PI 3-kinase activity came from experiments with l -6 cultured skeletal muscle cells treated with dexamethazone This treatment significantly reduced PI 3-kinase activity, despite an almost fourfold increase in expression of p85α no change in p85β and only a minimal increase in p The authors concluded that p85α competes with the pp heterodimer, thus, reducing PI 3-kinase activity Table 2.

To determine this ratio, the authors immunodepleted p and blotted both the immunoprecipitates and the supernatant with p85 antibody.

The amounts of p85 in the p immunoprecipitates denote p85 bound to p, while the amount of p85 in the supernatant represents free excess p The greater the ratio of bound to free, the greater the insulin sensitivity the mice display. The same group of authors then overexpressed p85α in cultured cells.

This overexpression significantly inhibited the PI 3-kinase activity 85 , 86 , Overexpression of p50α or p55α did not inhibit PI 3-kinase activity to the same extent. These experimental results were consistent with the competition hypothesis. Recently, Barbour and colleagues 87 , 93 demonstrated that insulin resistance of pregnancy is likely due to increased expression of skeletal muscle p85 in response to increasing concentrations of human placental growth hormone.

Furthermore, women remaining insulin resistant postpartum have been found to display higher levels of p85 in the muscle Thus, results reported in the literature support the hypothesis that the p85 monomer completes with a pp dimer and that the removal of the excess of p85 improves insulin sensitivity by allowing the remaining isoforms to bring p to its site of action.

Finally, in a small study of eight healthy lean women without a family history of diabetes, Cornier et al. Within this experimental time frame, overfeeding did not cause any change in serine phosphorylation of either IRS-1 or S6K1 88 , suggesting that increased expression of p85α may be an early molecular step in the pathogenesis of the nutritionally induced insulin resistance.

There have been substantial strides made in our understanding of the genesis of insulin resistance. A number of serine kinases that could phosphorylate serine residues of IRS-1 and thereby diminish insulin signal transduction have been identified. Potential triggering mechanisms such as mitochondrial dysfunction have also been proposed and supported by experimental and observational data.

On the other hand, an additional and possibly complementary mechanism involving increased expression of p85α has also been found to play an important role in the pathogenesis of insulin resistance under certain circumstances.

Further studies are needed in order to evaluate this hypothesis. Inhibition of the metabolic insulin signaling. IRS-1 is phosphorylated by the tyrosine kinase of the insulin receptor in response to insulin binding. PI 3-kinase is then activated and initiates a downstream cascade of events leading to the phosphorylation and activation of Akt, mTOR, and p70S6 kinase.

Activation of Akt appears to be important for glucose transport, while activation of mTOR and p70S6 kinase participates in the process of protein synthesis.

In addition, serine phosphorylation of IRS-1 can be promoted by JNK, PKC, IKKβ, and TNFα. B : Increased expression of p85α monomer competes with and displaces the pp heterodimer from the IRS-1 binding sites.

The resultant decrease in association of p with IRS-1 diminishes PI 3-kinase activity and the downstream effects of this kinase. Steroids, growth hormone GH , human placental growth hormone hPGH , short-term overfeeding, obesity, and type 2 diabetes T2DM have been shown to increase p85α expression see text for details and references.

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. Advanced Search. User Tools Dropdown. Sign In. Skip Nav Destination Close navigation menu Article navigation.

Volume 55, Issue 8. Previous Article. Article Navigation. Perspectives in Diabetes August 01 Molecular Mechanisms of Insulin Resistance: Serine Phosphorylation of Insulin Receptor Substrate-1 and Increased Expression of p85α : The Two Sides of a Coin Boris Draznin Boris Draznin.

This Site. Google Scholar. Address correspondence and reprint requests to Dr. Boris Draznin, Research Service , Denver VA Medical Center, Clermont St. E-mail: boris. draznin med. Diabetes ;55 8 — Article history Received:. Get Permissions. toolbar search Search Dropdown Menu.

toolbar search search input Search input auto suggest. View large Download slide. TABLE 1 Causes of IRS-1 serine phosphorylation. View Large. TABLE 2 Causes of an imbalance between PI 3-kinase subunits. Steroids 89 Growth hormone 93 Human placental growth hormone 87 , 93 Short-term overfeeding 88 Obesity and diabetes Olefsky JM: The insulin receptor: a multifunctional protein.

Reaven GM: Role of insulin resistance in human disease. Ginsberg H: Insulin resistance and cardiovascular disease. J Clin Invest. Shulman GI: Cellular mechanisms of insulin resistance in humans.

Am J Cardiol. Ullrich A, Bell JR, Chen EY, Herrera R, Petruzzelli IM, Dull TJ, Gray A, Coussens L, Liao Y-C, Tsubokawa M, Mason A, Seeburg PH, Grunfeld C, Rosen OM, Ramachandran J: Human insulin receptor and its relationship to the tyrosine kinase family of oncogenes. Ebina Y, Ellis L, Jarnagin K, Edery M, Grat L, Clauser E, Ou J-H, Masiarz F, Kan YW, Goldfine ID, Roth RA, Rutter WJ: The human insulin receptor cDNA: the structural basis for hormone-activated transmembrane signaling.

Seino S, Seino M, Nishi S, Bell GI: Structure of human insulin receptor gene and characterization of its promoter. Proc Natl Acad Sci U S A. Kasuga M, Karisson FA, Kahn CR: Insulin stimulates the phosphorylation of the 95, Dalton subunit of its own receptor.

Wilden PA, Siddle K, Haring E, Backer JM, White MF, Kahn CR: The role of insulin receptor-kinase domain autophosphorylation in receptor-mediated activities. J Biol Chem. De Meyts P, Christoffersen CT, Tornqvist H, Seedorf K: Insulin receptors and insulin action. Curr Opin Endocrinol Diabetes.

The homeostatic control of blood glucose is determined Signaliing two major factors: the concentration of insulin in sensifivity circulation, Berry Infused Water correlates with Berry Infused Water cell function, and the sensitivity of signalling organs an. muscle, adipose tissue and Insulinn to insulin. Natural weight loss foods resistance is defined as Greek yogurt granola failure of target organs to respond to physiological insulin concentrations, thus leading to the development of diabetes, an ever-increasing epidemic of the 21st century. Ongoing studies in our lab focus on the molecular basis of insulin resistance and its effects on growth and survival of the pancreatic inslin β-cells. We could show that inducers of insulin resistance e. pro-inflammatory cytokines exploit phosphorylation-based negative feedback control mechanisms, to uncouple the insulin receptor IR from its downstream effectors, the IRS insilin IRS-1 and IRS-2 and thereby terminate insulin signal transduction.

Insulin sensitivity and insulin signaling -

Precise modulation of this pathway is vital for adaption as the individual moves from the fed to the fasted state. The positive and negative modulators acting on different steps of the signaling pathway, as well as the diversity of protein isoform interaction, ensure a proper and coordinated biological response to insulin in different tissues.

Whereas genetic mutations are causes of rare and severe insulin resistance, obesity can lead to insulin resistance through a variety of mechanisms. Understanding these pathways is essential for development of new drugs to treat diabetes, metabolic syndrome, and their complications.

Copyright © by Cold Spring Harbor Laboratory Press. Insulin Receptor Signaling in Normal and Insulin-Resistant States Jérémie Boucher 1 , 2 , André Kleinridders 1 , 2 and C.

Abstract In the wake of the worldwide increase in type-2 diabetes, a major focus of research is understanding the signaling pathways impacting this disease.

CiteULike Delicious Digg Facebook Reddit Twitter What's this? Also in this Collection. This Article doi: a Cold Spring Harb. Article Category Perspective Molecular Pathology.

Services Alert me when this article is cited Alert me if a correction is posted Similar articles in this journal Similar articles in Web of Science Similar articles in PubMed Download to citation manager Permissions.

Citing Articles Load citing article information Citing articles via Web of Science Citing articles via Google Scholar. Google Scholar Articles by Boucher, J. Articles by Kahn, C. Search for related content. Subject Collections Signaling by Receptor Tyrosine Kinases. Advanced Search. User Tools Dropdown.

Sign In. Skip Nav Destination Close navigation menu Article navigation. Volume 55, Issue 8. Previous Article. Article Navigation. Perspectives in Diabetes August 01 Molecular Mechanisms of Insulin Resistance: Serine Phosphorylation of Insulin Receptor Substrate-1 and Increased Expression of p85α : The Two Sides of a Coin Boris Draznin Boris Draznin.

This Site. Google Scholar. Address correspondence and reprint requests to Dr. Boris Draznin, Research Service , Denver VA Medical Center, Clermont St.

E-mail: boris. draznin med. Diabetes ;55 8 — Article history Received:. Get Permissions. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. View large Download slide. TABLE 1 Causes of IRS-1 serine phosphorylation. View Large. TABLE 2 Causes of an imbalance between PI 3-kinase subunits.

Steroids 89 Growth hormone 93 Human placental growth hormone 87 , 93 Short-term overfeeding 88 Obesity and diabetes Olefsky JM: The insulin receptor: a multifunctional protein.

Reaven GM: Role of insulin resistance in human disease. Ginsberg H: Insulin resistance and cardiovascular disease. J Clin Invest. Shulman GI: Cellular mechanisms of insulin resistance in humans. Am J Cardiol. Ullrich A, Bell JR, Chen EY, Herrera R, Petruzzelli IM, Dull TJ, Gray A, Coussens L, Liao Y-C, Tsubokawa M, Mason A, Seeburg PH, Grunfeld C, Rosen OM, Ramachandran J: Human insulin receptor and its relationship to the tyrosine kinase family of oncogenes.

Ebina Y, Ellis L, Jarnagin K, Edery M, Grat L, Clauser E, Ou J-H, Masiarz F, Kan YW, Goldfine ID, Roth RA, Rutter WJ: The human insulin receptor cDNA: the structural basis for hormone-activated transmembrane signaling.

Seino S, Seino M, Nishi S, Bell GI: Structure of human insulin receptor gene and characterization of its promoter. Proc Natl Acad Sci U S A. Kasuga M, Karisson FA, Kahn CR: Insulin stimulates the phosphorylation of the 95, Dalton subunit of its own receptor.

Wilden PA, Siddle K, Haring E, Backer JM, White MF, Kahn CR: The role of insulin receptor-kinase domain autophosphorylation in receptor-mediated activities. J Biol Chem. De Meyts P, Christoffersen CT, Tornqvist H, Seedorf K: Insulin receptors and insulin action.

Curr Opin Endocrinol Diabetes. Rhodes CJ, White MF: Molecular insights into insulin action and secretion. Eur J Clin Invest.

White MF, Shoelson SE, Keutmann H, Kahn CR: A cascade of tyrosine autophosphorylation in the beta-subunit activates the phosphotransferase of the insulin receptor. Tornqvist HE, Avruch J: Relationship of site-specific beta subunit tyrosine autophosphorylation to insulin activation of the insulin receptor tyrosine protein kinase activity.

Myers MG Jr, White MF: Insulin signal transduction and the IRS proteins. Annu Rev Pharmacol Toxicol. Paz K, Voliovitch H, Hadari YR, Roberts CT, LeRoith D, Zick Y: Interaction between the insulin receptor and its downstream effectors. Kolterman OG, Insel J, Saekow M, Olefsky JM: Mechanisms of insulin resistance in human obesity: evidence for receptor and post-receptor defects.

Marshal S, Olefsky JM: Effects if insulin incubation on insulin binding, glucose transport, and insulin degradation by isolated rat adipocytes: evidence for hormone-induced desensitization at the receptor and post-receptor level. Haring HU: The insulin receptor: signaling mechanism and contribution to the pathogenesis of insulin resistance.

Cheatham B, Kahn CR: Insulin action and the insulin signaling network. Endocrine Rev. Kahn CR: Insulin action, diabetogenes, and the cause of type 2 diabetes. Cheatham B: Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp70S6 kinase, DNA synthesis, and glucose transporter translocation.

Mol Cell Biol. Shepherd PR, Nave BT, Siddle K: Insulin stimulation of glycogen synthesis and glycogen synthase activity is blocked by wortmannin and rapamycin in 3T3—L1 adipocytes: evidence for the involvement of phosphoinositide 3-kinase and p70 ribosomal protein-S6 kinase. Biochem J. Lazar D: Mitogen-activated kinase kinase inhibition does not block the stimulation of glucose utilization by insulin.

Sutherland C, Waltner-Law M, Gnudi L, Kahn BB, Granner DK: Activation of the Ras mitogen-activated protein kinase-ribosomal protein kinase pathway is not required for the repression of phosphoenolpyruvate carboxykinase gene transcription by insulin.

Bandyopadhyay GK, Standaert ML, Zhao L, Yu B, Avignon A, Galloway L, Karnam P, Moscat J, Farese RV: Activation of protein kinase α, β, and ξ by insulin in 3T3—L1 cells: transfection studies suggest a role for PKC-zeta in glucose transport.

Cusi K, Maezono K, Osman A, Pendergrass M, Patti ME, Pratipanawatr T, DeFronzo RA, Kahn CR, Mandarino LJ: Insulin resistance differentially affects the PI 3-kinase- and MAP kinase-mediated signaling in human muscle.

Montagnani M, Golovchenko I, Kim I, Koh GY, Goalstone ML, Mundhekar AN, Johansen M, Kucik DF, Quon MJ, Draznin B: Inhibition of phosphatidylinositol 3-kinase enhances mitogenic action of insulin in endothelial cells. Wang C, Gurevich I, Draznin B: Insulin affects vascular smooth muscle cell phenotype and migration via distinct signaling pathways.

Sartipy P, Loskutoff DJ: Monocyte chemoattractant protein 1 in obesity and insulin resistance. Kahn BB, Flier JS: Obesity and insulin resistance. Pessin JE, Saltiel AR: Signaling pathways in insulin action: molecular targets of insulin resistance. LeRoith D, Zick Y: Recent advances in our understanding of insulin action and insulin resistance.

Diabetes Care. Qiao L, Goldberg JL, Russell JC, Sun XJ: Identification of enhanced serine kinase activity in insulin resistance. White MF: Insulin signaling in health and disease.

Birnbaum MJ: Turning down insulin signaling. Um SH, Frogerio 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.

Patti M-E, Kahn BB: Nutrient sensor links obesity with diabetes risk. Nat Med. Aguirre V, Werner ED, Giraud J, Lee YH, Shoelson SE, White MF: Phosphorylation of Ser in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action.

Qiao L, Zhande R, Jetton TL, Zhou G, Sun XJ: In vivo phosphorylation of insulin receptor substrate 1 at serine by a novel serine kinase in insulin-resistant rodents.

Curr Biol. Raught B, Gingras AC, Sonenberg N: The target of rapamycin TOR proteins. Rohde J, Heitman J, Cardenas ME: The TOR kinases link nutrient sensing to cell growth.

Khamzina L, Veilleux A, Bergeron S, Marette A: Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: possible involvement in obesity-linked insulin resistance.

Trembley 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.

Burnett PE, Barrow RK, Cohen NA, Snyder SH, Sabatini DM: RAFT1 phosphorylation of the translational regulators p70S6 kinase and 4E-BP1.

Hara K, Yonezawa K, Kozlowski MT, Sugimoto T, Andrabi K, Weng OP, Kasuga M, Nishimoto I, Avruch J: Regulation of eIF-4E BP1 phosphorylation by mTOR. Isotani S, Hara K, Tokunaga C, Inoue H, Avruch J, Yonezawa K: Immunopurified mammalian target of rapamycin phosphorylates and activates p70S6 kinase α in vitro.

Nave BT, Ouwens M, Withers DJ, Alessi DR, Shepherd PR: Mammalian target of rapamycin is a direct target for protein kinase B: identification of a convergence point for opposing effects of insulin and amino-acid deficiency on protein translation. Scott PH, Brunn GJ, Kohn AD, Roth RA, Lawrence JC Jr: Evidence of insulin-stimulated phosphorylation and activation of the mammalian target of rapamycin mediated by a protein kinase B signaling pathway.

FASEB J. Pham P-TT, Heydrick SJ, Fox HL, Kimball SR, Jefferson LS Jr, Lynch CJ: Assessment of cell-signaling pathways in the regulation of mammalian target of rapamycin mTOR by amino acids in rat adipocytes.

J Cell Biochem. Pende M, Kozma SC, Jaquet M, Oorshcot V, Burcelin R, Le Marchand-Brustel Y, Klumperman J, Thorens B, Thomas G: Hypoinsulinaemia, glucose intolerance and diminished β-cell size in S6K1-deficient mice. Tremblay F, Krebs M, Dombrowski L, Brehm A, Bernroider E, Roth E, Nowotny P, Waldhausl W, Marette A, Roden M: Overactivation of S6 kinase 1 as a cause of human insulin resistance during increased amino acid availability.

Hirosumi J, Tuncman G, Chang L, Gorzun CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS: A central role for JNK in obesity and insulin resistance. Gao Z, Zhang X, Zuberi A, Hwang D, Quon MJ, Lefevre M, Ye J: Inhibition of insulin sensitivity by free fatty acids requires activation of multiple serine kinases in 3T3—L1 adipocytes.

Mol Endocrinol. Nguyen MTA, Satoh H, Favelyukis S, Babendure JL, Imamura T, Sbodio JI, Zalevsky J, Dahiyat B, Chi N-W, Olefsky JM: JNK and tumor necrosis factor-α mediate free fatty acid-induced insulin resistance in 3T3—L1 adipocytes.

Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE: Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of IKK-beta. Perseghin G, Petersen K, Shulman GI: Cellular mechanism of insulin resistance: potential links with inflammation.

Int J Obes Relat Metab Disord. Gao Z, Hwang D, Bataille F, Lefevre, York D, Quon MJ, Ye J: Serine phosphorylation of insulin receptor substrate 1 by inhibitor kB kinase complex. Kim JK, Kim Y-J, Fillmore JJ, Chen Y, Moore I, Lee J, Yuan M, Li ZW, Karin M, Perret P, Shoelson SE, Shulman GI: Prevention of fat-induced insulin resistance by salicylate.

Hundal RS, Petersen KF, Mayerson AB, Rahdhawa PS, Inzucchi S, Shoelson SE, Shulman GI: Mechanism by which high-dose aspirin improves glucose metabolism in type 2 diabetes.

Hotamisligil GS, Spiegelman BM: Tumor necrosis factor α: a key component of the obesity-diabetes link. Qi C, Pekala PH: Tumor necrosis factor-alpha-induced insulin resistance in adipocytes. Proc Soc Exp Biol Med. Hotamisligil GS, Shargill NS, Spiegelman BM: Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance.

Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS: Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM: IRSmediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha and obesity-induced insulin resistance.

Lowell BB, Shulman GI: Mitochondrial dysfunction and type 2 diabetes. 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. N Engl J Med. Li Y, Soos TJ, Li X, Wu J, Degennaro M, Sun X, Littman DR, Birnbaum MJ, Polakiewicz RD: Protein kinase θ inhibits insulin signaling by phosphorylating IRS1 at Ser Bell KS, Shcmitz-Peiffer C, Lim-Fraser M, Biden TJ, Cooney GJ, Kraegen EW: Acute reversal of lipid-induced muscle insulin resistance is associated with rapid alteration in PKC-θ localization.

Am J Physiol Endocrinol Metab. Itani SI, Pories WJ, Macdonald KG, Dohm GL: Increased protein kinase C θ in skeletal muscle of diabetic patients. Haruta T, Uno T, Kawahara J, Takano A, Egawa K, Sharma PM, Olefsky JM, Kobayashi M: A rapamycin-sensitive pathway down-regulates insulin signaling via phosphorylation and proteasomal degradation of insulin receptor substrate Harrington LS, Findlay GM, Gray A, Tolkacheva T, Wigfield S, Rebholz H, Barnett J, Leslie NR, Cheng S, Shepherd PR, Gout I, Downes CP, Lamb RE: The TSC1—2 tumor suppressor controls insulin-PI3K signaling via regulation of IRS proteins.

J Cell Biol. Lee YH, Giraud J, Davis RJ, White MF: C-Jun N-terminal kinase JNK mediates feedback inhibition of the insulin signaling cascade.

Ueki K, Fruman DA, Brachmann SM, Tseng YH, Cantley LC, Kahn CR: Molecular balance between the regulatory and catalytic subunits of phosphoinositide 3-kinase regulates cell signaling and survival. Shepherd PR, Withers DJ, Siddle K: Phosphoinositide 3-kinase: the key switch mechanism in insulin signaling.

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, Kadowaki T: Increased insulin sensitivity and hypoglycaemia in mice lacking the p85α subunit of phosphoinositide 3-kinase.

Nat Genet. Ueki K, Algenstaedt P, Mauvais-Jarvis F, Kahn CR: Positive and negative regulation of phosphoinositide 3-kinase-dependent signaling pathways by three different gene products of the p85α regulatory subunit.

Mauvais-Jarvis F, Ueki K, Fruman DA, Hirshman MF, Sakamoto K, Goodyear LJ, Iannacone M, Accili D, Cantley LC, Kahn CR: Reduced expression of the murine p85α subunit of phosphoinositide 3-kinase improves insulin signaling and ameliorates diabetes.

Ueki K, Fruman DA, Yballe CM, Fasshauer M, Klein J, Asano T, Cantley LC, Kahn CR: Positive and negative roles of p85α and p85β regulatory subunits of phosphoinositide 3-kinase in insulin signaling. Barbour LA, Shao J, Qiao L, Leitner W, Anderson M, Friedman JE, Draznin B: Human placental growth hormone increases expression of p85 regulatory unit of phosphatidylinositol 3-kinase and triggers severe insulin resistance in skeletal muscle.

Although core components of the pathway are well defined, less signaing known about mechanisms Insulin sensitivity and insulin signaling adjust the sensitivity of Organic post-workout recovery pathway Insulin sensitivity and insulin signaling extracellular Berry Infused Water. Snesitivity humans, disturbance in insulin Ways to boost immunity leads insukin impaired xnd of glucose from the blood stream, xensitivity is a hallmark of diabetes. Here we present the results of a genetic screen in Drosophila designed to identify regulators of insulin sensitivity in vivo. This mechanism permits physiological adjustment of insulin sensitivity and subsequent maintenance of circulating glucose at appropriate levels. Insulin signaling is an important and conserved physiological regulator of growth, metabolism, and longevity in multicellular animals. Disturbance in insulin signaling is common in human metabolic disorders. For example insulin resistance is a hallmark of diabetes and metabolic syndrome. Insuli is a signalign hormone Insulin sensitivity and insulin signaling predominantly functions to sensjtivity Insulin sensitivity and insulin signaling glucose levels. Sensitivitj is anr from beta cells found in the islets of the Mindful portion control for fewer cravings in response to nutrient uptake and increased insulim glucose levels. When insulin binds to Inulin receptors on sensiticity cells, such as skeletal muscle cells and Berry Infused Water, sensirivity signaling cascade is initiated, which culminates in the translocation of the glucose transporter GLUT4 from intracellular vesicles to the cell membrane. Once GLUT4 is incorporated into the plasma membrane, it functions to promote the uptake of extracellular glucose, which is then stored as glycogen in these cells, thereby regulating blood glucose [1]. Insulin also regulates blood sugar through inhibiting gluconeogenesis de novo glucose production and glycogenolysis glycogen breakdown in the liver. Besides regulating blood glucose levels, insulin also plays critical roles in facilitating protein and lipid synthesis and preventing the conversion of protein and fat to glucose. While insulin is widely viewed as a glucose homeostasis regulating hormone, an increasing body of research is illuminating broader roles for this peptide.

Author: Kagagore

3 thoughts on “Insulin sensitivity and insulin signaling

  1. Ich meine, dass Sie den Fehler zulassen. Es ich kann beweisen. Schreiben Sie mir in PM, wir werden umgehen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com