首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 263 毫秒
1.
Diabetes mellitus is a major risk factor in the development of atherosclerosis and cardiovascular disease conditions, involving intimal injury and enhanced vascular smooth muscle cell (VSMC) migration. We report a mechanistic basis for divergences between insulin's inhibitory effects on migration of aortic VSMC from control Wistar Kyoto (WKY) rats versus Goto-Kakizaki (GK) diabetic rats. In normal WKY VSMC, insulin increased MAPK phosphatase-1 (MKP-1) expression as well as MKP-1 phosphorylation, which stabilizes it, and inhibited PDGF-mediated MAPK phosphorylation and cell migration. In contrast, basal migration was elevated in GK diabetic VSMCs, and all of insulin's effects on MKP-1 expression and phosphorylation, MAPK phosphorylation, and PDGF-stimulated migration were markedly inhibited. The critical importance of MKP-1 in insulin inhibition of VSMC migration was evident from several observations. MKP-1 small interfering RNA inhibited MKP-1 expression and abolished insulin inhibition of PDGF-induced VSMC migration. Conversely, adenoviral expression of MKP-1 decreased MAPK phosphorylation and basal migration rate and restored insulin's ability to inhibit PDGF-directed migration in GK diabetic VSMCs. Also, the proteasomal inhibitors lactacystin and MG132 partially restored MKP-1 protein levels in GK diabetic VSMCs and inhibited their migration. Furthermore, GK diabetic aortic VSMCs had reduced cGMP-dependent protein kinase Ialpha (cGK Ialpha) levels as well as insulin-dependent, but not sodium nitroprusside-dependent, stimulation of cGMP. Adenoviral expression of cGK Ialpha enhanced MKP-1 inhibition of MAPK phosphorylation and VSMC migration. We conclude that enhanced VSMC migration in GK diabetic rats is due at least in part to a failure of insulin-stimulated cGMP/cGK Ialpha signaling, MKP-1 expression, and stabilization and thus MAPK inactivation.  相似文献   

2.
In this study, we examined the roleof insulin in the control of vascular smooth muscle cell (VSMC)migration in the normal vasculature. Platelet-derived growth factor(PDGF) increased VSMC migration, which was inhibited by pretreatmentwith insulin in a dose-dependent manner. Insulin also caused a 60%decrease in PDGF-stimulated mitogen-activated protein kinase (MAPK)phosphorylation and activation. Insulin inhibition of MAPK wasaccompanied by a rapid induction of MAPK phosphatase (MKP-1), whichinactivates MAPKs by dephosphorylation. Pretreatment with inhibitors ofthe nitric oxide (NO)/cGMP pathway, blocked insulin-induced MKP-1 expression and restored PDGF-stimulated MAPK activation and migration. In contrast, adenoviral infection of VSMCs with MKP-1 or cGMP-dependent protein kinase I (cGK I), the downstream effector of cGMPsignaling, blocked the activation of MAPK and prevented PDGF-directedVSMC migration. Expression of antisense MKP-1 RNA prevented insulin's inhibitory effect and restored PDGF-directed VSMC migration and MAPKphosphorylation. We conclude that insulin inhibition of VSMC migrationmay be mediated in part by NO/cGMP/cGK I induction of MKP-1 andconsequent inactivation of MAPKs.

  相似文献   

3.
The mechanisms for the effect of hyperglycemia on insulin-induced mitogenesis were investigated using rat vascular smooth muscle cells (VSMC). VSMC were preincubated in serum-free medium with low (5 mM) glucose (LG condition) or high (25 mM) glucose (HG condition), and examined for DNA synthesis using bromodeoxyuridine (BrdUrd) incorporation. Mitogen-activated protein kinase (MAPK) activity and MAPK phosphatase (MKP-1) protein expression were detected by Western blot analysis. Phosphatidylinositol 3-kinase (PI-3K) activity was detected by thin layer chromatography. Insulin induced a dose-dependent increase in BrdUrd incorporation (123.3+/-2.6% over basal level with 1 microM insulin) in the LG group and this effect was significantly enhanced (161.6+/-10.4% over basal level) in the HG group. In the LG group, MAPK activity was transient with a peak activation (137.4+/-11.2% over basal level) after 10 min exposure to 100 nM insulin. In the HG group, the MAPK activity was significantly potentiated (two-fold compared to the LG group) and was sustained even after 60 min. Insulin also induced PI-3K activity and MKP-1 expression, both of which were blocked by the PI-3K inhibitor wortmannin. In the HG group, insulin-induced PI-3K and MKP-1 expression was almost abolished. In conclusion, high glucose enhances insulin-induced mitogenesis associated with the potentiation of insulin-stimulated MAPK activity in VSMC. These effects of glucose might in part be due to the attenuation of MKP-1 expression through the blockage of the insulin-PI-3K signal pathway.  相似文献   

4.
Zhu JH  Liu Z  Huang ZY  Li S 《生理学报》2005,57(5):587-592
本文研究血管紧张素Ⅱ(angiotensin Ⅱ,Ang Ⅱ)对自发性高血压大鼠(spontaneously hypertensive rat,SHR)和Wistar- Kyoto(WKY)大鼠血管平滑肌细胞(vascular smooth muscle cells.VSMCs)细胞外信号调节激酶(extracellular signal-regulated pro- tein kinases,ERKs)信号途径的影响。体外培养SHR和WKY大鼠的VSMCs,先在培养基中加入终浓度为1×105mmol/L 的缬沙坦或1×105mmol/L的PD98059或不加药物,再给予1×107mmol/L的Ang Ⅱ刺激24 h后收集细胞,以无血清培养基 培养的VSMCs作对照。用免疫沉淀法测定ERK活性;用Western-blot方法检测总ERK(total ERK,t-ERK)、磷酸化ERK (phosphorylated-ERK,p-ERK)及丝裂素活化蛋白激酶磷酸酶-1(mitogen-activated protem kinases phosphatase-1,MKP-1)水 平;用RT-PCR法半定量测定MKP-1 mRNA的含量。结果显示:(1)SHR和WKY大鼠Ang Ⅱ刺激组VSMCs中ERK活 性、p-ERK、MKP-1及MKP-1 mRNA水平均明显高于对照组(P<0.05);SHR和WKY大鼠Ang Ⅱ+缬沙坦组和Ang Ⅱ +PD98059组的上述指标与对照组比较均无显著性差异。(2)SHR大鼠VSMCs中ERK活性、P-ERK、MKP-1及MKP-1 mRNA均显著高于相同干预的WKY大鼠(P<0.01)。(3)SHR和WKY大鼠之间以及对照组、Ang Ⅱ刺激组、Ang Ⅱ+缬沙 坦组和Ang Ⅱ+PD98059组间VSMCs中t-ERK水平均无显著性差异。以上结果表明,Ang Ⅱ可能主要通过其1型(Ang Ⅱ type 1,AT)受体激活SHR和WKY大鼠VSMCs中ERK途径,增加ERK活性和p-ERK蛋白水平,继而引起MKP-1及 MKP-1 mRNA水平升高。  相似文献   

5.
Our laboratory has recently demonstrated arole for the phosphatidylinositol 3-kinase-mediatedinducible NO synthase (iNOS) signaling pathway in acute regulation ofinsulin-induced mitogen-activated protein phosphatase-1 (MKP-1)expression in primary cultures of rat aortic vascular smooth musclecells (VSMCs) (N. Begum, L. Ragolia, M. McCarthy, and N. Duddy.J. Biol. Chem. 273: 25164-25170, 1998). We now show that prolonged treatment of VSMCs with 100 nMinsulin and high glucose (25 mM) for 12-24 h, to mimichyperinsulinemia and hyperglycemia, completely blocked MKP-1 mRNA andprotein expression in response to subsequent acute insulin treatment.To understand the mechanism of insulin resistance induced by highglucose and insulin, we studied the regulation of iNOS proteininduction in these cells. Both high glucose and chronic insulintreatment caused a marked impairment of iNOS induction in response toacute insulin. Blocking of signaling via the p38 mitogen-activatedprotein kinase (MAPK) pathway by prior treatment for 1 h withSB-203580, a synthetic p38 MAPK inhibitor, completely prevented theinhibition of iNOS induced by high glucose and insulin and restoredMKP-1 induction to levels observed with acute insulin treatment. Incontrast, PD-98059, a MEK inhibitor, had no effect. Furthermore, highglucose and chronic insulin treatment caused sustained p38 MAPKactivation. We conclude 1) thatchronic insulin and high glucose-induced insulin resistance isaccompanied by marked reductions in both iNOS and MKP-1 inductions dueto p38 MAPK activation that leads to excessive cell growth and2) that p38 MAPK/extracellularsignal-regulated kinase pathways regulate iNOS induction, therebycontrolling MKP-1 expression, which in turn inactivates MAPKs as afeedback mechanism and inhibits cell growth.

  相似文献   

6.
Aortic stiffening is an independent risk factor that underlies cardiovascular morbidity in the elderly. We have previously shown that intrinsic mechanical properties of vascular smooth muscle cells (VSMCs) play a key role in aortic stiffening in both aging and hypertension. Here, we test the hypothesis that VSMCs also contribute to aortic stiffening through their extracellular effects. Aortic stiffening was confirmed in spontaneously hypertensive rats (SHRs) vs. Wistar‐Kyoto (WKY) rats in vivo by echocardiography and ex vivo by isometric force measurements in isolated de‐endothelized aortic vessel segments. Vascular smooth muscle cells were isolated from thoracic aorta and embedded in a collagen I matrix in an in vitro 3D model to form reconstituted vessels. Reconstituted vessel segments made with SHR VSMCs were significantly stiffer than vessels made with WKY VSMCs. SHR VSMCs in the reconstituted vessels exhibited different morphologies and diminished adaptability to stretch compared to WKY VSMCs, implying dual effects on both static and dynamic stiffness. SHR VSMCs increased the synthesis of collagen and induced collagen fibril disorganization in reconstituted vessels. Mechanistically, compared to WKY VSMCs, SHR VSMCs exhibited an increase in the levels of active integrin β1‐ and bone morphogenetic protein 1 (BMP1)‐mediated proteolytic cleavage of lysyl oxidase (LOX). These VSMC‐induced alterations in the SHR were attenuated by an inhibitor of serum response factor (SRF)/myocardin. Therefore, SHR VSMCs exhibit extracellular dysregulation through modulating integrin β1 and BMP1/LOX via SRF/myocardin signaling in aortic stiffening.  相似文献   

7.
Intracellular Mg2+ depletion has been implicated in vascular dysfunction in hypertension. We demonstrated that transient receptor potential melastatin 7 (TRPM7) cation channels mediate Mg2+ influx in VSMCs. Whether this plays a role in [Mg2+]i deficiency in hypertension is unclear. Here, we tested the hypothesis that downregulation of TRPM7 and its homologue TRPM6 is associated with reduced [Mg2+]i and that ANG II negatively regulates TRPM6/7 in vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR). Cultured VSMCs from Wistar Kyoto (WKY) and SHR were studied. mRNA and protein expression of TRPM6 and TRPM7 were assessed by RT-PCR and immunoblotting, respectively. Translocation of annexin-1, specific TRPM7 substrate, was measured as an index of TRPM7 activation. [Mg2+]i was determined using mag fura-2. VSMCs from WKY and SHR express TRPM6 and TRPM7. Basal TRPM6 expression was similar in WKY and SHR, but basal TRPM7 content was lower in VSMCs from SHR vs. WKY. This was associated with significantly reduced [Mg2+]i in SHR cells (P < 0.01). ANG II time-dependently increased TRPM6 expression, with similar responses in WKY and SHR. ANG II significantly increased TRPM7 expression in WKY (P < 0.05), but not in SHR. Annexin-1 translocation was reduced 1.5-2-fold in SHR vs. WKY. Our findings demonstrate that TRPM6 and TRPM7 are differentially regulated in VSMCs from SHR and WKY. Whereas TRPM6 is unaltered in SHR, expression of TRPM7 is blunted. This was associated with attenuated annexin-1 translocation and decreased VSMC [Mg2+]i in SHR. Downregulation of TRPM7, but not TRPM6, may play a role in altered Mg2+ homeostasis in VSMCs from SHR.  相似文献   

8.
Acetylsalicylic acid (aspirin), used to reduce risk of cardiovascular disease, plays an important role in the regulation of cellular proliferation. However, mechanisms responsible for aspirin-induced growth inhibition are not fully understood. Here, we investigated whether aspirin may exert therapeutic effects via AMP-activated protein kinase (AMPK) activation in vascular smooth muscle cells (VSMC) from wistar kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Aspirin increased AMPK and acetyl-CoA carboxylase phosphorylation in a time- and dose-dependent manner in VSMCs from WKY and SHR, but with greater efficacy in SHR. In SHR, a low basal phosphorylation status of AMPK resulted in increased VSMC proliferation and aspirin-induced AMPK phosphorylation inhibited proliferation of VSMCs. Compound C, an AMPK inhibitor, and AMPK siRNA reduced the aspirin-mediated inhibition of VSMC proliferation, this effect was more pronounced in SHR than in WKY. In VSMCs from SHR, aspirin increased p53 and p21 expression and inhibited the expression of cell cycle associated proteins, such as p-Rb, cyclin D, and cyclin E. These results indicate that in SHR VSMCs aspirin exerts anti-proliferative effects through the induction of AMPK phosphorylation.  相似文献   

9.
Hyperinsulinemia is a risk factor in atherosclerosis formation that it stimulated vascular smooth muscle cells (VSMCs) proliferation and migration. To understand the underlying molecular mechanism involved in the processes of cellular response to insulin, VSMCs from Wistar-Kyoto rat (WKY) and spontaneous hypertensive rat (SHR) were isolated and cultured, and its proteome was comparatively analyzed with normal control by two-dimensional gel electrophoresis (2-DE). Results showed that the proliferation of VSMCs from SHR be more sensitive to insulin stimulation than that VSMCs from WKY. The detectable spots ranged from 537 to 608 on the gels in VSMCs of SHR, and 413 ± 31 spots in VSMCs of WKY. The different expressed protein spots in VSMCs of SHR were then isolated and measured by matrix-assisted desorption/ionization time of flight mass spectrometry (MALDI-TOF-MS). A total of 18 spots showed a sharp clear spectrum, and 13 spots matched with the known proteins from database. These proteins were mainly involved in cytoskeleton, glycometabolism, and post-translational processes. Among these proteins, OPN and matrix gla protein were up-regulated expression proteins, while α-SM actin was down-regulated. Furthermore, these preliminarily identified proteins confirmed by RT-PCR and western blotting analysis were coincident with the changes in 2-DE check. In addition, the cytoskeleton changes and migration rate of VSMCs from SHR treated by insulin increased significantly. The results showed that insulin plays a crucial role in activating proliferation and migration of VSMCs, by regulating the phenotype switch of VSMCs.  相似文献   

10.
Insulin at physiological concentrations can suppress catecholamine activation of the membrane transport of long chain fatty acids in the adipocyte. We have previously shown that the stimulatory effect of catecholamines was mediated by a beta-receptor interaction and cAMP (Abumrad, N.A., Park, C.R., and Whitesell, R. R. (1986) J. Biol. Chem. 261, 13082-13086). In this study we have investigated the mechanism of insulin action to antagonize transport activation. Fatty acid transport was stimulated using different cAMP derivatives with varying susceptibilities to hydrolysis by the cAMP-degrading enzyme phosphodiesterase. Insulin was effective in antagonizing the effect of cAMP analogs which were good substrates for the phosphodiesterase and failed to suppress the effect of those which were poorly hydrolyzed by the enzyme. Addition of increasing concentrations (1-100 microM) of the phosphodiesterase inhibitor methylisobutylxanthine (MIX) to norepinephrine (0.1 microgram/ml) gradually abolished insulin's antagonism. Insulin was completely ineffective in inhibiting stimulation by norepinephrine and 20 microM methylisobutylxanthine. Also consistent with involvement of cAMP lowering in insulin action was the finding that adenosine removal greatly diminished insulin's responsiveness. Treatment of cells with adenosine deaminase (1 unit/ml) enhanced the effect of norepinephrine by about 30%. A 10-fold higher range of insulin concentrations was then required to produce inhibition of fatty acid transport. The effect of adenosine removal was reversed by addition of phenylisopropyladenosine (500 nM), which is resistant to hydrolysis by the deaminase. Finally, exposure of insulin-treated cells (1 nM for 5 min) to dinitrophenol (1 mM for 5 min) reversed insulin action, consistent with reports of reversal of insulin's activation of the phosphodiesterase. In conclusion, our studies support the involvement of cAMP lowering in insulin's antagonism of fatty acid transport stimulation in the adipocyte.  相似文献   

11.
Wang XK  Wang Y  He ZY  Liu GY  Yang CM 《生理学报》2002,54(2):165-170
血管平滑肌细胞增殖的同时伴有细胞内肌动蛋白的改变,这种改变受PKC-MAPK信号转导途径调控,但目前机制尚不清楚。为探讨胰岛素对PKC-MAPK信号转导途径参与调控血管平滑肌细胞增殖及细胞内肌动蛋白分布的影响,本研究用PKC抑制剂预处理SHR在鼠体外培养的血管平滑肌细胞,观察预处理的血管平滑肌细胞经胰岛素刺激后细胞内DNA的合成、MAPK的活性、表达及细胞内肌动蛋白的分布。发现,胰岛素刺激后可使血管平滑肌细胞增殖,同时伴有[^3H]TdR掺入增加、MAPK活性及表达与对照组比较明显升高。这些作用可被PKC抑制剂阻断。胰岛素在刺激血管平滑肌细胞增殖的同时也使细胞内肌动蛋白重新分布,这一效应也可被PKC抑制剂阻断。 上述结果提示,胰岛素使血管平滑肌细胞增殖的效应可能与MAPK信号转导途径有关。  相似文献   

12.
We have recently shown that insulin induced myogenesis in the mouse C2C12 skeletal muscle cell line by activation of phosphatidylinositol (PI) 3-kinase/p70S6-kinase and p38-mitogen-activated protein kinase (MAPK) and downregulation of p42/p44-MAPK. This study investigated the insulin-signaling pathways involved in mitogenesis, survival, and membrane ruffling in C2C12 myoblasts, a cellular system that besides IGF-I receptors, expressed a high number of functional insulin receptors. Insulin (10 nM) rapidly stimulated beta-chain insulin receptor and IRS-1 tyrosine phosphorylation, IRS-2 being poorly and SHC not phosphorylated at all. However, an association of SHC with IRS-1 was found under insulin stimulation. Insulin stimulated IRS-1 association with p85alpha leading to the activation of PI3-kinase, and, subsequently AKT and p70S6-kinases. Moreover, both p42/p44- and p38-MAPKs resulted in phosphorylation after insulin stimulation. Insulin treatment for 24 h produced mitogenesis, as demonstrated by the increase in ((3)H)-thymidine incorporation, DNA content, the expression of PCNA and cyclin D1 proteins, and the proportion of cells in S + G2/M phases of the cell cycle. This mitogenic effect of insulin was precluded by inhibition of p70S6-kinase (either by rapamycin or by the PI3-kinase inhibitor LY294002) as well as by inhibition of p44/p42-MAPK with PD098059, but was not affected by inhibition of p38-MAPK. Serum deprivation of C2C12 myoblasts resulted in growth arrest at the GO/G1 phases of the cell cycle and apoptosis, as detected either by DNA laddering or by increase in the percentage of hypodiploid cells. Insulin rescued serum-deprived cells from apoptosis in an AKT-dependent manner, as demonstrated by the inhibition of AKT-activity by the use of LY294002 and ML-9, meanwhile neither inhibition of p70S6-kinase, nor MAPK affected insulin-induced survival. Finally, we evaluated the capacity of insulin to modulate actin cytoskeleton rearrangement. Insulin stimulation of myoblasts produced membrane ruffling and decreased actin stress fibers; this biological response being dependent of p38-MAPK, as demonstrated by the use of the p38-MAPK inhibitors SB203580 or PD169316, but independent of PI3-kinase and p42/p44-MAPK.  相似文献   

13.
14.
We have recently characterized a mutant insulin receptor (Y/F2) in which the two tyrosines in the carboxyl terminus (Tyr1316, Tyr1322) were mutated to phenylalanine. Compared with wild type receptors, the Y/F2 receptor exhibited markedly enhanced sensitivity to insulin-stimulated DNA synthesis with normal insulin-stimulated glucose uptake (Takata, Y., Webster, N. J. G., and Olefsky, J. M. (1991) J. Biol. Chem. 266, 9135-9139). In this paper, we present further evidence for the divergence of the metabolic and mitogenic signaling pathways utilized by the insulin receptor. The mutant receptor showed normal sensitivity and responsiveness for insulin-stimulated glucose incorporation into glycogen. The insulin sensitivity for phosphorylation of two substrates (pp180 and pp220) was the same in both Y/F2 cells and HIRc cells. Phosphotyrosine content, however, was greater in Y/F2 cells than in HIRc cells, especially in the basal state. Insulin stimulated S6 kinase activity 2-6-fold, with an ED50 of -10 nM in Rat 1 cells and 0.5 nM in HIRc cells. The sensitivity to insulin was enhanced in Y/F2 cells with an ED50 of 0.1 nM. These effects were insulin-specific, since insulin-like growth factor (IGF)-I-stimulated mitogenesis was normal. In summary: 1) Y/F2 receptors exhibit normal metabolic and enhanced mitogenic signaling; 2) the enhanced mitogenic signaling is specific for the insulin receptor in the Y/F2 cells, since IGF-I-stimulated mitogenesis is normal; 3) Y/F2 cells display increased endogenous substrate phosphorylation and augmented insulin-stimulated S6 kinase activity placing these responses among insulin's mitogenic effects; and 4) these results are consistent with the concept that the COOH-terminal tyrosine residues of the insulin receptor are normally inhibitory to mitogenic signaling.  相似文献   

15.
Insulin stimulates production of NO in vascular endothelium via activation of phosphatidylinositol (PI) 3-kinase, Akt, and endothelial NO synthase. We hypothesized that insulin resistance may cause imbalance between endothelial vasodilators and vasoconstrictors (e.g., NO and ET-1), leading to hypertension. Twelve-week-old male spontaneously hypertensive rats (SHR) were hypertensive and insulin resistant compared with control Wistar-Kyoto (WKY) rats (systolic blood pressure 202 +/- 11 vs. 132 +/- 10 mmHg; fasting plasma insulin 5 +/- 1 vs. 0.9 +/- 0.1 ng/ml; P < 0.001). In WKY rats, insulin stimulated dose-dependent relaxation of mesenteric arteries precontracted with norepinephrine (NE) ex vivo. This depended on intact endothelium and was blocked by genistein, wortmannin, or N(omega)-nitro-l-arginine methyl ester (inhibitors of tyrosine kinase, PI3-kinase, and NO synthases, respectively). Vasodilation in response to insulin (but not ACh) was impaired by 20% in SHR (vs. WKY, P < 0.005). Preincubation of arteries with insulin significantly reduced the contractile effect of NE by 20% in WKY but not SHR rats. In SHR, the effect of insulin to reduce NE-mediated vasoconstriction became evident when insulin pretreatment was accompanied by ET-1 receptor blockade (BQ-123, BQ-788). Similar results were observed during treatment with the MEK inhibitor PD-98059. In addition, insulin-stimulated secretion of ET-1 from primary endothelial cells was significantly reduced by pretreatment of cells with PD-98059 (but not wortmannin). We conclude that insulin resistance in SHR is accompanied by endothelial dysfunction in mesenteric vessels with impaired PI3-kinase-dependent NO production and enhanced MAPK-dependent ET-1 secretion. These results may reflect pathophysiology in other vascular beds that directly contribute to elevated peripheral vascular resistance and hypertension.  相似文献   

16.
Myocardial ischemia-reperfusion injury contributes significantly to morbidity and mortality in patients with diabetes. Insulin decreases myocardial infarct size in animals and the rate of apoptosis in cultured cells. Ischemia-reperfusion activates p38 mitogen-activated protein kinase (MAPK), which regulates cellular apoptosis. To examine whether p38 MAPK affects insulin's cardioprotection against ischemia-reperfusion injury, we studied overnight-fasted adult male rats by use of an in vivo rat model of myocardial ischemia-reperfusion. A euglycemic clamp (3 mU.min(-1).kg(-1)) was begun either 10 min before ischemia (InsulinBI), 5 min before reperfusion (InsulinBR), or 30 min after the onset of reperfusion (InsulinAR), and continued until the end of the study. Compared with saline control, insulin decreased the infarct size in both InsulinBI (P < 0.001) and InsulinBR (P < 0.02) rats but not in InsulinAR rats. The ischemic area showed markedly increased phosphorylation of p38 MAPK compared with the nonischemic area in saline animals. Acute activation of p38 MAPK with anisomycin (2 mg/kg iv 10 min before ischemia) had no effect on infarct size in saline rats. However, it completely abolished insulin's protective effect in InsulinBI and InsulinBR rats. Activation of p38 MAPK by anisomycin was associated with marked and persistent elevation in IRS-1 serine phosphorylation. Treatment of animals with SB-239063, a potent and specific inhibitor of p38 MAPK, 10 min before reperfusion enabled insulin-mediated myocardial protection in InsulinAR rats. We conclude that insulin protects myocardium against ischemia-reperfusion injury when given prior to ischemia or reperfusion, and activation of p38 MAPK abolishes insulin's cardioprotective effect.  相似文献   

17.
In this study, we examined the molecular mechanism of myosin-bound protein phosphatase (MBP) regulation by insulin and evaluated the role of MBP in insulin-mediated vasorelaxation. Insulin rapidly stimulated MBP in confluent primary vascular smooth muscle cell (VSMC) cultures. In contrast, VSMCs isolated from diabetic and hypertensive rats exhibited impaired MBP activation by insulin. Insulin-mediated MBP activation was accompanied by a rapid time-dependent reduction in the phosphorylation state of the myosin-bound regulatory subunit (MBS) of MBP. The decrease observed in MBS phosphorylation was due to insulin-induced inhibition of Rho kinase activity. Insulin also prevented a thrombin-mediated increase in Rho kinase activation and abolished the thrombin-induced increase in MBS phosphorylation and MBP inactivation. These data are consistent with the notion that insulin inactivates Rho kinase and decreases MBS phosphorylation to activate MBP in VSMCs. Furthermore, treatment with synthetic inhibitors of phosphatidylinositol-3 kinase (PI3-kinase), nitric oxide synthase (NOS), and cyclic guanosine monophosphate (cGMP) all blocked insulin's effect on MBP activation. We conclude that insulin stimulates MBP via its regulatory subunit, MBS partly by inactivating Rho kinase and stimulating NO/cGMP signaling via PI3-kinase as part of a complex signaling network that controls 20-kDa myosin light chain (MLC20) phosphorylation and VSMC contraction.  相似文献   

18.
We investigated the long-term effect of metformin treatment on blood pressure, insulin sensitivity, and vascular responses to insulin in conscious spontaneously hypertensive rats (SHR). The rats were instrumented with intravascular catheters and pulsed Doppler flow probes to measure blood pressure, heart rate, and blood flow. Insulin sensitivity was assessed by the euglycemic hyperinsulinemic clamp technique. Two groups of SHR received metformin (100 or 300 mg x kg(-1) x day(-1)) for 3 wk while another group of SHR and a group of Wistar Kyoto (WKY) rats were left untreated. We found that vasodilation of skeletal muscle and renal vasculatures by insulin is impaired in SHR. Moreover, a reduced insulin sensitivity was detected in vivo and in vitro in isolated soleus and extensor digitorum longus muscles from SHR compared with WKY rats. Three weeks of treatment with metformin improves the whole-body insulin-mediated glucose disposal in SHR but has no blood pressure-lowering effect and no influence on vascular responses to insulin (4 mU x kg(-1) x min(-1)). An improvement in insulin-mediated glucose transport activity was detected in isolated muscles from metformin-treated SHR, but in the absence of insulin no changes in basal glucose transport activity were observed. It is suggested that part of the beneficial effect of metformin on insulin resistance results from a potentiation of the hormone-stimulating effect on glucose transport in peripheral tissues (mainly skeletal muscle). The results argue against a significant antihypertensive or vascular effect of metformin in SHR.  相似文献   

19.
Electroconvulsive shock (ECS) activates MAPKs in rat brain and also induces immediate early genes. We investigated whether ECS induces MKP-1, a specific MAPK phosphatase and an immediate early gene, for feedback regulation of MAPK activity. ECS induced MKP-1 in the cortex, but MAPK activity returned to its basal level before MKP-1 protein increased, within 10 min of ECS. MKP-1 protein amount peaked 1 hr after ECS. MKP-1 induced did not lower the basal level of MAPK activity or attenuate MAPK activation by second ECS. MAPK activation in cerebellum was very weak, but the MKP-1 induction was faster and more prominent than in the cortex. These results suggest that ECS induces MKP-1 in various rat brain regions, however, the induction may not be related to the activation of MAPK and the MKP-1 induced may be independent of the regulation of MAPK activity after ECS.  相似文献   

20.
Membrane transport of long chain fatty acids in the isolated rat adipocyte can be strongly stimulated by epinephrine (Abumrad, N. A., Perry, P. R., and Whitesell, R. R. (1985) J. Biol. Chem. 260, 9969-9971). We now report that insulin at physiological concentrations can completely block or reverse the epinephrine effect. Insulin was optimally effective at a concentration of about 0.1 nM in inhibiting transport activation by 0.3 and 3 microM epinephrine (0.1 and 1.0 microgram/ml). High concentrations of insulin (above 1 nM) were generally less effective and this was particularly true at the highest dose of epinephrine (1.0 microgram/ml). The insulin effect was shown to be on the transport process since insulin inhibited epinephrine activation of transport in both directions (influx and efflux). No effect of insulin on basal transport was observed over a wide range of concentrations (0.01-10 nM). Insulin's antagonism of transport activation by epinephrine appeared dependent on ATP metabolism since it was abolished by preincubating the cells with dinitrophenol (1 mM). Dinitrophenol, however, could not reverse the insulin effect when exposure to the hormone preceded that to dinitrophenol, consistent with an action of insulin at the transport step. The data indicate that regulation of the membrane transport of fatty acids is a potential site for insulin's action to suppress lipid mobilization.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号