Open access

Molecular Basis of Insulin Resistance and Its Relation to Metabolic Syndrome

Written By

Sarika Arora

Submitted: 24 October 2012 Published: 12 December 2012

DOI: 10.5772/54620

From the Edited Volume

Insulin Resistance

Edited by Sarika Arora

Chapter metrics overview

2,836 Chapter Downloads

View Full Metrics

1. Introduction

The metabolic syndrome is an agglomeration of interrelated risk factors that is associated with nearly 5-fold increased risk for type 2 diabetes mellitus (DM) and a 2-fold increased risk of coronary artery disease (CAD) [1]. Reaven first suggested this cluster of metabolic abnormalities in 1988. It is characterized by insulin resistance, visceral adiposity, dyslipidemia and a systemic pro-inflammatory and pro-coagulant state [2]. Insulin resistance is defined as reduced insulin action in metabolic and vascular target tissues, hence higher than normal concentration of insulin is required to maintain normoglycemia. On a cellular level, it indicates an inadequate strength of insulin signaling from the insulin receptor downstream to the final substrates of insulin action involved in multiple metabolic and mitogenic aspects of cellular function [3].

The development of insulin resistance leads to many of the metabolic abnormalities associated with this syndrome. Patients with insulin resistance tend to have impaired fasting plasma glucose levels, which increase the prevalence of more atherogenic, small dense low-density lipoprotein (LDL) particles. The growing incidence of insulin resistance and metabolic syndrome (MS) is seriously threatening human health globally. Individuals with MS have a 30%–40% probability of developing diabetes and/or CVD within 20 years, depending on the number of components present [4].

In the United States (US), the prevalence of the MS in the adult population was estimated to be more than 25%. Similarly, the prevalence of MS in seven European countries was approximately 23%. It was estimated that 20%–25% of South Asians have developed MS and many more may be prone to it [5,6]. The main reason why MS is attracting scientific and commercial interest is that the factors defining the syndrome are all factors associated with increased morbidity and mortality in general and from CVD in particular [7].

Though, Insulin resistance has been recognized as a basis of CVD and diabetes type II, its etiology still remains elusive. Recent studies have contributed to a deeper understanding of the underlying molecular mechanisms of Insulin resistance. This review provides a detailed understanding of these basic pathophysiological mechanisms which may be critical for the development of novel therapeutic strategies to treat/ prevent metabolic syndrome.

Advertisement

2. Signalling through Insulin receptor and its downstream Pathways

Insulin action is initiated by an interaction of insulin with its cell surface receptor [8]. The insulin receptor (IR) is a heterotetramer consisting of two α subunits and two β subunits that are linked by disulphide bonds. Insulin binds to the extracellular α subunit of the insulin receptor and activates the tyrosine kinase in the β subunit {figure 1). Binding of insulin to IR effects a series of intramolecular transphosphorylation reactions, where one β subunit phosphorylates its adjacent partner on a specific tyrosine residue. Once the tyrosine kinase of insulin receptor is activated, it promotes autophosphorylation of the β subunit itself, where phosphorylation of three tyrosine residues (Tyr-1158, Tyr-1162, and Tyr-1163) is required for amplification of the kinase activity [9]. It then recruits different substrate adaptors such as the Insulin Receptor Substrate (IRS) family of proteins. Although IRs are present on the surface of virtually all cells, their expression in classical insulin target tissues, i.e. muscle, liver and fat, is extremely high [10]. Tyrosine phosphorylated IRS then displays binding sites for numerous signaling partners. Phosphorylated IRS proteins serve as multisite docking proteins for various effector molecules possessing src homology 2 (SH2) domains, including phosphatidylinositol 3-kinase (PI 3-kinase) regulatory subunits (p85, p55 p50, p85, and p55PIK), the tyrosine kinases Fyn and Csk, the tyrosine protein phosphatase SHP-2/Syp, as well as several smaller adapter molecules such as the growth factor receptor binding proteins Grb-2, Crk, and Nck [11]. Activation of these SH2 domain proteins initiates signaling cascades, leading to the activation of multiple downstream effectors that ultimately transmit the insulin signal to a branching series of intracellular pathways that regulate cell differentiation, growth, survival, and metabolism. Four members of the IRS family have been identified that are considerably similar in their general architecture [12-15]. IRS proteins share a similar structure characterized by the presence of an NH2-terminal pleckstrin homology (PH) domain adjacent to a phosphotyrosine-binding (PTB) domain followed by a variable-length COOH-terminal tail that contains a number of Tyr and Ser phosphorylation sites. The PH domain is critical for IR-IRS interactions. Plasma membrane phospholipids, cytoskeletal elements, and protein ligands mediate these interactions [16, 17]. In contrast, the PTB domain interacts directly with the juxtamembrane (JM) domain of the insulin and IGF-I receptors [18, 19], and hindrance of these interactions (by Ser/Thr phosphorylation) negatively affects insulin signaling [19]. A third domain, the kinase regulatory loop binding (KRLB) is found only in IRS-2 [20, 21]. This domain interacts with the phosphorylated regulatory loop of the IR, whereas the phosphorylation of two Tyr residues within the KRLB are crucial for this interaction [22].

PI3 kinase is a target of the IRS proteins (IRS-1 and IRS-2) which phosphorylates specific phosphoinositides to form phosphatidylinositol 4,5 bisphosphate (PIP2) to phosphatidylinositol 3,4,5 triphosphate; in turn, this activates ser/thr kinase, i.e. phosphoinositide-dependent kinase-1 (PDK1) [23, 24]. Known substrates of the PDKs are the protein kinase B (PKB) and also atypical forms of protein kinase C (PKC) [25].

Downstream from PI 3-kinase, the serine/threonine kinase Akt (also called PKB) triggers insulin effects on the liver. Phosphatidylinositol-dependent kinase (PDK) and PKB/Akt have a pleckstrin homology domain that enables these molecules to migrate toward the plasma membrane [26]. Activated Akt induces glycogen synthesis, through inhibition of GSK-3; protein synthesis via mTOR and downstream elements; and cell survival, through inhibition of several pro-apoptotic agents (Bad, Forkhead family transcription factors, GSK-3). Insulin stimulates glucose uptake in muscle and adipocytes via translocation of GLUT4 vesicles to the plasma membrane [27- 29]. This suggests that the impairment of insulin activity leading to insulin resistance is linked to insulin signalling defects.

Recently, an alternative PI 3-kinase independent mechanism to enhance GLUT4 translocation and glucose uptake was described. According to this model, binding of insulin to its receptor finally activates the small G-protein TC10 via the scaffolding protein CAP (Cbl-associated protein) resulting in GLUT4 translocation and enhanced glucose uptake [30]. Insulin signaling also has growth and mitogenic effects, which are mostly mediated by the Akt cascade as well as by activation of the Ras/MAPK pathway. A negative feedback signal emanating from Akt/PKB, PKCΖ, p70 S6K and the MAPK cascades results in serine phosphorylation and inactivation of IRS signaling [31, 32]. Insulin signalling molecules involved in metabolic and mitogenic action have been demonstrated to play a role in cellular insulin resistance. A few recent reports indicate that some PKC isoforms may have a regulatory effect on insulin signalling. The expression levels and activity of a few PKC isoforms are found to be associated with insulin resistance [33-35].

Recent data from PKB knockout animal models provide an insight into the role of PKB in normal glucose homeostasis. While disruption of PKB/Akt1 isoform in mice have not shown to cause any significant perturbations in metabolism, mice with a knock out of the PKB(Akt2) isoform show insulin resistance ending up with a phenotype closely resembling Type 2 diabetes in humans [36-37]. Subsequent studies [38- 40] in insulin-resistant animal models and humans have consistently demonstrated a reduced strength of insulin signaling via the IRS-1/PI 3-kinase pathway, resulting in diminished glucose uptake and utilization in insulin target tissues. Recent studies on inherited insulin post-receptor mutations in humans have detected a missense mutation in the kinase domain of PKB (Akt2) in a family of severely insulin resistant patients. The mutant PKB was unable to phosphorylate downstream targets and to mediate inhibition of phosphoenolpyruvate carboxykinase (PEPCK), a gluconeogenic key enzyme [41]. Another recent study, involving the stimulation of PI3K and Akt-1, -2, and -3 by insulin and epidermal growth factors (EGFs) in skeletal muscles from lean and obese insulin-resistant humans showed that Insulin activated all Akt isoforms in lean muscles, whereas only Akt-1 was activated in obese muscles. Insulin receptor substrate (IRS)-1 expression was reduced in obese muscles, and this was accompanied by decreased Akt-2 and -3 stimulation. In contrast, insulin- or EGF-stimulated phosphotyrosine-associated PI3K activity was not different between lean and obese muscles. These results showed that a defect in the ability of insulin to activate Akt-2 and -3 may explain the impaired insulin-stimulated glucose transport in insulin resistance [42].

This suggests that the impairment of insulin activity leading to insulin resistance is linked to insulin signalling defects. These insulin signalling pathways are shown in figure1.

Advertisement

3. Mechanisms related to Insulin resistance

Two separate, but likely, complementary mechanisms have recently emerged as a potential explanations for Insulin resistance. First, changes in IRS-1 either due to mutations or serine phosphorylation of IRS proteins can reduce their ability to attract PI 3-kinase, thereby minimizing its activation. A number of serine kinases that phosphorylate serine residues of IRS-1 and weaken insulin signal transduction have been identified. Additionally, mitochondrial dysfunction has been suggested to trigger activation of several serine kinases, leading to a serine phosphorylation of IRS-1. Second, a distinct mechanism involving increased expression of p85α has also been found to play an important role in the pathogenesis of insulin resistance. Conceivably, a combination of both increased expression of p85α and increased serine phosphorylation of IRS-1 is needed to induce clinically apparent insulin resistance.

Advertisement

4. Mutations of IRS as a cause of Insulin resistance

IRS-1 protein is a gene product of IRS-1 gene. In humans, rare mutations of the IRS-1 protein are associated with insulin resistance [43] and disruption of the IRS-1 gene in mice results in insulin resistance mainly of muscle and fat [44]. The genetic analysis of the IRS-1 gene has revealed several base-pair changes that result in amino acid substitutions [45-47]. The most common amino acid change is a glycine to arginine substitution at codon 972 (G972R), which has an overall frequency of ≈6% in the general population [48], with a carrier prevalence of 9% among Caucasians [49]. This mutation has been reported to significantly impair IRS-1 function in experimental models [50], and clinical studies have shown that this genetic variant is associated with reduced insulin sensitivity [51]. Expression of this variant in 32-D cells is associated with a significant (20-30%) impairment of insulin-stimulated PI3-kinase activity, as well as reduced binding of IRS-1 to the p85 regulatory subunit of PI3-kinase. Genotype/phenotype studies stratified according to body mass index (BMI) indicate that obese subjects who are heterozygous for the mutant allele have a 50% decrease in insulin sensitivity, compared with wild-type obese subjects. This suggests that there may be an interaction between the mutant allele and obesity, such that, in the presence of obesity, the mutant variant may aggravate the obesity-associated insulin resistance [49]. Moreover, earlier observations have indicated that the presence of a mutated IRS-1 gene is associated with dyslipidemia, further suggesting that this gene variant may have a significant effect on several risk factors for CAD [48, 50-52].

Interestingly, IRS-2 knockout mice not only show insulin resistance of muscle, fat and liver, but also manifest diabetes as a result of cell failure [53]. This phenotype with severe hyperglycemia as a consequence of peripheral insulin resistance and insufficient insulin secretion due to a significantly reduced β-cell mass reveals many similarities to type 2 diabetes in man and outlines the role of IRS proteins for the development of cellular insulin resistance. Homozygous knockout mice lacking a single allele of IRS-1 gene lack any significant phenotype, whereas homozygous disruption of the IRS-1 gene results in a mild form of insulin resistance [54]. IRS-1 homozygous null mice (IRS-1-/-) do not show a clear diabetic phenotypic expression, presumably because of pancreatic β cell compensation. IRS-2−/− mice, on the other hand, developed diabetes as a result of severe insulin resistance paired with β-cell failure [55, 56]. Even though β cell mass was reduced in IRS-2−/− mice, individual β cell showed normal or increased insulin secretion in response to glucose [55].

In regard to insulin signaling, experiments in immortalized neonatal hepatocytes show that the lack of IRS-2 is not compensated for by an elevation of IRS-1 protein content or an increase in tyrosine phosphorylation [57]. Previous experiments performed in peripheral tissues of IRS-1−/− mice by Yamauchi et al. [44] suggested that IRS-2 could be a major player in hepatic insulin action. However, to what extent reduced IRS-2 contributes to insulin resistance in the liver remains uncertain. In humans, a number of polymorphisms have been identified in the IRS-2 gene. Among those, the amino acid substitution Gly1057Asp has been found in various populations with a prevalence sufficiently high to modulate a population’s risk of type 2 diabetes. In Caucasians, Finns, and Chinese, however, this variant has not shown an associated with type 2 diabetes [58, 59]. Although the polymorphism was associated with decreased insulin sensitivity and impaired glucose tolerance in women with polycystic ovary syndrome [60], it showed no association with insulin sensitivity in other studies [59, 61, 62]. In contrast, another study in women with polycystic ovary syndrome found that homozygous carriers of the Gly1057 allele had higher 2-h plasma glucose concentrations during an oral glucose tolerance test (OGTT) [63]. Decreased serum insulin and C-peptide concentrations during an OGTT were reported in middle-aged glucose-tolerant Danish males carrying the Asp1057 allele [62]. However, using formal β-cell function tests, associations with insulin secretion were not reproduced in German, Finnish, and Swedish populations [59, 61, 62].

Advertisement

5. Serine phosphorylation of IRS as a cause of Insulin resistance

IRS-1 contains 21 putative tyrosine phosphorylation sites, several of which are located in amino acid sequence motifs that bind to SH-2 domain proteins, including the p85 regulatory subunit of PI 3-kinase, Grb-2, Nck, Crk, Fyn, Csk, phospholipase Cγ, and SHP-2 [64]. IRS-1 contains also > 30 potential serine/threonine phosphorylation sites in motifs recognized by various kinases such as casein kinase II, protein kinase C, protein kinase B/Akt, and mitogen-activated protein (MAP) kinases [12, 64].

Human IRS-2 contains 22 potential tyrosine phosphorylation sites, but only 13 are conserved in IRS-1. The amino acid sequence identity between IRS-1 and IRS-2 is 43%, with some domains such as the PH and PTB domains exhibiting higher degrees of identity (65 and 75%, respectively). The COOH-terminal domains of IRS-1 and IRS-2 are poorly conserved, displaying only 35% identity, which arises largely from similar tyrosine phosphorylation motifs surrounded by variable stretches of amino acid sequence. The middle of IRS-2 possesses a unique region comprising amino acids 591–786 that interacts specifically with the kinase regulatory loop binding (KRLB) domain of the insulin receptor β subunit [65]. Since this region is absent in IRS-1, this domain may contribute to the signaling specificity of IRS-2. In addition, IRS-1 and IRS-2 may regulate unique signaling pathways because of different tissue distribution, subcellular localization, kinetics of activation/deactivation, or specificity of interaction with downstream effectors [66-68]. For example, it has been shown that IRS-1 and IRS-2 differ in their subcellular localization since IRS-1 is twofold more concentrated in the intracellular membrane compartment than in cytosol, whereas IRS-2 is twofold more concentrated in cytosol than in the intracellular membrane compartment [69]. Further studies have shown that IRS-2 is dephosphorylated more rapidly and activates PI 3-kinase more transiently than IRS-1, thus indicating that differences in kinetics of activation may contribute to the diversity of the insulin signaling transduced by IRS-1 and IRS-2 [69,70].

Since, IRS-1 and IRS-2 have the longest tails, which contain ∼20 potential Tyr phosphorylation sites. Many of the Tyr residues gather into common Tyr-phosphorylated consensus motifs (YMXM or YXXM) that bind SH2 domains of their effector proteins. Spatial matching is required for successful protein-protein interaction. Ser/Thr phosphorylation of IRS proteins in close proximity to their PTB (receptor-binding) region impedes the binding of the SH2 domains of these effectors, thus inhibiting insulin signaling [71].

Serine phosphorylation of IRS proteins can occur in response to a number of intracellular serine kinases [72].The causes of IRS-1 serine phosphorylation are-

  1. mTOR- p70S6 kinase, Amino acids, Hyperinsulinemia

  2. JNK- Stress, Hyperlipidemia, Inflammation

  3. IKK- Inflammation

  4. TNFα- Obesity, Inflammation

  5. Mitochondrial dysfunction

  6. PKC θ- Hyperglycemia, Diacylglycerol, Inflammation

Recent studies have demonstrated hyper-serine phosphorylation of IRS-1 on Ser302, Ser307, Ser612, and Ser632 in several insulin-resistant rodent models [73- 76] as well as in lean insulin-resistant offspring of type 2 diabetic parents [77]. Further evidence for this hypothesis stems from recent studies in a muscle-specific triple serine to alanine mutant mouse (IRS-1 Ser → Ala302, Ser → Ala307, and Ser → Ala612), which has been shown to be protected from high-fat diet–induced insulin resistance in vivo [78]. Based on in vitro studies, serine phosphorylation may lead to dissociation between insulin receptor/IRS-1 and/or IRS-1/PI 3-kinase, preventing PI 3-kinase activation [79, 80] or increasing degradation of IRS-1 [81].

Ser318 of IRS-1 is a potential target for PKCζ [82], JNK, and kinases along the PI3K-mTOR pathway [83]. It is located in close proximity to the PTB domain. Therefore, its phosphorylation presumably disrupts the interaction between IR and IRS-1. Phosphorylation of Ser318 is not restricted to insulin stimulation. Elevated plasma levels of leptin, an adipokine produced by adipocytes [84], also stimulates the phosphorylation of Ser318. This down regulates insulin-stimulated Tyr phosphorylation of IRS-1 and glucose uptake.

In a recent study using skeletal muscle biopsies from 11 humans, the mTOR-S6K pathway was shown to negatively modulate glucose metabolism under nutrient abundance [151]. In agreement with previous studies, phosphorylation of Ser312 and Ser636 of IRS-1 was implicated as part of this negative regulation [85, 86]. Increased phosphorylation of Ser636 of IRS-1 was observed in myotubes of patients with type 2 diabetes. Inhibition of ERK1/2 with PD-98059 reduced this phosphorylation, thereby implicating ERK1/2 in the phosphorylation of Ser636 in human muscle [87].

To unveil the importance of phosphorylated Ser/Thr residues of human IRS-1, Yi et al. [88] adopted a mass spectrometry approach. More than 20 Ser residues of IRS-1 were found to undergo insulin-stimulated phosphorylation in human muscle biopsies, three of which were newly identified sites: Thr495, Ser527, and Ser1005. This report validates previous in vitro and in vivo studies in animal models and suggests that the same strategy could be employed to identify phosphorylated Ser/Thr sites under conditions of insulin resistance, obesity, or type 2 diabetes.

Impaired hepatic glycogen storage and glycogen synthase activity is a common finding in insulin resistance [89] and polymorphisms in the glycogen synthase gene have been described in insulin resistant patients. The most frequent mutations are the so-called XbaI mutations and Met416Val within intron 14 and exon 10, respectively. Currently, there are conflicting data on the correlation of these polymorphisms with insulin resistance and Type 2diabetes mellitus [90-92].

Recently, a hypothesis that mitochondrial dysfunction or reduced mitochondrial content accompanied by a decreased mitochondrial fatty acid oxidation and accumulation of fatty acid acyl CoA and diacylglycerol can cause insulin resistance has gained substantial experimental support [93- 95]. The mechanism of insulin resistance in these cases has been suggested to involve activation of a novel PKC that either by itself or via IKKβ or JNK-1 could lead to increased serine phosphorylation of IRS-1. Severe mitochondrial dysfunction can result in diabetes that is typically associated with severe β-cell dysfunction and neurological abnormalities [96]. In a study,using 13C/31P MRS, it was found that in the healthy lean elderly volunteers with severe muscle insulin resistance, there is∼40% reduction in rates of oxidative phosphorylation activity associated with increased intramyocellular and intrahepatic lipid content [94]. This study suggests that an acquired loss of mitochondrial function associated with aging predisposes elderly subjects to intramyocellular lipid accumulation, which results in insulin resistance [78]. Further, it was found that mitochondrial density was reduced by 38% in the insulin-resistant offspring [77].

[This topic has been dealt in details in subsequent chapter by Wang etal.]

The proinflammatory novel PKCθ has been found to cause serine phosphorylation of IRS-1 [97, 98], while PKCθ knockout mice have been shown to be protected from fat-induced insulin resistance [75]. Increased activity of PKCθ, along with increased activity of JNK, has also been found in skeletal muscle of obese and type 2 diabetic subjects [99, 100], supporting a potential role of these serine kinases in the pathogenesis of insulin resistance.

Figure 1.

Insulin signaling pathway showing that the binding of insulin with Insulin receptor (IR) leads to phosphorylation of tyrosine residues followed by activation of downstream signalling pathways which result in recruitment in recruitment of GLUT-4 transporter to the plasma membrane and entry of glucose molecules within the cell. Serine phosphorylation of IRS protein has an inhibitory effect on downstream pathways resulting in insulin Resistance.

Advertisement

6. Increased expression of p85

A molecular mechanism that can potentially lead to insulin resistance is a disruption in the balance between the amounts of the PI 3-kinase subunits [101]. PI 3-kinase belongs to the class 1a 3-kinases [102], which exist as heterodimers, consisting of a regulatory subunit p85, which is tightly associated with a catalytic subunit, p110. Most tissues express two forms of regulatory subunit, p85α and p85β, and two forms of catalytic subunit, p110α and p110β [102]. p85α and p85β share the highest degree of homology in the C-terminal half of the molecules, which contains two SH2 domains that bind to tyrosine-phosphorylated proteins and an inter-SH2 domain that interacts with the catalytic subunit. The N-terminal halves of p85α and p85β contain an SH3 domain, a BCR homology region, and two proline-rich domains, but these domains are less well conserved between the two molecules. Two isoforms of p85α truncated in the N-terminal region, identified as AS53 (or p55α) [103, 104] and p50α [105, 106], as well as p85α itself, are derived from a single gene (Pik3r1). p85β and another short isoform with limited tissue distribution termed p55γ/p55PIK are encoded by separate genes [107]. Normally, the regulatory subunit exists in stoichiometric excess to the catalytic one, resulting in a pool of freep85 monomers not associated with the p110 catalytic subunit. However, there exists a balance between the free p85 monomer and the p85-p110 heterodimer, with the latter being responsible for the PI 3-kinase activity [108-110]. Because the p85 monomer and the p85-p110 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 [111]. Increase or decrease in expression of p 85 would result in a shift in the balance either in the favour of free p85 or p85-p110 complexes [108-110].

One of the first indications that an imbalance between the abundance of p85 and p110 can alter PI 3-kinase activity came from experiments with l-6 cultured skeletal muscle cells treated with dexamethazone [111]. 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 p110. The authors concluded that p85α competes with the p85-p110 heterodimer, thus, reducing PI 3-kinase activity.

Subsequently, animals with a targeted disruption of p85α (p85+/− heterozygous mice) have been found to have a higher ratio of p85-p110 dimer to free p85 and to be more sensitive to insulin [101, 111-114].

The possibility of mismatch between free p85 and p85-p110 complexes has been recently supported by studies in insulin-resistant states induced by human placental growth hormone [115], obesity, and type 2 diabetes [100] and by short-term overfeeding of lean non-diabetic women [116]. Barbour etal [117] have 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 [118].

Another small study of eight healthy lean women without a family history of diabetes, by Cornier et al showed that 3 days of overfeeding (50% above usual caloric intake) led to a significant increase in expression of p85α, ratio of p85α to p110, and a decline in insulin sensitivity. Within this experimental time frame, overfeeding did not cause any change in serine phosphorylation of either IRS-1 or S6K1, suggesting that increased expression of p85α may be an early molecular step in the pathogenesis of the nutritionally induced insulin resistance [116].

Advertisement

7. Role of the adipose tissue in insulin resistance

Insulin has 3 major target tissues—skeletal muscle, liver and adipose tissue. It has been postulated that the insulin receptor (IR) is overexpressed in the cells of these tissues. Also only these three organs in the body are capable of glucose deposition and storage; no other cells can store glucose. Removal of excess postprandial glucose by insulin occurs due to glucose uptake and storage in insulin sensitive target cells. About 75% of insulin-dependent postprandial glucose disposal occurs into the skeletal muscle [119]; therefore, it is the major target cell. While insulin-stimulated glucose disposal in adipose tissue is of little quantitative importance compared with that in muscle, regulation of lipolysis with subsequent release of glycerol and FFA into the circulation by insulin has major implications for glucose homeostasis.

It is widely accepted that increased availability and utilization of FFA contribute to the development of skeletal muscle insulin resistance [120-122]. Moreover, FFA have been shown to increase endogenous glucose production both by stimulating key enzymes and by providing energy for gluconeogenesis [123]. Finally, the glycerol released during triglyceride hydrolysis serves as a gluconeogenic substrate [124]. Consequently, resistance to the antilipolytic action of insulin in adipose tissue resulting in excessive release of FFA and glycerol would have deleterious effects on glucose homeostasis.

Patients suffering from insulin resistance and type 2 diabetes frequently display signs of abnormal lipid metabolism, increased circulatory concentration and elevated deposition of lipids in the skeletal muscle [125]. Increase in plasma FFA reduces insulin-stimulated glucose uptake, whereas a decrease in lipid content improves insulin activity in the skeletal muscle cells, adipocytes and liver [126]. Lipid-associated insulin resistance has also been shown to be linked to Glut4 translocation defects [27]. Studies have shown that raising plasma fatty acids in both rodents [75] and humans [127] abolishes insulin activation of IRS-1–associated PI 3-kinase activity in skeletal muscle where IRS-1 is most prevalent.

Adipose tissue can modulate whole body glucose metabolism by regulating levels of circulating free fatty acids (FFA) and also by secreting adipokines, thereby acting as an endocrine organ. However, the underlying mechanism of FFA-induced impairment of insulin signals is still unclear. The molecular mechanism underlying defective insulin-stimulated glucose transport activity can be attributed to increases in intramyocellular lipid metabolites such as fatty acyl CoAs and diacylglycerol, which in turn activate a serine/threonine kinase cascade, thus leading to defects in insulin signaling through Ser/Thr phosphorylation of insulin receptor substrate-1 [78].

Some of the PKC isoforms represent such signalling molecules. PKC isoforms are classified as classical (cPKCα, βI, βII, γ), novel (nPKCδ, ε, θ, η) and atypical (aPKCζ, λ). cPKCs are activated by Ca+2 and diacylglycerol (DAG), nPKCs are activated by only DAG and aPKCs respond to neither Ca+2 nor DAG [128]. Among all these PKC isoforms, nPKCs are said to have a modulatory role in insulin signalling. Recent reports also demonstrate a link between nPKCs and FFA induced insulin resistance.

Diacylglycerol is an attractive trigger for fat-induced insulin resistance in skeletal muscle, since it has been shown to increase in muscle during both lipid infusions and fat feeding and it is a known activator of novel protein kinase C (PKC) isoforms [78].

Recent studies have revealed that accumulation of intracellular lipid metabolites activate a serine kinase cascade involving PKC-ε, leading to decreased insulin receptor kinase activity resulting in 1) lower insulin-stimulated IRS-2 tyrosine phosphorylation, 2) lower IRS-2–associated PI 3-kinase activity, and 3) lower AKT2 activity [129]. These fat-induced defects in insulin signalling in turn result in reduced insulin stimulation of glycogen synthase activity, resulting in decreased insulin-stimulated hepatic glucose uptake and reduced insulin stimulation of hepatic glucose production. Furthermore, reduced activity of AKT2 results in decreased phosphorylation of forkhead box protein O (FOXO), allowing it to enter the nucleus and activate the transcription of the rate-controlling enzymes of gluconeogenesis (phosphoenolpyruvate carboxykinase, glucose-6-phosphate phosphatase).

Increased gluconeogenesis further exacerbates hepatic insulin resistance and results in fasting hyperglycemia [129- 131]. Mitochondrial glycerol-3-phosphate acyltransferase (mtGPAT) is a key enzyme in de novo fat synthesis in liver, and recent studies in mtGPAT knockout mice have clearly implicated intracellular accumulation of diacylglycerol in triggering fat-induced insulin resistance in liver through activation of PKC-ε [132]. These data have important implications for the development of novel therapeutic agents to reverse and prevent hepatic insulin resistance associated with non-alcoholic fatty liver and type 2 diabetes [133].

Lipid infusion in rats and humans impaired insulin-stimulated glucose disposal into the muscle and concomitant activation of PKCθ and PKCδ [134, 135]. PKCδ has been shown to be a possible candidate for phosphorylation of the IR on serine residues [136]. These result in defects in the insulin signalling pathway imposing insulin resistance.

Recently, the PPARγ co-activator-1 (PGC-1) has been recognized as playing a major role in glucose homeostasis of the organism. Work mainly by Spiegelman’s group demonstrated a crucial role of PGC-1 in the regulation of GLUT4 in muscle cells [137]. (PGC)-1α and PGC-1 β are transcriptional factor co-activators that regulate mitochondrial biogenesis. In addition AMP kinase, which is activated during exercise and ischemia by a reduction in the ATP/AMP ratio, has been shown to be an important regulator of mitochondrial biogenesis, mediating its effects through MEF2- and CREB-mediated increased PGC-1α expression [138-141]. Extracellular stimuli such as cold, thyroid hormone, and exercise stimulate mitochondrial biogenesis through PGC-1 in brown fat and skeletal muscle. Increased PGC-1 protein expression leads to increases in the target genes, including nuclear respiratory factor (NRF)-1. NRF-1 is a transcription factor stimulating many nuclear-encoded mitochondrial genes such as OXPHOS genes and also mitochondrial transcription factor A (mtTFA), a key transcriptional factor for the mitochondrial genome. mtTFA can bind to the D-loop of the mitochondrial genome and increase transcription of mitochondrial genes and replication of mitochondrial DNA [142].

A recent study by Ling et al. [143] demonstrated an age dependent decrease in muscle gene expression of PGC-1 α and PGC-1 β in young and elderly dizygotic and monozygotic twins without known diabetes

Adipose tissue also acts as an endocrine organ producing adipokines which modulate glucose homeostasis [144]. Currently, those most intensely discussed are tumor necrosis factor-α (TNF α), leptin, adiponectin and resistin. At a molecular level, TNF α increases serine phosphorylation of IRS-1 and down-regulates GLUT4 expression, thereby contributing to insulin resistance [38]. Furthermore, mice lacking TNF α function were protected from obesity-induced insulin resistance [145]. The role of leptin in regulating food intake and energy expenditure is well established. Humans with leptin deficiency or leptin receptor mutations are severely obese [146,147]. The adiponectin has insulin-sensitizing effects as it enhances inhibition of hepatic glucose output as well as glucose uptake and utilization in fat and muscle. The expression of adiponectin is decreased in obese humans and mice [148]. Thus, in humans, adiponectin levels correlate with insulin sensitivity. Because of its insulin-antagonistic effects, the adipocytokine resistin has attracted a lot of research interest. This is mainly based on data obtained in-vitro and from some animal models. Resistin decreases insulin-dependent glucose transport in-vitro and increases fasting blood glucose concentrations and hepatic glucose production in-vivo [149, 150].

Advertisement

8. Insulin resistance and Forkhead box protein O (FOXO)

The fasting hyperglycaemia in patients with Type 2 diabetes is the clinical correlate of the increased glucose production by the liver because of insulin resistance. This is as a result of the lack of inhibition of the two key gluconeogenic enzymes, phospho-enolpyruvate carboxykinase (PEPCK) and the glucose-6- phosphatase (G6Pase) catalytic subunit. Studies in hepatoma cells [151,152] suggest that Foxo1 and -3 regulate the transcription of reporter genes containing insulin response elements from the PEPCK and G6Pase promoters. Furthermore, Foxo1 is phosphorylated in an insulin-responsive manner by PIP3-dependent kinases, such as Akt. Reduced activity of AKT2 results in decreased phosphorylation of Foxo protein, allowing it to enter the nucleus and activate the transcription of these rate-controlling enzymes of gluconeogenesis [151,153]. There is increasing evidence that Foxo-proteins are critically involved in the insulin dependent regulation of gluconeogenic gene expression and insulin-resistancein-vivo [154, 155]. In addition, the PPARγco-activator-1 (PGC-1), a factor integrating the effects of glucocorticoids and cAMP on gluconeogenic geneexpression in the liver [156, 157] is also regulated by PKB and Foxo1 [158].

Advertisement

9. FFA induced Inhibition of Insulin receptor (IR) gene expression by PKCε

Clearly, the IR is one of the major targets in FFA-induced impairment of insulin activity. Recent studies performed in-vivo suggested that glucose uptake rather than intracellular glucose metabolism is the rate-limiting step for fatty acid induced insulin resistance in humans [159]. This indicates a mechanism in which accumulation of intracellular fatty acids or their metabolites results in an impairment of signaling through IRS/PI 3-kinase.

Recent evidence has shown that PDK1 can directly phosphorylate all PKCs including nPKCs [160]. The PKCε isotype has recently been shown to be related to insulin resistance. Insulin stimulation of PDK1 phosphorylation is inhibited by an FFA, i.e. palmitate. PKCε phosphorylation is dependent on PDK1; FFA incubation of skeletal muscle cells and adipocytes inhibited PDK1 phosphorylation but surprisingly increased PKCε phosphorylation. Inhibition of PDK1 by FFA is reflected in Akt phosphorylation as Akt phosphorylation is also dependent on PDK1 [161]. It has been shown that myristic acid incubation of HEPG2 cells causes myristoylation of PKCε which results in constitutive phosphorylation of PKCεat thr566/ser729 in the kinase domain required for PKCε activity. This phosphorylation was totally independent of PDK1, which the workers demonstrated by using PDK1 knockout cells. In the same way, addition of palmitate to skeletal muscle cells or adipocytes may affect palmitoylation of PKCε resulting in constitutive phosphorylation of PKCε [162, 163]. Taken together, it is clear that FFA causes PDK1-independent phosphorylation of PKCε which in turn translocates to the nucleus, and its time of entry into the nucleus coincides with inhibition of IR gene transcription.

Advertisement

10. Conclusion

In this review, current developments contributing to understanding of insulin resistance and to the pathogenesis of metabolic syndrome has been discussed. Among the many molecules involved in the intracellular processing of the signal provided by insulin, IRS-2, PKB, Foxo protein and p85 regulatory subunit of PI-3 kinase have attracted particular interest, because their dysfunction results in insulin resistance in-vivo. It has been well established that FFA are responsible for insulin resistance. This review focuses on the current trends in research in this important domain and throws light on certain possibilities regarding the manner in which FFA inhibits insulin activity.

References

  1. 1. RosensonR. SNew approaches in the intensive management of cardiovascular risk in the metabolic syndrome.Curr Probl Cardiol 200520313317
  2. 2. ReavenG. MBanting lecture 1988role of insulin resistance in human disease. Diabetes 1988; 3715961607
  3. 3. GinsbergHInsulin resistance and cardiovascular diseaseJ Clin Invest 2000106453458
  4. 4. EnasE. AMohanVDeepaMFarooqSPazhoorSChennikkaraHThe metabolic syndrome and dyslipidemia among Asian Indians : a population with high rates of diabetes and premature coronary artery disease. J Cardiometab Syndr 20072267275
  5. 5. NestelPLyuRLowL. PSheuW. HNitiyanantWSaitoIetalMetabolic syndrome: Recent prevalence in East and Southeast Asian populations.Asia Pac J Clin Nutr 200716362367
  6. 6. EapenDKalraG. LMerchantNAroraAKhanB. VMetabolic syndrome and cardiovascular disease in South AsiansVasc Health Risk Manag 20095731743
  7. 7. Borch-johnsenKThe metabolic syndrome in a global perspective. The public health impact. Dan Med Bull 200754157159
  8. 8. ShulmanG. ICellular mechanisms of insulin resistance in humans.Am J Cardiol 19993J EOF10J EOF
  9. 9. WhiteM. FShoelsonS. EKeutmannHKahnC. RA cascade of tyrosine autophosphorylation in the beta-subunit activates the phosphotransferase of the insulin receptor.J Biol Chem198826329692980
  10. 10. BrunettiAManfiolettiGChiefariEGoldfineI. DFotiDTranscriptional regulation of human insulin receptor gene by the high-mobility group protein HMGI(Y). FASEB J.200115492500
  11. 11. VirkamakiAUekiKKahnC. RProtein-protein interaction in insulin signaling and the molecular mechanisms of insulin resistance.J Clin. Invest. 1999103931943
  12. 12. SunX. JRothenbergPKahnC. RBackerJ. MArakiEWildenP. AetalStructure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein.NatureLondon) 19913527377
  13. 13. SunX. JWangL. MZhangYYenushLMyersM. GGlasheenEetalRole of IRS-2 in insulin and cytokine signalling.NatureLondon) 1995377173177
  14. 14. LavanB. ELaneW. SLienhardG. EThe 60-kDa phosphotyrosine protein in insulin-treated adipocytes is a new member of the insulin receptor substrate family.J Biol Chem 19972721143911443
  15. 15. LavanB. EFantinV. RChangE. TLaneW. SKellerS. RLienhardG. EA novel 160-kDa phosphotyrosine protein in insulin-treated embryonic kidney cells is a new member of the insulin receptor substrate familyJ. Biol. Chem 19972722140321407
  16. 16. Farhang-fallahJRandhawaV. KNimnualAKlipABar-sagiDRozakis-adcockMThe pleckstrin homology (PH) domain-interacting protein couples the insulin receptor substrate 1 PH domain to insulin signaling pathways leading to mitogenesis and GLUT4 translocationMol Cell Biol 20022273257336
  17. 17. GreeneM. WSakaueHWangLAlessiD. RRothR. AModulation of insulin-stimulated degradation of human insulin receptor substrate-1 by Serine 312 phosphorylation.J Biol Chem 200327881998211
  18. 18. PazKVoliovitchHHadariY. RRoberts CT Jr, LeRoith D, Zick Y. Interaction between the insulin receptor and its downstream effectors. Use of individually expressed receptor domains for structure/function analysis.J Biol Chem 199627169987003
  19. 19. VoliovitchHSchindlerD. GHadariY. RTaylorS. IAcciliDZickYTyrosine phosphorylation of insulin receptor substrate-1 in vivo depends upon the presence of its pleckstrin homology region.J Biol Chem 19952701808318087
  20. 20. HeWCraparoAZhuYONeillT. JWangL. MPierceJ. HGustafsonTA. Interaction of insulin receptor substrate-2 (IRS-2) with the insulin and insulin-like growth factor I receptors. Evidence for two distinct phosphotyrosine-dependent interaction domains within IRS-2.J Biol Chem 19962711164111645
  21. 21. Sawka-verhelleDTartare-deckertSWhiteM. FVan ObberghenEInsulin receptor substrate-2 binds to the insulin receptor through its phosphotyrosine-binding domain and through a newly identified domain comprising amino acids 591-786.J Biol Chem 199627159805983
  22. 22. Sawka-verhelleDBaronVMotheIFillouxCWhiteM. FVan ObberghenETyr624 and Tyr628 in insulin receptor substrate-2 mediate its association with the insulin receptor.J Biol Chem 19972721641416420
  23. 23. AlessiD. RCohenPMechanism of activation and function of protein kinase B.Curr Opin Genet Dev 199885562
  24. 24. Le Good J AZiegler WH, Parekh D B, Alessi D R, Cohen P, Parker P J. Protein kinase C isotypes controlled by phosphoinositide 3-kinase through the protein kinase PDK1. Science 199828120422045
  25. 25. KotaniKOgawaWMatsumotoMKitamuraTSakaueHHinoYet alRequirement of atypical protein kinase clamdafor insulin stimulation of glucose uptake but not for Akt activation in 3T3-L1 adipocytes. Mol Cell Biol 19981869716982
  26. 26. TaniguchiC. MEmanuelliBKahnC. RCritical nodes in signalling pathways: insights into insulin action.Nat Rev Mol Cell Biol 200678596
  27. 27. PessinJ EThurmondD CElmendorfJ SCokerK Jand OkadaSMolecular basis of insulin-stimulated GLUT4 vesicle trafficking. J. Biol. Chem 199927425932596
  28. 28. KupriyanovaT. AKandrorK. VAkt-2 binds to Glut4-containing vesicles and phosphorylates their component proteins in response to insulin.J. Biol. Chem.199927414581464
  29. 29. MartinSMillarC. ALyttleC. TMeerlooTMarshB JGouldG. WetalEffects of insulin on intracellular GLUT4 vesicles in adipocytes: evidence for a secretory mode of regulation.J Cell Sci 200011334273438
  30. 30. KhanA. HPessinJ. EInsulin regulation of glucose uptake: a complex interplay of intracellular signalling pathways.Diabetologia20024514751483
  31. 31. CheathamBPhosphatidylinositol 3-kinase activation is required for insulin stimulation of 70S6kinase, DNA synthesis, and glucose transporter translocation. Mol Cell Biol 1994
  32. 32. ShepherdP. RNaveB. TSiddleKInsulin 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 70ribosomal protein-S6 kinase.Biochem J 1995
  33. 33. GreeneM. WMorriceNGarofaloR. SRothR. AModulation of human receptor substrate 1 tyrosine phosphorylation by protein kinase C δ. Biochem J 2004378105116
  34. 34. ZickYInsulin resistance: a phosphorylation-based uncoupling of insulin signallingTrends Cell Biol 200111437441
  35. 35. WhiteM. FIRS proteins and the common path to diabetes.Am J Physiol Endocrinol Metab 2002E413422
  36. 36. ChoHThorvaldsenJ. LChuQFengFBirnbaumM. JAkt1/PKBα is required for normal growth but dispensable for maintenance of glucose homeostasis in mice. J Biol Chem 20012763834938352
  37. 37. ChoHMuJKimJ. KThorvaldsenJ. LChuQCrenshawE. Brd et alInsulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKBα). Science 200129217281731
  38. 38. KahnB. BFlierJ. SObesity and insulin resistanceJ Clin Invest 2000106473481
  39. 39. PessinJ. ESaltielA. RSignaling pathways in insulin action: molecular targets of insulin resistanceJ Clin Invest 2000106165169
  40. 40. LeRoith DZick Y. Recent advances in our understanding of insulin action and insulin resistance. Diabetes Care 200124588597
  41. 41. GeorgeSRochfordJWolfrumCGrayS. LSchinnerSWilsonJ. Cet alHuman insulin resistance and diabetes mellitus due to a missense mutation AKT2. Science 200430413251328
  42. 42. Brozinick JT JrRoberts 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. Diabetes 200352935941
  43. 43. WhiteheadJ. PHumphreysPKrookAJacksonRHaywardALewisHet alMolecular scanning of the insulin receptor substrate 1 gene in subjects with severe insulin resistance: detection and functional analysis of a naturally occurring mutation in a YMXM motif. Diabetes 199847837839
  44. 44. YamauchiTTobeKTamemotoHUekiKKaburagiYYamamoto-hondaRet alInsulin signalling and insulin actions in the muscles and livers of insulin-resistant, insulin receptor substrate 1-deficient mice. Mol Cell Biol 19961630743084
  45. 45. AlmindKBjorbaekCVestergaardHHansenTEchwaldSPedersenOAmino acid polymorphism of insulin receptor substrate-1 in non-insulin-dependent diabetes mellitus. Lancet. 1993342828832
  46. 46. LaaksoMMalkkiMKekalainenPKuusistoJDeebS. SInsulin receptor substrate-1 variants in non-insulin-dependent diabetes.J Clin Invest. 19949411411146
  47. 47. ImaiYFuscoASuzukiYLesniakM. ADAlfonsoRSestiG, etal. Variant sequences of insulin receptor substrate-1 in non-insulin-dependent-diabetes mellitus. J Clin Endocrinol Metab. 19947916551658
  48. 48. HitmanG. AHawramiKMccarthyM. IViswanathanMSnehalathaCRamachandranAetalInsulin receptor substrate-1 gene mutations in NIDDM: implications for the study of polygenic disease.Diabetologia199538481486
  49. 49. PedersonOGenetics of Insulin resistance. Exp Clin Endocrinol Diabet 199910721138
  50. 50. AlmindKInoueGPedersenOKahnC. RA common amino acid polymorphism in insulin receptor substrate-1 causes impaired insulin signaling: evidence from transfection studies.J Clin Invest 19969725692575
  51. 51. ClausenJ. OHansenTBjorbaekCEchwaldS. MUrhammerS. ARasmussenSetalInsulin resistance: interactions between obesity and a common variant of insulin receptor substrate-1.Lancet1995346397402
  52. 52. BaroniM. GDAndreaM. PMontaliAPannitteriGBarillaFCampagnaF, etal. A common mutation of the insulin receptor substrate-1 gene is a risk factor for coronary artery disease.Arterioscler Thromb Vasc 19991912297580
  53. 53. PrevisS. FWithersD. JRenJ. MWhiteM. FShulmanG. IContrasting effects of IRS-1 versus IRS-2 gene disruption on carbohydrate and lipid metabolism in vivo.J Biol Chem 20002753899038994
  54. 54. ArakiEShimadaFUzawaHMoriMand EbinaYCharacterization of the promoter region of the human insulin receptor gene. J Biol Chem 19942621618616191
  55. 55. KubotaNTobeKTerauchiYEtoKYamauchiTSuzukiRetalDisruption of insulin receptor substrate 2 causes type 2 diabetes because of liver insulin resistance and lack of compensatory β-cell hyperplasia. Diabetes 20004918801889
  56. 56. KidoYBurksD. JWithersDBrunningJ. CKahnC. RWhiteM. FetalTissue-specific insulin resistance in mice with combined mutations of the insulin receptor. J Clin Invest 2000105199205
  57. 57. ValverdeA. MBurksD. JFabregatIFisherT. LCarreteroJWhiteM. FetalMolecular mechanisms of insulin resistance in IRS-2-deficient hepatocytes.Diabetes20035222392248
  58. 58. BernalDAlmindKYenushLAyoubMZhangYRosshaniLetalInsulin receptor substrate-2 amino acid polymorphisms are not associated with random type 2 diabetes among Caucasians.Diabetes199847976979
  59. 59. WangHRissanenJMiettinenRKarkkainenPKekalainenPKuusistoJetalNew amino acid substitutions in the IRS-2 gene in Finnish and Chinese subjects with late-onset type 2 diabetes.Diabetes20015019491951
  60. 60. El MkademS. ALautierCMacariFMolinariNLefebvrePRenardEetalRole of allelic variants Gly972Arg of IRS-1 and Gly1057Asp of IRS-2 in moderate-to-severe insulin resistance of women with polycystic ovary syndrome.Diabetes20015021642168
  61. 61. FritscheAMadausARennWTschritterOTeigelerAWeisserMetalThe prevalent Gly1057Asp polymorphism in the insulin receptor substrate-2 gene is not associated with impaired insulin secretion.J Clin Endocrinol Metab 20018648224825
  62. 62. AlmindKFrederiksenS. KBernalDHansenTAmbyeLUrhammerSetalSearch for variants of the gene-promoter and the potential phosphotyrosine encoding sequence of the insulin receptor substrate-2 gene: evaluation of their relation with alterations in insulin secretion and insulin sensitivity.Diabetologia19994212441249
  63. 63. EhrmannD. ATangXYoshiuchiICoxN. JBellG. IRelationship of insulin receptor substrate-1 and-2 genotypes to phenotypic features of polycystic ovary syndrome. J Clin Endocrinol Metab 20028742974300
  64. 64. WhiteM. FThe insulin signaling system and the IRS proteins.Diabetologia1997S2S17.
  65. 65. Sawka-verhelleDTartare-deckertSWhiteM. FVan ObberghenEInsulin receptor substrate-2 binds to the insulin receptor through its phosphotyrosine-binding domain and through a newly identified domain comprising amino acid 591-786.J Biol Chem 199627159805983
  66. 66. GiovannoneBScaldaferriM. LFedericiMPorzioOLauroDFuscoAetalInsulin receptor substrate (IRS) transduction system: distinct and overlapping signaling potential.Diabetes Metab. Res. Rev 200016434441
  67. 67. SunX. JPonsSWangL. MZhangYYenushLBurksDetalThe IRS-2 gene on murine chromosome 8 encodes a unique signaling adapter for insulin and cytokine action.Mol. Endocrinol 199711251262
  68. 68. ShuppinG. TPonsSHuglSAielloL. PKingG. LWhiteM. FetalA specific increased expression of Insulin Receptor Substrate 2 in pancreatic β-cell lines is involved in mediating serum-stimulated-cell growth. Diabetes 19984710741085
  69. 69. InoueGCheathamBEmkeyRKahnC. RDynamics of insulin signaling in 3T3-L1 adipocytes. Differential compartmentalization and trafficking of insulin receptor substrate (IRS)-1 and IRS-2.J Biol Chem. 19982731154811555
  70. 70. OgiharaTShinB. CAnaiMKatagiriHInukaiKFunakiMetalInsulin receptor substrate (IRS)-2 is dephosphorylated more rapidly than IRS-1 via its association with phosphatidylinositol 3-kinase in skeletal muscle cells. J Biol Chem 19972721286812873
  71. 71. Boura-halfonSZickYPhosphorylation of IRS proteins, insulin action, and insulin resistance. AJP- Endo 2009E581E591.
  72. 72. DrazninBMolecular Mechanisms of Insulin Resistance: Serine phosphorylation of insulin receptor substrate-1 and increased expression of 85The two sides of a coin. Diabetes 2006
  73. 73. UmS. HFrogerioFWatanabeMPicardFJoaquinMStickerMetalAbsence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity.Nature2004431200205
  74. 74. YuC. LChenYClineG. WZhangDZongHWangYetalMechanisms by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. J. Biol Chem 2002275023050236
  75. 75. KimJ. KFillmoreJ. JSunshineM. JAlbrechtBHigashimoriTKimD. WetalPKC-theta knockout mice are protected from fat-induced insulin resistance.J Clin Invest 2004114823827
  76. 76. FurukawaNOngusahaPJahngW. JArakiKChoiC. SKimH. JetalRole of Rho-kinase in regulation of insulin action and glucose homeostasis.Cell Metab 20052119129
  77. 77. MorinoKPetersenK. FDufourSBefroyDFrattiniJShatzkesNetalReduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents.J Clin Invest 200511535873593
  78. 78. MorinoKPetersenK. FSchulmanG. IMolecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction.Diabetes2006S9S15.
  79. 79. MoeschelKBeckAWeigertCLammersRKalbacherHVoelterWetalProtein kinase C-zeta-induced phosphorylation of Ser(318) in insulin receptor substrate-1 (IRS-1) attenuates the interaction with the insulin receptor and the tyrosine phosphorylation of IRS-1. J Biol Chem 20042792515725163
  80. 80. LiJ. PDefeaKRothR. AModulation of insulin receptor substrate-1 tyrosine phosphorylation by an Akt/phosphatidylinositol 3-kinase pathway.J BiolChem 199927493519356
  81. 81. EgawaKNakashimaNSharmaP. MMaegawaHNagaiYKashiwagiAetalPersistent activation of phosphatidylinositol 3-kinase causes insulin resistance due to accelerated insulin-induced insulin receptor substrate-1 degradation in 3T3-L1 adipocytes.Endocrinology200014119301935
  82. 82. MoeschelKBeckAWeigertCLammersRKalbacherHVoelterWetalProtein kinase C-zeta-induced phosphorylation of Ser318 in insulin receptor substrate-1 (IRS-1) attenuates the interaction with the insulin receptor and the tyrosine phosphorylation of IRS-1.J Biol Chem 20042792515725163
  83. 83. MussigKFiedlerHStaigerHWeigertCLehmannRSchleicherE. DetalInsulin-induced stimulation of JNK and the PI 3-kinase/mTOR pathway leads to phosphorylation of serine 318 of IRS-1 in C2C12 myotubes.Biochem Biophys Res Commun 2005335819825
  84. 84. ArgilesJ. MLopez-sorianoJAlmendroVBusquetsSLopez-sorianoF. JCross-talk between skeletal muscle and adipose tissue: a link with obesity? Med Res Rev 2005254965
  85. 85. KrebsMBrunmairBBrehmAArtwohlMSzendroediJNowotnyPetalThe Mammalian target of rapamycin pathway regulates nutrient-sensitive glucose uptake in man.Diabetes20075616001607
  86. 86. TremblayFBruleSHee Um S, Li Y, Masuda K, Roden M, etal. Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient- and obesity-induced insulin resistance.Proc Natl Acad Sci USA 20071041405614061
  87. 87. BouzakriKRoquesMGualPEspinosaSGuebre-egziabherFRiouJ. PetalReduced activation of phosphatidylinositol-3 kinase and increased serine 636 phosphorylation of insulin receptor substrate-1 in primary culture of skeletal muscle cells from patients with type 2 diabetes.Diabetes20035213191325
  88. 88. YiZLanglaisPDe FilippisE. ALuoMFlynnC. RSchroederSetalGlobal assessment of regulation of phosphorylation of insulin receptor substrate-1 by insulin in vivo in human muscle.Diabetes20075615081516
  89. 89. DamsboPVaagAHother-nielsenOBeck-nielsenHReduced glycogen synthase activity in skeletal muscle from obese patients with and without type 2 (non-insulin-dependent) diabetes mellitus.Diabetologia199134239245
  90. 90. GroopL. CKankuriMSchalin-janttiCEkstrandANikula-ijasPWidenEet alAssociation between polymorphism of the glycogen synthase gene and non-insulin-dependent diabetes mellitus. N Engl J Med 19933281014
  91. 91. RissanenJPihlajamakiJHeikkinenSKekalainenPMykkanenLKuusistoJet alNew variants in the glycogen synthase gene (Gln71His, Met416Val) in patients with NIDDM from eastern Finland. Diabetologia 19974013131319
  92. 92. St-ongeJJoanisseD. RSimoneauJ-AThe stimulation-induced increase in skeletal muscle glycogen synthase content is impaired in carriers of the glycogen synthase XbaI gene polymorphism.Diabetes200150195198
  93. 93. LowellB. BShulmanG. IMitochondrial dysfunction and type 2 diabetes.Science 2005307384387
  94. 94. PetersenK. FBefroyDDufourSDziuraJAriyanCRothmanD. LetalMitochondrial dysfunction in the elderly: possible role in insulin resistance.Science 200330011401142
  95. 95. PetersenK. FDufourSBefroyDGarciaRShulmanG. IImpaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes.N Engl J Med 2004350664671
  96. 96. DiMauro SSchon EA. Mechanisms of disease: mitochondrial respiratory-chain diseases. N Engl J Med 200334826562668
  97. 97. LiYSoosT. JLiXWuJDegennaroMSunXetalProtein kinase θ inhibits insulin signaling by phosphorylating IRS1 at Ser1101. J Biol Chem 20042794530445307
  98. 98. BellK. SShcmitz-peifferCLim-fraserMBidenT. JCooneyG. JKraegenE. WAcute reversal of lipid-induced muscle insulin resistance is associated with rapid alteration in PKC-θ localization. Am J Physiol Endocrinol Metab 2000E1196E1201.
  99. 99. ItaniS. IPoriesW. JMacdonaldK. GDohmG. LIncreased protein kinase C θ in skeletal muscle of diabetic patients. Metabolism 200150553557
  100. 100. BandyopadhyayG. KYuJ. GOfrecioJOlefskyJ. MIncreasedpexpression and decreased phosphatidylinositol 3-kinase activity in insulin-resistant human skeletal muscle. Diabetes 20055423512359
  101. 101. UekiKFrumanD. ABrachmannS. MTsengY. HCantleyL. CKahnC. RMolecular balance between the regulatory and catalytic subunits of phosphoinositide 3-kinase regulates cell signaling and survival.Mol Cell Biol 200222965977
  102. 102. ShepherdP. RWithersD. JSiddleKPhosphoinositide 3-kinase: the key switch mechanism in insulin signaling.Biochem J 1998333471490
  103. 103. InukaiKAnaiMVan BredaEHosakaTKatagiriHFunakiMet alA novel 55-kDa regulatory subunit for phosphatidylinositol 3-kinase structurally similar to 55PIKis generated by alternative splicing of the p85a gene. J Biol Chem 1996
  104. 104. AntonettiD. AAlgenstaedtPKahnC. RInsulin receptor substrate 1 binds two novel splice variants of the regulatory subunit of phosphatidylinositol 3-kinase in muscle and brain.Mol Cell Biol 19961621952203
  105. 105. FrumanD. ACantleyL. CCarpenterC. LStructural organization and alternative splicing of the murine phosphoinositide 3-kinase 85agene. Genomics 1996
  106. 106. InukaiKFunakiMOgiharaTKatagiriHKandaAAnaiMet al85agene generates three isoforms of regulatory subunit for phosphatidylinositol 3-kinase (PI 3-kinase), p50a, p55a, and p85a, with different PI 3-kinase activity elevating responses to insulin. J Biol Chem 1997
  107. 107. PonsSAsanoTGlasheenEMiralpeixMZhangYFischerT. Let alThe structure and function of 55PIKreveal a new regulatory subunit for phosphatidylinositol 3-kinase.Mol Cell Biol 1995
  108. 108. TerauchiYTsujiYSatohSMinouraHMurakamiKOkunoAetalIncreased insulin sensitivity and hypoglycaemia in mice lacking the 85subunit of phosphoinositide 3-kinase. Nat Genet 1999
  109. 109. Mauvais-jarvisFUekiKFrumanD. AHirshmanM. FSakamotoKGoodyearL. JetalReduced expression of the murine 85subunit of phosphoinositide 3-kinase improves insulin signalling and ameliorates diabetes. J Clin Invest 2000
  110. 110. UekiKFrumanD. AYballeC. MFasshauerMKleinJAsanoTetalPositive and negative roles of 85and p85β regulatory subunits of phosphoinositide 3-kinase in insulin signaling. J BiolChem 2003
  111. 111. GiorginoFPedriniM. TMateraLSmithR. JSpecific increase in 85expression in response to dexamethazone is associated with inhibition of insulin-like growth factor-I stimulated phosphatidylinositol 3-kinase activity in cultured muscle cells. J Biol Chem 1997
  112. 112. LeeY. HGiraudJDavisR. JWhite MF. C-Jun N-terminal kinase (JNK) mediates feedback inhibition of the insulin signaling cascade.J Biol Chem 200327828962902
  113. 113. UekiKYballeC. MBrachmannS. MVicentDWattJ. MKahnC. RetalIncreased insulin sensitivity in mice lacking 85subunit of phosphoinositide 3-kinase. Proc Natl Acad Sci U S A 2002
  114. 114. LamiaK. APeroniO. DKimY-BRamehL. EKahnB. BCantleyL. CIncreased insulin sensitivity and reduced adiposity in phosphatidylinositol 5-phosphate 4-kinase β-/- mice. Mol Cell Biol 20042450805087
  115. 115. BarbourL. AShaoJQiaoLLeitnerWAndersonMFriedmanJ. EetalHuman placental growth hormone increases expression of 85regulatory unit of phosphatidylinositol 3-kinase and triggers severe insulin resistance in skeletal muscle.Endocrinology2004
  116. 116. CornierM-ABessesenD. HGurevichILeitnerJ. WDrazninBNutritional up-regulation of 85expression is an early molecular manifestation of insulin resistance. Diabetologia 2006
  117. 117. BarbourL. ARahmanS. MGurevichILeitnerJ. WFisherSRoperMetalIncreased 85alphais a potent negative regulator of skeletal muscle insulin signaling and induces in vivo insulin resistance associated with growth hormone excess.J BiolChem 2005
  118. 118. KirwanJVarastehpourAJingMPresleyLShaoJFriedmanJ. EetalReversal of insulin resistance post-partum is linked to enhanced skeletal muscle insulin signaling. J ClinEndocrinolMetab 20048946784684
  119. 119. KlipAPaquetM. RGlucose transport and glucose transporters in muscle and their metabolic regulation.Diabetes Care199013228243
  120. 120. RandleP. JPriestmanD. AMistryS. CHalsallAGlucose fatty acid interactions and the regulation of glucose disposalJ Cell Biochem 1994111
  121. 121. SalorantaCGroopLInteractions between glucose and FFA metabolism in man.Diabetes Metab Rev 1996121536
  122. 122. BodenGRole of fatty acids in the pathogenesis of insulin resistance and NIDDM [published erratum appears in Diabetes 1997Mar;46(3):536]. Diabetes 1997; 46310
  123. 123. FoleyJ. ERationale and application of fatty acid oxidation inhibitors in treatment of diabetes mellitus.Diabetes Care199215773784
  124. 124. NurjhanNConsoliAGerichJIncreased lipolysis and its consequences on gluconeogenesis in non-insulin-dependent diabetes mellitus.J Clin Invest 199289169175
  125. 125. McGarryBanting lecture. Dysregulation of fatty acid metabolism in the etiology of type 2 diabetes.Diabetes200151718
  126. 126. MollerD. ENew drug targets for type 2 diabetes and the metabolic syndrome. Nature (London) 2001414821827
  127. 127. DresnerALaurentDMarcucciMGriffinM. EDufourSClineG. WetalEffects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity.J Clin Invest 1999103253259
  128. 128. NewtonA. CRegulation of the ABC kinases by phosphorylation: protein kinase C as a paradigm.Biochem J 2003370361371
  129. 129. SamuelV. TLiuZ. XQuX. QElderB. DBilzSBefroyDetalMechanism of hepatic insulin resistance in non-alcoholic fatty liver disease.J Biol Chem 20042793234532353
  130. 130. SavageD. BChoiC. SSamuelV. TLiuZ. XZhangD. YWangAetalReversal of diet-induced hepatic steatosis and hepatic insulin resistance by antisense oligonucleotide inhibitors of acetyl-CoA carboxylases 1 and 2.J Clin Invest 2006116817824
  131. 131. AcciliDArdenK. CFoxOs at the crossroads of cellular metabolism, differentiation, and transformation.Cell2004117421426
  132. 132. NeschenSMorinoKHammondL. EZhangD. YLiuZ. XRomanelliA. JetalPrevention of hepatic steatosis and hepatic insulin resistance in mitochondrial acyl-CoA: glycerol-sn-3-phosphate acyltransferase 1 knockout miceCell Metab 200525565
  133. 133. PetersenK. FDufourSBefroyDLehrkeMHendlerR. EShulmanG. IReversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes.Diabetes200554603608
  134. 134. BodenGShulmanG IFree fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction.Eur. J. Clin. Invest.2002Suppl. 31423
  135. 135. ItaniS IRudermanN BFrankSBodenGLipid induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and Ikb-α. Diabetes 20025120052011
  136. 136. StrackVStoyanovBBossenmaierBMosthafLKellererMHaringH UImpact of mutations at different serine residues on the tyrosine kinase activity of the insulin receptor.Biochem. Biophys. Res. Commun.1997239235239
  137. 137. MichaelL. FWuZCheathamR. BPuigserverPAdelmantGLehmanJ. Jet alRestoration of insulin-sensitive glucose transporter (GLUT4) gene expression in muscle cells by the transcriptional coactivator PGC-1Proc Natl Acad Sci USA 20019838203825
  138. 138. AkimotoTRibarT. JWilliamsR. SYanZSkeletal muscle adaptation in response to voluntary running in Ca2/calmodulin-dependent protein kinase IV-deficient mice.Am J Physiol Cell Physiol 2004C1311C1319.
  139. 139. BergeronRRenJ. MCadmanK. SMooreI. KPerretPPypaertMetalChronic activation of AMP kinase results in NRF-1 activation and mitochondrial biogenesis.Am J Physiol Endocrinol Metab 2001E1340E1346.
  140. 140. ZongH. HRenJ. MYoungL. HPypaertMMuJBirnbaumM. JShulman GI: AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivationProc Natl Acad Sci U S A9920021598315987
  141. 141. WinderW. WHolmesB. FRubinkD. SJensenE. BChenMHolloszyJ. OActivation of AMP-activated protein kinase increases mitochondrial enzymes in skeletal muscle.J Appl Physiol 20008822192226
  142. 142. Scarpulla RC: Nuclear control of respiratory gene expression in mammalian cellsJ Cell Biochem 200697673683
  143. 143. LingCPoulsenPCarlssonERidderstraleMAlmgrenPWojtaszewskiJetalMultiple environmental and genetic factors influence skeletal muscle PGC-1 alpha and PGC-1 beta gene expression in twins. J Clin Invest 200411415181526
  144. 144. SaltielA. RKahnC. RInsulin signalling and the regulation of glucose and lipid metabolism.Nature2001414799806
  145. 145. UysalK. TWiesbrockS. MMarinoM. WHotamisligilG. SProtection from obesity-induced insulin resistance in mice lacking TNF-alpha function.Nature1997389610614
  146. 146. MontagueC. TFarooqiI. SWhiteheadJ. PSoosM. ARauHWarehamN. Jet alCongenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 1997387903908
  147. 147. FarooqiI. SJebbS. ALangmackGLawrenceECheethamC. HPrenticeA. Met alEffects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 1999341879884
  148. 148. StumvollMHäringHResistin and adiponectin-of mice and men.Obes Res 20021011971199
  149. 149. MoonBKwanJ. JDuddyNSweeneyGBegumNResistin inhibits glucose uptake in L6 cells independently of changes in insulin signalling and GLUT4 translocationAm J PhysiolEndocrinolMetab 2003E106115
  150. 150. PravenecMKazdovaLLandaVZidekVMlejnekPJansaPet alTransgenic and recombinant resistin impair skeletal muscle glucose metabolism in the spontaneously hypertensive rat. J BiolChem 20032784520945215
  151. 151. HallR. KYamasakiTKuceraTWaltner-lawMOBrienRGrannerDK. Regulation of phosphoenolpyruvatecarboxykinase and insulin-like growth factor-binding protein-1 gene expression by insulin. J BiolChem 20002753016930175
  152. 152. SchmollDWalkerK. SAlessiDGremplerRBurchellAGuoSetalRegulation of glucose-6-phosphatase gene expression by protein kinase B alpha and the forkhead transcription factor FKHR. J BiolChem 20002753632436333
  153. 153. WolfrumCBesserDLucaEStoffelMInsulin regulates the activity of forkhead transcription factor Hnf-3/Foxa-2 by Akt-mediated phosphorylation and nuclear/ cytosolic localizationProc Natl Acad Sci USA 20031001162411629
  154. 154. NakaeJBiggsW. Hrd, Kitamura T, Cavenee WK, Wright CV, Arden KC et al. Regulation of insulin action and pancreatic β-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1. Nat Genet 200232245253
  155. 155. ZhaoXGanLPanHKanDMajeskiMAdamS. Aet alMultiple elements regulate nuclear/cytoplasmic shuttling of FOXO1: characterization of phosphorylation- and 14-3-3-dependent and-independent mechanisms. Biochem J 2004378839849
  156. 156. YoonJ. CPuigserverPChenGDonovanJWuZRheeJet alControl of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 2001413131138
  157. 157. HerzigSLongFJhalaU. SHedrickSQuinnRBauerAet alCREB regulates hepatic gluconeogenesis through the co-activator PGC- 1. Nature 2001413179183
  158. 158. PuigserverPRheeJDonovanJWalkeyC. JYoonJ. COrienteFet alInsulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1α interaction. Nature 2003423550555
  159. 159. ShulmanG. ICellular mechanisms of insulin resistance. J Clin Invest 2000106171176
  160. 160. TokerANewtonA. CCellular signaling: pivoting around PDK-1.Cell 2000103185188
  161. 161. BhattacharyaSDeyDRoyS. SMolecular mechanisms of insulin resistance. J. Biosci. 200732405413
  162. 162. DeyDBasuDRoyS SBandyopadhyayABhattacharyaSInvolvement of novel PKC isoforms in FFA induced defects in insulin signalling. Mol. Cell. Endocrinol.2006266064
  163. 163. DeyDMukherjeeMBasuDDattaMRoyS SBandyopadhyayAetalInhibition of insulin receptor gene expression and insulin signaling by fatty acid: interplay of PKC isoforms thereinCell. Physiol. Biochem.200516217228

Written By

Sarika Arora

Submitted: 24 October 2012 Published: 12 December 2012