Molecular Mechanism of Insulin Resistance in Obesity and Type 2 Diabetes

Article information

Korean J Intern Med. 2010;25(2):119-129
Publication date (electronic) : 2010 June 01
doi : https://doi.org/10.3904/kjim.2010.25.2.119
Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
Correspondence to Young-Bum Kim, Ph.D. Beth Israel Deaconess Medical Center, 330 Brookline Ave., Center for Life Science-736, Boston, MA 02115, USA. Tel: 1-617-735-3216, Fax: 1-617-735-3323, ykim2@bidmc.harvard.edu

Abstract

Insulin resistance is a major risk factor for developing type 2 diabetes caused by the inability of insulin-target tissues to respond properly to insulin, and contributes to the morbidity of obesity. Insulin action involves a series of signaling cascades initiated by insulin binding to its receptor, eliciting receptor autophosphorylation and activation of the receptor tyrosine kinase, resulting in tyrosine phosphorylation of insulin receptor substrates (IRSs). Phosphorylation of IRSs leads to activation of phosphatidylinositol 3-kinase (PI3K) and, subsequently, to activation of Akt and its downstream mediator AS160, all of which are important steps for stimulating glucose transport induced by insulin. Although the mechanisms underlying insulin resistance are not completely understood in skeletal muscle, it is thought to result, at least in part, from impaired insulin-dependent PI3K activation and downstream signaling. This review focuses on the molecular basis of skeletal muscle insulin resistance in obesity and type 2 diabetes. In addition, the effects of insulin-sensitizing agent treatment and lifestyle intervention of human insulin-resistant subjects on insulin signaling cascade are discussed. Furthermore, the role of Rho-kinase, a newly identified regulator of insulin action in insulin control of metabolism, is addressed.

INTRODUCTION

A fundamental mechanism for the maintenance of glucose homeostasis is the rapid action of insulin to stimulate glucose uptake and metabolism in peripheral tissues. Skeletal muscle is the primary site of glucose disposal in the insulin-stimulated state [1]. Resistance to the actions of insulin in skeletal muscle is a major pathogenic factor in type 2 or type 1 diabetes mellitus [2,3]; it also contributes to the morbidity of obesity, and complicates poorly controlled type 1 (autoimmune) diabetes [2]. The ability of insulin to increase glucose transport in skeletal muscle is elicited by the translocation of glucose transporter 4 (Glut4), the major insulin regulated glucose transporter, from intracellular vesicles to the plasma membrane and transverse tubules [4]. In muscle of type 2 diabetic subjects, the expression of the Glut4 gene is normal, and impaired glucose uptake by insulin action most likely results from altered trafficking or impaired function of Glut4 [5-7]. Because glucose transport in response to other stimuli that use different signaling pathways is normal in muscle of type 2 diabetic subjects [4], the resistance to insulin stimulation may be due to impaired insulin signal transduction [8]. In this review, we mainly summarize the updated information on insulin signaling over the past decade, with particular emphasis on the molecular mechanism of human insulin resistance, and also address the physiological role of the newly identified player of insulin action.

Insulin receptor signaling

Insulin signaling involves a cascade of events initiated by insulin binding to its cell surface receptor, followed by receptor autophosphorylation, and activation of receptor tyrosine kinases, which result in tyrosine phosphorylation of insulin receptor substrates (IRSs) including IRS1, IRS2, IRS3, IRS4, Gab1, and Shc [9,10]. Binding of IRSs to the regulatory subunit of phosphoinositide 3-kinase (PI3K) via Src homology 2 (SH2) domains results in activation of PI3K, which phosphorylates membrane phospholipids and phosphatidylinositol 4,5-bisphosphate (PIP2) on the 3' position. This complex activates the 3-phosphoinositide-dependent protein kinases (PDK-1 and PDK-2) resulting in activation of Akt/protein kinase B (PKB) and atypical protein kinase C λ and ζ, (PKCλ/ζ), each of which are serine/threonine kinases [11,12]. Activated Akt phosphorylates its 160 kDa substrate (AS160), which stimulates the translocation of insulin-mediated Glut4 from intracellular vesicles to the plasma membrane [13]. Moreover, activation of PKCλ/ζ is also involved in the regulation of Glut4 translocation in response to insulin [14,15]. However, the insulin receptor (IR) is also dephosphorylated and inactivated by protein tyrosine phosphatases (PTPs), which comprise an extensive family of proteins that exert negative effects on insulin action and glucose metabolism [16,17]. In addition, phosphatase and tension homologue deleted on chromosome 10 (PTEN), a lipid phosphatase, serves as an important negative modulator for the insulin signaling pathway by hydrolyzing phosphatidylinositol 3,4,5-triphosphate to PIP2, antagonizing the PI3K pathway [18,19]. Thus, the physiological regulation of insulin action is controlled by the balance between phosphorylation and dephosphorylation (Fig. 1). Most importantly, the PI3K pathway is thought to be a key component of the insulin signaling cascade, which is necessary for the metabolic effects of insulin on glucose transport and Glu4 translocation [20,21]. Indeed, insulin-stimulated PI3K activity decreases in skeletal muscle of type 2 diabetic subjects [8,22], providing evidence for a defect in insulin signaling that could contribute to impaired Glut4 translocation and insulin resistance.

Figure 1

The insulin signaling pathway. PTP1B, protein-tyrosine phosphatase 1B; IRS, insulin receptor substrate; ROCK, Rho-kinase; PIP, phosphatidylinositol phosphate; PTEN, phosphatase and tension homologue deleted on chromosome 10; PH domain, pleckstrin homology domain; PDK, phosphoinositide-dependent protein kinase; GβL, G-protein beta subunit like; mTOR, mammalian target of rapamycin; AS160, 160 kDa Akt substrate; PKCλ/ζ, protein kinase C λ and ζ; Glut4, glucose transporter 4.

Role of Akt in insulin signaling

The serine/threonine kinase Akt is a downstream mediator of PI3K [12]. Three Akt isoforms have been cloned [23,24]; Akt1, Akt2, and Akt3, all of which are ubiquitously expressed in the tissues. Insulin has differential effects on Akt isoforms in a tissue-, isoform-, and species-specific manner [25,26]. In obese rats, insulin-stimulated Akt1 activity decreases in muscle and adipose tissue but increases in liver, while insulin-stimulated Akt2 activity decreases in muscle and liver but increases in adipose tissue [25]. Some studies have shown that inhibition of Akt activation by DN-Akt expression has no significant effect on insulin-stimulated glucose transport, whereas Akt siRNA inhibits insulin-stimulated glucose transport in 3T3-L1 adipocytes [27]. Furthermore, in skeletal muscles from insulin-resistant models, including PTP-1B transgenic mice [28], leukocyte antigen-related phosphatase (LAR) transgenic mice [29], and glucosamine-infused rats [30], insulin-induced Akt activation is normal but insulin-stimulated glucose uptake is impaired. In skeletal muscle of obese insulin resistant humans with or without type 2 diabetes, the effect of insulin on the activity of all Akt isoforms in muscle in vivo is normal [8]. However, at very high insulin levels in vitro, Akt activity is diminished in muscle from non-obese type 2 diabetics [31]. Moreover, Akt2 phosphorylation is impaired in adipocytes from obese type 2 diabetics [32]. Whether these impairments in Akt1 or Akt2 activity are sufficient to play a role in insulin resistant states is unclear. Akt1 knockout mice do not have insulin resistance, although their growth retardation or developmental effects could mask this [33]. Akt2 knockout mice have impaired insulin action on liver and modest effects in muscle and adipocytes that could possibly be secondary to the liver defect [34]. In this regard, our studies have also demonstrated that insulin-stimulated Akt activity in skeletal muscle of these obese rats is very mildly reduced while PI3K activity is markedly reduced, as compared to lean rats [25]. These data lead us to propose the hypothesis that full activation of PI3K is not necessary to maximally activate Akt, and that other pathways that are independent of PI3K are also involved in activation of Akt by insulin, at least in insulin resistant states. Data in support of the hypothesis come from studies showing that insulin-stimulated PI3K activation is reduced by 50% to 60% while Akt activation is normal in hepatoma cells transduced with a dominant-interfering mutant of dynamin [35]. Conceivably, such pathways could be induced in altered metabolic states such as obesity or type 2 diabetes. Thus, the potential impact of altered Akt activity in insulin-dependent glucose metabolism remains uncertain. This important issue can be resolved by studying an animal model that has Akt selectively deleted in peripheral tissues. Until now, there are no data regarding the effects of tissue-specific deletion of Akt on glucose metabolism and insulin signaling.

AS160 is an Akt substrate involved in Glut4 translocation

Investigation of steps down-stream of Akt could shed light on the role of Akt in insulin resistance. Akt directly phosphorylates cytosol proteins that have the RXRXXS/T motif. Using a phosphospecific antibody that only recognizes serine or threonine residues, which are phosphorylated by Akt [36], the Lienhard group has discovered the AS160 from 3T3-L1 adipocytes. This molecule was originally named TBC1 domain family member 4 (TBC1D4) and contains two phosphotyrosine binding (PTB) domains at the NH2 terminus and a Rab GTPase activating protein (GAP) domain at the COOH terminus [37]. In the basal state of 3T3-L1 adipocytes, AS160 was mainly localized in the low density microsomes (LDM) fraction. However, upon insulin stimulation, it was redistributed from the LDM compartment to the cytosol, but these effects were significantly blocked by treatment with a PI3K inhibitor [36]. Given that one of the LDM components are vesicles containing the glucose transporter Glut4 [38], and they move to and fuse with the plasma membrane in response to insulin [39], it is conceivable that AS160 may be involved in the process of GLUT4 trafficking that is dependent on PI3K signaling. The Lienhard group further demonstrated the important role of AS160 in the regulation of Glut4 translocation by showing that insulin causes a marked increase in AS160 phosphorylation at Ser318, Ser570, Ser588, Thr642, and Thr751 residues, and that mutation of these sites inhibits Glut4 translocation in response to insulin [13]. Support for this finding comes from skeletal muscle studies showing that insulin stimulates AS160 phosphorylation in skeletal muscle in a PI3K-dependent fashion, and that contraction and the AMP-activated protein kinase (AMPK) activator aminoimidazole carboxamide ribonucleotide (AICAR) also increases AS160 phosphorylation in isolated rat epitrochlearis muscle and other muscle systems [40-44]. Moreover, in skeletal muscle of human insulin-resistant subjects, including those with polycystic ovary syndrome and type 2 diabetes, the ability of insulin to increase AS160 phosphorylation is significantly impaired [45,46]. Collectively, the current available data provide important evidence that insulin-stimulated phosphorylation of AS160 is required to regulate Glut4 translocation, which is a critical step in controlling glucose homeostasis, and that decreased insulin-induced AS160 phosphorylation in skeletal muscle may play an important role in insulin resistance in vivo.

Role of PKCλ/ζ in the regulation of insulin-mediated glucose transport

The atypical PKC isoforms λ and ζ, are downstream PI3K mediators, and their activation is required for insulin stimulation of glucose uptake [47-49]. Overexpression of a dominant negative mutant of PKCλ or PKCζ abrogates insulin-stimulated glucose transport and Glut4 translocation in adipose [47,50] and muscle cells [48,49]. Overexpression of constitutively active PKCλ in adipocytes [47] or wildtype PKCζ in muscle in vivo [51] enhances both basal and insulin-stimulated glucose transport. In addition, PKCλ and ζ appear to function interchangeably, as overexpression of wild type PKCλ restores the inhibitory effects of a dominant negative mutant of PKCζ on insulin-stimulated Glut4 translocation, and vice versa [52]. The possibility that PKCλ/ζ could play an important role in insulin resistance in vivo is supported by studies showing impaired insulin-stimulated PKCλ/ζ activity in skeletal muscle and adipose tissue of non-obese type 2 diabetic GK rats [53,54]. Moreover, insulin-stimulated PKCλ/ζ activity decreases in cultured myotubes of obese humans with impaired glucose tolerance [55] and in muscle of obese diabetic monkeys [56]. Along with these findings, the Farese group have demonstrated that muscle-specific deletion of PKCλ/ζ causes insulin resistance by reducing insulin-mediated glucose transport in skeletal muscle [15]. Importantly, insulin-stimulated PKCλ/ζ activity is reduced in the muscle of obese nondiabetic and obese type 2 diabetic subjects. This contrasts with our findings that Akt activation is normal in obese and diabetic subjects with similar metabolic characteristics [8]. Thus, although insulin-induced PI3K activity is reduced in type 2 diabetic subjects, not all downstream pathways are similarly affected. These data imply that reduced insulin-stimulated atypical PKC activity may play an important role in insulin resistance in vivo.

Insulin-sensitizer agents and insulin signaling

Thiazolidinediones (TZDs) are a new class of insulin-sensitizing agents being used for the treatment of type 2 diabetes [57]. The molecular targets of these compounds are thought to include the nuclear receptor, peroxisome proliferator activator receptor-γ (PPARγ), which regulates the expression of numerous genes that affect glucose and lipid metabolism [58]. Evidence suggests that TZDs ameliorate insulin resistance in humans primarily by increasing insulin-stimulated glucose disposal in skeletal muscle [59]. Our previous studies revealed that treatment with troglitazone, a member of the TZD family, increases insulin-stimulated IRS-1-associated PI3K activity and Akt activity in skeletal muscle of type 2 diabetic patients [60]. The troglitazone effect on PI3K activity was associated with an increase in the amount of the p110β catalytic subunit of PI3K [60]. Consistently, enhanced Akt phosphorylation was also detected in skeletal muscle in normal, glucose-tolerant, insulin-resistant, first-degree relatives of type 2 diabetic patients [61]. These findings suggest that the mechanism for the insulin-sensitizing effect of TZDs could involve enhanced PI3K activation in skeletal muscle of obese type 2 diabetic subjects. However, Karsson et al. [62] demonstrated that insulin action on Akt phosphorylation and PI3K activity is unaltered in skeletal muscle of human subjects with newly diagnosed type 2 diabetes after treatment of rosiglitazone, another member of the TZD family. This discrepancy could be due to either the nature of the human subjects (obese type 2 diabetic vs. lean type 2 diabetic) or the different kinds of TZDs. From the view of the current human data, it is somewhat unlikely that changes in insulin signaling can fully account for the improvement of insulin sensitivity and glucose disposal in skeletal muscle in response to TZD treatment.

Given that correlative changes in fatty acid metabolism and improvements in glucose homeostasis and insulin sensitivity may imply an indirect effect on skeletal muscle via adipose tissue [63,64], it is possible that some of the effects of TZDs on insulin signaling work through secondary mechanisms. One potential factor is fatty acids, as elevations of fatty acids in plasma and the lipid content of muscle are associated with insulin resistance [65]. As previously reported [66,67], troglitazone treatment tends to lower triglyceride and free fatty acid concentrations. TZDs also reduce accumulation of muscle triglycerides and diacylglycerol [68]. Activation of PKC by elevated diacylglycerol levels in muscle impairs insulin signaling [69,70]. This raises the possibility that decreases in plasma lipid concentrations with TZD treatment could lead to a reduction of diacylglycerol in muscle, reducing PKC activation, resulting in an improvement of the insulin signaling cascade. It is also possible that TZDs reduce intramyocellular lipid content by promoting storage of free fatty acids in adipocyte triglycerides via PPARγ, redirecting free fatty acids from skeletal muscle to adipose tissue. Thus, the ability of troglitazone to improve insulin action in skeletal muscle could involve actions on both adipose tissue and muscle.

Metformin is a member of another class of drugs that are effective in lowering glucose concentrations in patients with type 2 diabetes [71]. A number of studies have demonstrated that metformin inhibits gluconeogenesis, reduces hepatic glucose output, and lowers fasting blood glucose concentration [59,72]. In addition to its effect on the liver, metformin also appears to decrease glucose concentrations by increasing peripheral insulin sensitivity and augmenting insulin-mediated glucose uptake in skeletal muscle of type 2 diabetic subjects [73]. The precise mechanism for this action of metformin is incompletely understood, but in vitro studies indicate it could involve multiple effects, including increased translocation of Glut1 and Glut4 glucose transporters from intracellular vesicles to the cell surface [74] and increased binding of insulin to cell surface insulin receptors [75]. Evidence shows that metformin normalizes insulin receptor tyrosine phosphorylation and PI3K and Akt activity in adipocytes exposed to high insulin levels in vitro for long periods [76]. However, our studies have demonstrated that the effects of metformin on PI3K and Akt activity in skeletal muscle of human insulin resistant subjects with obesity and type 2 diabetes in vivo are unchanged [60]. These data are further confirmed in skeletal muscle of patients with newly diagnosed type 2 diabetes [62]. Overall, it is clear that enhanced insulin-mediated glucose disposal by metformin therapy in type 2 diabetic patients is independent of improved insulin signaling in skeletal muscle of type 2 diabetic subjects.

Effects of weight loss therapy on skeletal muscle insulin signaling

Given that obesity is the major risk factor for developing insulin resistance, it is no doubt that lifestyle intervention that produces weight loss, improves insulin sensitivity. A reduction of fat mass by weight loss results in a significant decrease in lipid oxidation and an enhanced glucose homeostasis [77]. In addition, insulin secretion and plasma insulin concentrations decrease significantly after weight loss. Our work has investigated whether weight loss therapy can alter the ability of insulin to activate PI3K and downstream signaling in skeletal muscle of obese nondiabetic patients. For these studies, ten obese nondiabetic subjects (body mass index [BMI], 30 to 45 kg/m2) were challenged on a very low calorie diet (VLCD) of 600 to 800 calories per day for up to 24 weeks or until 10 to 15% of initial body weight was lost [78]. Once the weight loss goal was met, the subjects were introduced to a weight-maintenance diet. Glucose disposal rate measured by euglycemic hyperinsulinemic clamp was increased by 30% after VLCD treatment, indicating enhanced systemic insulin sensitivity. VLCD treatment significantly increased IRS-1 tyrosine phosphorylation, compared with the pretreatment level [78]. In parallel, insulin-stimulated IRS-1-associated PI3K activity was increased 2-fold post-treatment with VLCD, compared with pretreatment [78]. These changes were independent of the total amount of IRS-1 protein. Importantly, the impaired PKCλ/ζ activation in obese nondiabetic humans was reversed with weight reduction. This could be due to increased signaling upstream of PKCλ/ζ. This, combined with the observation that treating diabetic subjects with TZDs reverses the defect in PKCλ/ζ activity [79], suggests that reversal of the PKCλ/ζ defect might enhance insulin sensitivity in obese insulin-resistant humans. In line with these findings, recent studies of severely obese individuals also demonstrated that weight loss with a very low energy diet (1,883 kJ/day) markedly enhances insulin sensitivity by improving insulin-stimulated glucose disposal [77]. This enhanced sensitivity was accompanied by increased insulin signaling at the level of AS160 and proline-rich Akt substrate 40, which is a component of the mammalian target of the rapamycin nutrient-sensing pathway. Taken together, weight reduction in obese individuals improves insulin sensitivity, which may result from an improvement in PI3K and its downstream signaling in skeletal muscle.

Although our studies have not investigated Glut4 translocation in response to insulin in skeletal muscle of obese subjects that have undergone weight loss, a number of studies have demonstrated that enhancement in the insulin signaling pathway was not accompanied by a significant improvement in Glut4 translocation to the plasma membrane in skeletal muscle from type 2 diabetic patients, TZD-treated type 2 diabetic patients, and obese subjects who underwent gastric bypass surgery [62,80,81]. This could be explained by the fact that intrinsic activity of Glut4 at the plasma membrane is increased or that an unidentified glucose transporter, which may contribute to increased glucose disposal in skeletal muscle, is involved in this event.

Protein tyrosine phosphatase 1B and insulin signaling

Tyrosine phosphorylation of the IR is reversible, and IR dephosphorylation takes place rapidly in intact cells even with the continued presence of insulin [82]. Because a critical regulatory step in insulin signal transduction is the dephosphorylation of signaling molecules by PTPs, it is plausible that enhanced activity of one or more PTPs could lead to insulin resistance. Several studies of obese humans and rodents have reported that the expression and/or activity of specific PTPs, including the transmembrane LAR and the intracellular enzymes protein-tyrosine phosphatase 1B (PTP1B) and Src-homology 2 domain-containing phosphatase-2 (SHP2, SHPTP2, or syp), increase in muscle and adipose tissue [83-87]. LAR and PTP1B show the greatest increases (3-fold in muscle, 2-fold in fat) [83,88]. There is a strong correlation between BMI and total PTP activity toward the IR in both skeletal muscle and adipose tissue from lean and obese subjects [83,88]. Recent studies have also observed increased levels of LAR and PTP1B expression in liver and muscle from obese rodents [89].

Support for the role of PTPs in the regulation of insulin action comes from transgenic and gene knockout studies. The Kahn group generated PTP1B null mice by targeted disruption of the ATG-coding exon [90]. Elchebly et al. [91] targeted exons 5 and 6 (Ex5/6-/-) and also obtained PTP1B-null mice. Both lines of mice have increased insulin sensitivity, manifested by enhanced insulin-stimulated phosphorylation of IR and IRS-1 in muscle and liver. PTP1B-deficient mice have reduced body fat and are protected from diet-induced obesity due, at least in part, to increased basal metabolic rate and total energy expenditure [90]. In addition, insulin-stimulated whole-body glucose disposal is enhanced in PTP1B-deficient mice [90]. Surprisingly, this effect is tissue-specific; insulin-stimulated glucose uptake is elevated in skeletal muscle but not in adipose tissue. These data suggest that overexpression of PTP1B in insulin-target tissues in vivo could contribute to insulin resistance. Consistent with this hypothesis, Zabolotny et al. [28] demonstrated that selective overexpression of PTP1B in skeletal muscle impairs insulin-stimulated PI3K activity and causes mild insulin resistance in vivo. Moreover, recent studies have indicated that liver-specific deletion of PTP1B improves insulin resistance and attenuates diet-induced endoplasmic reticulum stress [92]. Similar to these findings, Haj et al. [93] showed that liver-specific re-expression of PTP1B in PTP1B deficient mice leads to marked attenuation of their enhanced insulin sensitivity. In obese diabetic insulin resistant ob/ob and db/db mice, PTP1B antisense oligonucleotide (ASO) treatment reduces PTP1B protein and mRNA level in liver and fat, and normalizes plasma glucose levels along with improved glucose tolerance and insulin sensitivity [94,95]. Taken together, the current data indicate that inhibition of PTP1B in peripheral tissues may be useful for treating metabolic-related disorders such as obesity and type 2 diabetes. In fact, the development of PTP1B inhibitors has received much attention by the pharmaceutical industry. However, it has been difficult to identify a selective, safe, and effective PTP1B inhibitor, although a novel PTP1B inhibitor (JTT-551) has been suggested as a potential therapeutic agent [96].

Rho-kinase and IRS-1 serine phosphorylation

Rho-kinase (ROCK) is a serine/threonine protein kinase identified as a GTP-Rho-binding protein [97]. There are two isoforms, ROCK1 (also known as ROKβ) [98,99] and ROCK2 (also known as ROKα) [98,100]. ROCK participates in the insulin signaling network by interacting with IRS-1 [101,102]. Our work has demonstrated that inhibiting ROCK decreases insulin-stimulated IRS-1-associated PI3K activity in adipocytes and myotubes. This effect is mainly due to decreased tyrosine phosphorylation of the YXXM motif in IRS-1, which can lead to reduced interaction of IRS-1 with the p85 subunit of PI3K. Indeed, insulin-stimulated IRS-1 binding to the p85 regulatory subunit of PI3K is impaired in adipocytes expressing dominant negative ROCK [103]. By mass spectrometry analysis, we identified the serine residues of IRS-1 at serine 632/635, serine 936, and serine 972, all of which are phosphorylated by ROCK. Interestingly, these sites are close to the YMXM motif domain in IRS-1, which is the binding site of the p85 regulatory subunit of PI3K. Evidence indicates that serine phosphorylation of IRS-1 is a key regulator of insulin signaling [104]. However, the effects of phosphorylation of individual IRS-1 serine residues on insulin signaling appear to be complex and may be context dependent; they are still being defined. Studies of the effects of IRS-1 serine 632/635 phosphorylation on insulin action have yielded conflicting results. Our studies have demonstrated that replacing IRS serines 632 and 635 with alanine causes a significant inhibition of insulin-stimulated IRS-1 tyrosine phosphorylation and PI3K activity [103], suggesting a positive role for IRS-1 serine 632/635 in insulin action.

Our recent studies have demonstrated that global deletion of ROCK1 in mice results in whole-body insulin resistance and impaired skeletal muscle insulin signaling. These effects are independent of changes in body adiposity. Insulin-stimulated IRS-1 serine 632/635 phosphorylation and PI3K activity associated with IRS-1 or phosphotyrosine are impaired in skeletal muscle of ROCK1 deficient mice [105]. Impaired insulin-induced IRS-1 serine 632/635 phosphorylation also accompanies decreased PI3K and ROCK activation in skeletal muscle of obese, insulin-resistant or diabetic mice and rats (unpublished data), suggesting that impaired IRS-1 serine 632/635 phosphorylation may be an important mechanism contributing to the pathogenesis of insulin resistance in obesity. In addition, insulin-stimulated ROCK activity is impaired in skeletal muscle of obese diabetic mice and insulin-resistant humans with obesity and type 2 diabetes (unpublished data). Importantly, insulin-stimulated ROCK1 activity was positively correlated with glucose disposal rate, suggesting that defective ROCK1 activation may contribute to the pathogenesis of human insulin resistance. Thus, our studies identify ROCK1 as a novel player regulating insulin-mediated glucose metabolism in vivo. Further studies of ROCK1 and ROCK2 functions in different metabolic tissues will be needed to precisely delineate ROCK isoform functions that regulate tissue and whole-body insulin sensitivity and glucose homeostasis. The emergence of ROCK1 as an important regulator of insulin action could lead to new treatment approaches for obesity and type 2 diabetes.

CONCLUSION

The earliest defect in the development of type 2 diabetes is insulin resistance characterized by decreased glucose transport and metabolism in skeletal muscle. Studies with the skeletal muscle of type 2 diabetic humans demonstrate impaired insulin activation of the IRS-1/PI3K/Akt signaling pathway, which is a critical step in the regulation of glucose transport in response to insulin. These defects are selectively restored by treatment with an insulin-sensitizing agent and lifestyle changes, representing the core of insulin signaling components. Recent advances have also revealed that insulin action on AS160 phosphorylation is diminished in skeletal muscle of type 2 diabetic patients, and that inhibiting AS160 causes a significant decrease in insulin-dependent translocation of Glut4, suggesting an important role for AS160 in glucose metabolism. Furthermore, defective ROCK activity in skeletal muscle may also contribute to impaired glucose homeostasis in type 2 diabetic patients. A better understanding of the disease pathogenesis and the potential alternative pathways for regulating glucose metabolism could lead to new therapeutic targets for obesity and type 2 diabetes by clarifying intracellular defects in the insulin signaling cascade.

Notes

No potential conflict of interest relevant to this article was reported.

References

1. DeFronzo RA. Pathogenesis of type 2 diabetes: metabolic and molecular implications for identifying diabetes. Diabetes Rev 1997;5:177–269.
2. Yki-Järvinen H, Sahlin K, Ren JM, Koivisto VA. Localization of rate-limiting defect for glucose disposal in skeletal muscle of insulin-resistant type I diabetic patients. Diabetes 1990;39:157–167. 2121571.
3. Petersen KF, Shulman GI. Pathogenesis of skeletal muscle insulin resistance in type 2 diabetes mellitus. Am J Cardiol 2002;90(5A):11G–18G.
4. Kahn BB. Lilly lecture 1995. Glucose transport: pivotal step in insulin action. Diabetes 1996;45:1644–1654. 8866574.
5. Garvey WT, Huecksteadt TP, Matthaei S, Olefsky JM. Role of glucose transporters in the cellular insulin resistance of type II non-insulin-dependent diabetes mellitus. J Clin Invest 1988;81:1528–1536. 3366906.
6. Pedersen O, Bak JF, Andersen PH, et al. Evidence against altered expression of GLUT1 or GLUT4 in skeletal muscle of patients with obesity or NIDDM. Diabetes 1990;39:865–870. 2354749.
7. Dohm GL, Elton CW, Friedman JE, et al. Decreased expression of glucose transporter in muscle from insulin-resistant patients. Am J Physiol 1991;260(3 Pt 1):E459–E463. 2003599.
8. Kim YB, Nikoulina SE, Ciaraldi TP, Henry RR, Kahn BB. Normal insulin-dependent activation of Akt/protein kinase B, with diminished activation of phosphoinositide 3-kinase, in muscle in type 2 diabetes. J Clin Invest 1999;104:733–741. 10491408.
9. White MF. The IRS-signaling system: a network of docking proteins that mediate insulin and cytokine action. Recent Prog Horm Res 1998;53:119–138. 9769706.
10. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol 2006;7:85–96. 16493415.
11. Sale EM, Sale GJ. Protein kinase B: signalling roles and therapeutic targeting. Cell Mol Life Sci 2008;65:113–127. 17952368.
12. Farese RV, Sajan MP, Standaert ML. Insulin-sensitive protein kinases (atypical protein kinase C and protein kinase B/Akt): actions and defects in obesity and type II diabetes. Exp Biol Med (Maywood) 2005;230:593–605. 16179727.
13. Sano H, Kane S, Sano E, et al. Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J Biol Chem 2003;278:14599–14602. 12637568.
14. Bandyopadhyay G, Sajan MP, Kanoh Y, et al. PKC-zeta mediates insulin effects on glucose transport in cultured preadipocyte-derived human adipocytes. J Clin Endocrinol Metab 2002;87:716–723. 11836310.
15. Farese RV, Sajan MP, Yang H, et al. Muscle-specific knockout of PKC-lambda impairs glucose transport and induces metabolic and diabetic syndromes. J Clin Invest 2007;117:2289–2301. 17641777.
16. Bourdeau A, Dubé N, Tremblay ML. Cytoplasmic protein tyrosine phosphatases, regulation and function: the roles of PTP1B and TC-PTP. Curr Opin Cell Biol 2005;17:203–209. 15780598.
17. Harley EA, Levens N. Protein tyrosine phosphatase 1B inhibitors for the treatment of type 2 diabetes and obesity: recent advances. Curr Opin Investig Drugs 2003;4:1179–1189.
18. Vinciguerra M, Foti M. PTEN and SHIP2 phosphoinositide phosphatases as negative regulators of insulin signalling. Arch Physiol Biochem 2006;112:89–104. 16931451.
19. Sasaoka T, Wada T, Tsuneki H. Lipid phosphatases as a possible therapeutic target in cases of type 2 diabetes and obesity. Pharmacol Ther 2006;112:799–809. 16842857.
20. Cheatham B, Vlahos CJ, Cheatham L, et al. Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp70 S6 kinase, DNA synthesis, and glucose transporter translocation. Mol Cell Biol 1994;14:4902–4911. 8007986.
21. Le Marchand-Brustel Y, Gautier N, Cormont M, Van Obberghen E. Wortmannin inhibits the action of insulin but not that of okadaic acid in skeletal muscle: comparison with fat cells. Endocrinology 1995;136:3564–3570. 7628394.
22. Björnholm M, Kawano Y, Lehtihet M, Zierath JR. Insulin receptor substrate-1 phosphorylation and phosphatidylinositol 3-kinase activity in skeletal muscle from NIDDM subjects after in vivo insulin stimulation. Diabetes 1997;46:524–527. 9032113.
23. Konishi H, Shinomura T, Kuroda S, Ono Y, Kikkawa U. Molecular cloning of rat RAC protein kinase alpha and beta and their association with protein kinase C zeta. Biochem Biophys Res Commun 1994;205:817–825. 7999118.
24. Konishi H, Kuroda S, Tanaka M, et al. Molecular cloning and characterization of a new member of the RAC protein kinase family: association of the pleckstrin homology domain of three types of RAC protein kinase with protein kinase C subspecies and beta gamma subunits of G proteins. Biochem Biophys Res Commun 1995;216:526–534. 7488143.
25. Kim YB, Peroni OD, Franke TF, Kahn BB. Divergent regulation of Akt1 and Akt2 isoforms in insulin target tissues of obese Zucker rats. Diabetes 2000;49:847–856. 10905496.
26. Walker KS, Deak M, Paterson A, et al. Activation of protein kinase B beta and gamma isoforms by insulin in vivo and by 3-phosphoinositide-dependent protein kinase-1 in vitro: comparison with protein kinase B alpha. Biochem J 1998;331:299–308. 9512493.
27. Jiang ZY, Zhou QL, Coleman KA, et al. Insulin signaling through Akt/protein kinase B analyzed by small interfering RNA-mediated gene silencing. Proc Natl Acad Sci U S A 2003;100:7569–7574. 12808134.
28. Zabolotny JM, Haj FG, Kim YB, et al. Transgenic overexpression of protein-tyrosine phosphatase 1B in muscle causes insulin resistance, but overexpression with leukocyte antigen-related phosphatase does not additively impair insulin action. J Biol Chem 2004;279:24844–24851. 15031294.
29. Zabolotny JM, Kim YB, Peroni OD, et al. Overexpression of the LAR (leukocyte antigen-related) protein-tyrosine phosphatase in muscle causes insulin resistance. Proc Natl Acad Sci U S A 2001;98:5187–5192. 11309481.
30. Kim YB, Zhu JS, Zierath JR, et al. Glucosamine infusion in rats rapidly impairs insulin stimulation of phosphoinositide 3-kinase but does not alter activation of Akt/protein kinase B in skeletal muscle. Diabetes 1999;48:310–320. 10334307.
31. Krook A, Roth RA, Jiang XJ, Zierath JR, Wallberg-Henriksson H. Insulin-stimulated Akt kinase activity is reduced in skeletal muscle from NIDDM subjects. Diabetes 1998;47:1281–1286. 9703329.
32. Rondinone CM, Carvalho E, Wesslau C, Smith UP. Impaired glucose transport and protein kinase B activation by insulin, but not okadaic acid, in adipocytes from subjects with Type II diabetes mellitus. Diabetologia 1999;42:819–825. 10440123.
33. Cho H, Thorvaldsen JL, Chu Q, Feng F, Birnbaum MJ. Akt1/PKBalpha is required for normal growth but dispensable for maintenance of glucose homeostasis in mice. J Biol Chem 2001;276:38349–38352. 11533044.
34. Cho H, Mu J, Kim JK, et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB beta). Science 2001;292:1728–1731. 11387480.
35. Ceresa BP, Kao AW, Santeler SR, Pessin JE. Inhibition of clathrin-mediated endocytosis selectively attenuates specific insulin receptor signal transduction pathways. Mol Cell Biol 1998;18:3862–3870. 9632770.
36. Kane S, Sano H, Liu SC, et al. A method to identify serine kinase substrates. Akt phosphorylates a novel adipocyte protein with a Rab GTPase-activating protein (GAP) domain. J Biol Chem 2002;277:22115–22118. 11994271.
37. Nagase T, Ishikawa K, Miyajima N, et al. Prediction of the coding sequences of unidentified human genes. IX. The complete sequences of 100 new cDNA clones from brain which can code for large proteins in vitro. DNA Res 1998;5:31–39. 9628581.
38. Morris NJ, Ross SA, Lane WS, et al. Sortilin is the major 110-kDa protein in GLUT4 vesicles from adipocytes. J Biol Chem 1998;273:3582–3587. 9452485.
39. Simpson F, Whitehead JP, James DE. GLUT4--at the cross roads between membrane trafficking and signal transduction. Traffic 2001;2:2–11. 11208163.
40. Bruss MD, Arias EB, Lienhard GE, Cartee GD. Increased phosphorylation of Akt substrate of 160 kDa (AS160) in rat skeletal muscle in response to insulin or contractile activity. Diabetes 2005;54:41–50. 15616009.
41. Deshmukh A, Coffey VG, Zhong Z, et al. Exercise-induced phosphorylation of the novel Akt substrates AS160 and filamin A in human skeletal muscle. Diabetes 2006;55:1776–1782. 16731842.
42. Geraghty KM, Chen S, Harthill JE, et al. Regulation of multisite phosphorylation and 14-3-3 binding of AS160 in response to IGF-1, EGF, PMA and AICAR. Biochem J 2007;407:231–241. 17617058.
43. Sriwijitkamol A, Coletta DK, Wajcberg E, et al. Effect of acute exercise on AMPK signaling in skeletal muscle of subjects with type 2 diabetes: a time-course and dose-response study. Diabetes 2007;56:836–848. 17327455.
44. Treebak JT, Glund S, Deshmukh A, et al. AMPK-mediated AS160 phosphorylation in skeletal muscle is dependent on AMPK catalytic and regulatory subunits. Diabetes 2006;55:2051–2058. 16804075.
45. Hojlund K, Glintborg D, Andersen NR, et al. Impaired insulin-stimulated phosphorylation of Akt and AS160 in skeletal muscle of women with polycystic ovary syndrome is reversed by pioglitazone treatment. Diabetes 2008;57:357–366. 17977950.
46. Karlsson HK, Zierath JR, Kane S, et al. Insulin-stimulated phosphorylation of the Akt substrate AS160 is impaired in skeletal muscle of type 2 diabetic subjects. Diabetes 2005;54:1692–1697. 15919790.
47. Kotani K, Ogawa W, Matsumoto M, et al. Requirement of atypical protein kinase c lambda for insulin stimulation of glucose uptake but not for Akt activation in 3T3-L1 adipocytes. Mol Cell Biol 1998;18:6971–6982. 9819385.
48. Bandyopadhyay G, Kanoh Y, Sajan MP, Standaert ML, Farese RV. Effects of adenoviral gene transfer of wild-type, constitutively active, and kinase-defective protein kinase C-lambda on insulin-stimulated glucose transport in L6 myotubes. Endocrinology 2000;141:4120–4127. 11089544.
49. Bandyopadhyay G, Standaert ML, Galloway L, Moscat J, Farese RV. Evidence for involvement of protein kinase C (PKC)-zeta and noninvolvement of diacylglycerol-sensitive PKCs in insulin-stimulated glucose transport in L6 myotubes. Endocrinology 1997;138:4721–4731. 9348199.
50. Bandyopadhyay D, Kusari A, Kenner KA, et al. Protein-tyrosine phosphatase 1B complexes with the insulin receptor in vivo and is tyrosine-phosphorylated in the presence of insulin. J Biol Chem 1997;272:1639–1645. 8999839.
51. Etgen GJ, Valasek KM, Broderick CL, Miller AR. In vivo adenoviral delivery of recombinant human protein kinase C-zeta stimulates glucose transport activity in rat skeletal muscle. J Biol Chem 1999;274:22139–22142. 10428775.
52. Bandyopadhyay G, Standaert ML, Sajan MP, et al. Dependence of insulin-stimulated glucose transporter 4 translocation on 3-phosphoinositide-dependent protein kinase-1 and its target threonine-410 in the activation loop of protein kinase C-zeta. Mol Endocrinol 1999;13:1766–1772. 10517677.
53. Kanoh Y, Bandyopadhyay G, Sajan MP, Standaert ML, Farese RV. Thiazolidinedione treatment enhances insulin effects on protein kinase C-zeta/lambda activation and glucose transport in adipocytes of nondiabetic and Goto-Kakizaki type II diabetic rats. J Biol Chem 2000;275:16690–16696. 10749857.
54. Kanoh Y, Bandyopadhyay G, Sajan MP, Standaert ML, Farese RV. Rosiglitazone, insulin treatment, and fasting correct defective activation of protein kinase C-zeta/lambda by insulin in vastus lateralis muscles and adipocytes of diabetic rats. Endocrinology 2001;142:1595–1605. 11250941.
55. Vollenweider P, Ménard B, Nicod P. Insulin resistance, defective insulin receptor substrate 2-associated phosphatidylinositol-3' kinase activation, and impaired atypical protein kinase C (zeta/lambda) activation in myotubes from obese patients with impaired glucose tolerance. Diabetes 2002;51:1052–1059. 11916925.
56. Standaert ML, Ortmeyer HK, Sajan MP, et al. Skeletal muscle insulin resistance in obesity-associated type 2 diabetes in monkeys is linked to a defect in insulin activation of protein kinase C-zeta/lambda/iota. Diabetes 2002;51:2936–2943. 12351430.
57. Mudaliar S, Henry RR. New oral therapies for type 2 diabetes mellitus: The glitazones or insulin sensitizers. Annu Rev Med 2001;52:239–257. 11160777.
58. Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature 2000;405:421–424. 10839530.
59. Inzucchi SE, Maggs DG, Spollett GR, et al. Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. N Engl J Med 1998;338:867–872. 9516221.
60. Kim YB, Ciaraldi TP, Kong A, et al. Troglitazone but not metformin restores insulin-stimulated phosphoinositide 3-kinase activity and increases p110beta protein levels in skeletal muscle of type 2 diabetic subjects. Diabetes 2002;51:443–448. 11812753.
61. Storgaard H, Song XM, Jensen CB, et al. Insulin signal transduction in skeletal muscle from glucose-intolerant relatives of type 2 diabetic patients [corrected]. Diabetes 2001;50:2770–2778. 11723060.
62. Karlsson HK, Hällsten K, Björnholm M, et al. Effects of metformin and rosiglitazone treatment on insulin signaling and glucose uptake in patients with newly diagnosed type 2 diabetes: a randomized controlled study. Diabetes 2005;54:1459–1467. 15855334.
63. Reginato MJ, Lazar MA. Mechanisms by which thiazolidinediones enhance insulin action. Trends Endocrinol Metab 1999;10:9–13. 10322388.
64. Kintscher U, Law RE. PPARgamma-mediated insulin sensitization: the importance of fat versus muscle. Am J Physiol Endocrinol Metab 2005;288:E287–E291. 15637349.
65. Roden M, Price TB, Perseghin G, et al. Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest 1996;97:2859–2865. 8675698.
66. Antonucci T, Whitcomb R, McLain R, Lockwood D, Norris RM. Impaired glucose tolerance is normalized by treatment with the thiazolidinedione troglitazone. Diabetes Care 1997;20:188–193. 9118772.
67. Petersen KF, Krssak M, Inzucchi S, et al. Mechanism of troglitazone action in type 2 diabetes. Diabetes 2000;49:827–831. 10905493.
68. Schmitz-Peiffer C, Oakes ND, Browne CL, Kraegen EW, Biden TJ. Reversal of chronic alterations of skeletal muscle protein kinase C from fat-fed rats by BRL-49653. Am J Physiol 1997;273(5 Pt 1):E915–E921. 9374677.
69. Lewis RE, Volle DJ, Sanderson SD. Phorbol ester stimulates phosphorylation on serine 1327 of the human insulin receptor. J Biol Chem 1994;269:26259–26266. 7929343.
70. De Fea K, Roth RA. Protein kinase C modulation of insulin receptor substrate-1 tyrosine phosphorylation requires serine 612. Biochemistry 1997;36:12939–12947. 9335553.
71. Bailey CJ, Turner RC. Metformin. N Engl J Med 1996;334:574–579. 8569826.
72. Stumvoll M, Nurjhan N, Perriello G, Dailey G, Gerich JE. Metabolic effects of metformin in non-insulin-dependent diabetes mellitus. N Engl J Med 1995;333:550–554. 7623903.
73. Dunn CJ, Peters DH. Metformin. A review of its pharmacological properties and therapeutic use in non-insulin-dependent diabetes mellitus. Drugs 1995;49:721–749. 7601013.
74. Matthaei S, Hamann A, Klein HH, et al. Association of Metformin's effect to increase insulin-stimulated glucose transport with potentiation of insulin-induced translocation of glucose transporters from intracellular pool to plasma membrane in rat adipocytes. Diabetes 1991;40:850–857. 1647995.
75. Fantus IG, Brosseau R. Mechanism of action of metformin: insulin receptor and postreceptor effects in vitro and in vivo. J Clin Endocrinol Metab 1986;63:898–905. 3745404.
76. Pryor PR, Liu SC, Clark AE, et al. Chronic insulin effects on insulin signalling and GLUT4 endocytosis are reversed by metformin. Biochem J 2000;348(Pt 1):83–91. 10794717.
77. Jazet IM, Schaart G, Gastaldelli A, et al. Loss of 50% of excess weight using a very low energy diet improves insulin-stimulated glucose disposal and skeletal muscle insulin signalling in obese insulin-treated type 2 diabetic patients. Diabetologia 2008;51:309–319. 18080107.
78. Kim YB, Kotani K, Ciaraldi TP, Henry RR, Kahn BB. Insulin-stimulated protein kinase C lambda/zeta activity is reduced in skeletal muscle of humans with obesity and type 2 diabetes: reversal with weight reduction. Diabetes 2003;52:1935–1942. 12882908.
79. Beeson M, Sajan MP, Dizon M, et al. Activation of protein kinase C-zeta by insulin and phosphatidylinositol-3,4,5-(PO4)3 is defective in muscle in type 2 diabetes and impaired glucose tolerance: amelioration by rosiglitazone and exercise. Diabetes 2003;52:1926–1934. 12882907.
80. Ryder JW, Yang J, Galuska D, et al. Use of a novel impermeable biotinylated photolabeling reagent to assess insulin- and hypoxia-stimulated cell surface GLUT4 content in skeletal muscle from type 2 diabetic patients. Diabetes 2000;49:647–654. 10871204.
81. Friedman JE, Dohm GL, Leggett-Frazier N, et al. Restoration of insulin responsiveness in skeletal muscle of morbidly obese patients after weight loss. Effect on muscle glucose transport and glucose transporter GLUT4. J Clin Invest 1992;89:701–705. 1737857.
82. Goldstein BJ, Ahmad F, Ding W, Li PM, Zhang WR. Regulation of the insulin signalling pathway by cellular protein-tyrosine phosphatases. Mol Cell Biochem 1998;182:91–99. 9609118.
83. Ahmad F, Considine RV, Goldstein BJ. Increased abundance of the receptor-type protein-tyrosine phosphatase LAR accounts for the elevated insulin receptor dephosphorylating activity in adipose tissue of obese human subjects. J Clin Invest 1995;95:2806–2812. 7769120.
84. Ahmad F, Goldstein BJ. Increased abundance of specific skeletal muscle protein-tyrosine phosphatases in a genetic model of insulin-resistant obesity and diabetes mellitus. Metabolism 1995;44:1175–1184. 7666792.
85. Ahmad F, Goldstein BJ. Functional association between the insulin receptor and the transmembrane protein-tyrosine phosphatase LAR in intact cells. J Biol Chem 1997;272:448–457. 8995282.
86. Ahmad F, Goldstein BJ. Effect of tumor necrosis factor-alpha on the phosphorylation of tyrosine kinase receptors is associated with dynamic alterations in specific proteintyrosine phosphatases. J Cell Biochem 1997;64:117–127. 9015760.
87. McGuire MC, Fields RM, Nyomba BL, et al. Abnormal regulation of protein tyrosine phosphatase activities in skeletal muscle of insulin-resistant humans. Diabetes 1991;40:939–942. 1647997.
88. Ahmad F, Azevedo JL, Cortright R, Dohm GL, Goldstein BJ. Alterations in skeletal muscle protein-tyrosine phosphatase activity and expression in insulin-resistant human obesity and diabetes. J Clin Invest 1997;100:449–458. 9218523.
89. Zabolotny JM, Kim YB, Welsh LA, et al. Protein-tyrosine phosphatase 1B expression is induced by inflammation in vivo. J Biol Chem 2008;283:14230–14241. 18281274.
90. Klaman LD, Boss O, Peroni OD, et al. Increased energy expenditure, decreased adiposity, and tissue-specific insulin sensitivity in protein-tyrosine phosphatase 1B-deficient mice. Mol Cell Biol 2000;20:5479–5489. 10891488.
91. Elchebly M, Payette P, Michaliszyn E, et al. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Science 1999;283:1544–1548. 10066179.
92. Delibegovic M, Zimmer D, Kauffman C, et al. Liver-specific deletion of protein-tyrosine phosphatase 1B (PTP1B) improves metabolic syndrome and attenuates diet-induced endoplasmic reticulum stress. Diabetes 2009;58:590–599. 19074988.
93. Haj FG, Zabolotny JM, Kim YB, Kahn BB, Neel BG. Liver-specific protein-tyrosine phosphatase 1B (PTP1B) re-expression alters glucose homeostasis of PTP1B-/-mice. J Biol Chem 2005;280:15038–15046. 15699041.
94. Rondinone CM, Trevillyan JM, Clampit J, et al. Protein tyrosine phosphatase 1B reduction regulates adiposity and expression of genes involved in lipogenesis. Diabetes 2002;51:2405–2411. 12145151.
95. Zinker BA, Rondinone CM, Trevillyan JM, et al. PTP1B antisense oligonucleotide lowers PTP1B protein, normalizes blood glucose, and improves insulin sensitivity in diabetic mice. Proc Natl Acad Sci U S A 2002;99:11357–11362. 12169659.
96. Fukuda S, Ohta T, Sakata S, et al. Pharmacological profiles of a novel protein tyrosine phosphatase 1B inhibitor, JTT-551. Diabetes Obes Metab 2010;12:299–306. 20380650.
97. Matsui T, Amano M, Yamamoto T, et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. EMBO J 1996;15:2208–2216. 8641286.
98. Nakagawa O, Fujisawa K, Ishizaki T, et al. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett 1996;392:189–193. 8772201.
99. Ishizaki T, Maekawa M, Fujisawa K, Lim L. The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. EMBO J 1996;15:1885–1893. 8617235.
100. Leung T, Manser E, Tan L, et al. A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to peripheral membranes. J Biol Chem 1995;270:29051–29054. 7493923.
101. Farah S, Agazie Y, Ohan N, Ngsee JK, Liu XJ. A rho-associated protein kinase, ROKalpha, binds insulin receptor substrate-1 and modulates insulin signaling. J Biol Chem 1998;273:4740–4746. 9468537.
102. Begum N, Sandu OA, Ito M, Lohmann SM, Smolenski A. Active Rho kinase (ROK-alpha) associates with insulin receptor substrate-1 and inhibits insulin signaling in vascular smooth muscle cells. J Biol Chem 2002;277:6214–6222. 11739394.
103. Furukawa N, Ongusaha P, Jahng WJ, et al. Role of Rho-kinase in regulation of insulin action and glucose homeostasis. Cell Metab 2005;2:119–129. 16098829.
104. Zick Y. Insulin resistance: a phosphorylation-based uncoupling of insulin signaling. Trends Cell Biol 2001;11:437–441. 11684411.
105. Lee DH, Shi J, Jeoung NH, et al. Targeted disruption of ROCK1 causes insulin resistance in vivo. J Biol Chem 2009;284:11776–11780. 19276091.

Article information Continued

Figure 1

The insulin signaling pathway. PTP1B, protein-tyrosine phosphatase 1B; IRS, insulin receptor substrate; ROCK, Rho-kinase; PIP, phosphatidylinositol phosphate; PTEN, phosphatase and tension homologue deleted on chromosome 10; PH domain, pleckstrin homology domain; PDK, phosphoinositide-dependent protein kinase; GβL, G-protein beta subunit like; mTOR, mammalian target of rapamycin; AS160, 160 kDa Akt substrate; PKCλ/ζ, protein kinase C λ and ζ; Glut4, glucose transporter 4.