首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 140 毫秒
1.
目的 体外以缺氧无血清条件模拟心肌梗死后的心脏缺血微环境,研究洛伐他汀能否抑制缺氧无血清引起的骨髓间充质干细胞(MSC)凋亡并探讨其机制.方法 以Hocchst33342染色荧光显微镜观察法及Annexin V/PI流式细胞术检测洛伐他汀的抗凋亡作用,并进一步采用Westernblot方法 检测洛伐他汀对线粒体凋亡途径的抑制作用以及对磷脂酰肌醇3激酶(PI3K)/丝氨酸苏氨酸激酶(Akt)途径和丝裂原活化的蛋白激酶(MAPK)的激酶(MEK)/细胞内信号调节蛋白激酶(ERK1/2)途径的激活作用.结果 0.01~1 μmol/L浓度范围的洛伐他汀能够有效地抑制缺氧无血清引起的MSC凋亡.洛伐他汀抑制线粒体凋亡途径,洛伐他汀抑制细胞色素C释放,降低天冬氨酸特异性半胱氨酸蛋白酶-3(caspase-3)活化,从而保护线粒体功能.洛伐他汀的抗凋亡效应以及其抑制细胞色素C释放的作用均可被PI3K抑制剂LY294002和MEK抑制剂U0126阻断.洛伐他汀能够激活PI3K/Akt和MEK/ERK1/2两条细胞存活信号通路,分别导致Akt和GSK-3β及ERK1/2磷酸化.结论 洛伐他汀能够抑制线粒体凋亡途径,并激活PI3K/Akt和MEK/ERK1/2细胞存活通路,最终发挥抗缺氧无血清引起的MSC凋亡.该研究为提高移植干细胞的存活率提供了一种可能有效的干预措施.  相似文献   

2.
目的观察趋化因子受体4(CXCR4)通过激活PI3K/Akt和Erk信号通路在卵巢癌SKOV3细胞上皮细胞-间充质转化(EMT)中的作用。方法 Boyden小室检测激活CXCR4后SKOV3细胞迁移和侵袭能力的变化情况。Western印迹方法检测激活CXCR4后SKOV3细胞中EMT相关蛋白的表达情况及PI3K/Akt和Erk信号通路的活化情况。PI3K/Akt和Erk信号通路抑制剂预处理SKOV3细胞后,采用Western印迹方法检测SKOV3细胞中EMT相关蛋白的表达情况。结果同对照组相比,激活CXCR4后可促进SKOV3细胞的迁移和侵袭能力(P0.01),下调SKOV3细胞中与EMT相关的E-钙黏蛋白表达,并上调EMT相关波形蛋白的表达。Western印迹结果显示随着CXCR4刺激时间的延长,SKOV3细胞中PI3K/Akt蛋白和Erk蛋白磷酸化水平逐渐升高。采用PI3K/Akt信号通路抑制剂LY294002和Erk信号通路抑制剂U0126预处理SKOV3细胞后,CXCR4介导的E-钙黏蛋白表达下调和波形蛋白表达上调被阻断。结论 CXCR4通过激活SKOV3细胞中的PI3K/Akt信号通路和Erk信号通路,下调SKOV3细胞中E-钙黏蛋白表达,并上调波形蛋白的表达,从而促进卵巢癌SKOV3细胞EMT的发生。  相似文献   

3.
目的通过建立人脐静脉内皮细胞(HUVEC)内质网应激(ERS)的细胞模型,研究雌激素抑制内质网应激引起的凋亡的信号传导机制,以探讨雌激素对心血管的保护机制。方法分别用10μmol/L的衣霉素(TM)或2 mmol/L的二硫苏糖醇(DTT)诱导HUVEC,建立内质网应激细胞模型,提前给予10-8mol/L的17-β雌二醇(E2)预处理1 h,用Western blot检测葡萄糖调节蛋白78(GRP78)判断模型是否建立成功,并探索E2对内质网应激的作用。检测内质网应激的三条主要信号通路蛋白的变化,上调最显著的为内质网应激最主要的信号通路。Western blot检测内质网应激凋亡蛋白C/EBP-同源蛋白(CHOP),Hochest染色检测细胞凋亡率,探索E2对内质网应激凋亡的作用。添加E2受体拮抗剂ICI182780(ERα、ERβ拮抗剂及GPER激动剂)和G15(GPER拮抗剂)后检测内质网应激最主要通路蛋白表达量的变化,探索雌激素受体在其抑制内质网应激中的作用。添加E2受体后信号通路阻断剂,检测雌激素抑制内质网应激的过程中活化其受体后激活的最主要受体后信号通路。结果 TM/DTT组GRP78的表达量显著上调,内质网应激三条信号通路中蛋白激酶R样内质网激酶(PERK)信号通路上调最明显,而TM/DTT+E2组上调显著回复。TM/DTT组CHOP的表达量显著上调且细胞凋亡率显著增加,而TM/DTT+E2组上调明显回复,凋亡细胞减少。E2有显著抑制p-PERK/PERK上调的作用,而E2的保护作用可分别被ICI182780和G15阻断,同时添加ICI182780和G15时阻断作用最显著。分别添加信号通路阻断剂后,E2抑制pPERK/PERK上调的作用均减弱,其中以磷脂酰肌醇-3羟基激酶(PI3K)通路阻断剂的作用最显著。结论 E2可抑制TM/DTT诱导的HUVEC内质网应激。p-PERK/PERK通路可能为TM/DTT诱导的HUVEC内质网应激最主要的信号通路。E2可抑制过度内质网应激引起的细胞凋亡。E2受体在E2抑制内质网应激凋亡的作用中起重要作用。E2受体激活包括PI3K-蛋白激酶B(PKB/Akt)、细胞外信号调节激酶1/2(ERK1/2)、c-Jun氨基末端激酶(JNK)和p38-丝裂原活化蛋白激酶(p38-MAPK)在内的信号通路快速起到抑制内质网应激的作用,其中PI3K-Akt通路可能为最主要的通路。雌激素通过抑制PERK信号通路引起的内质网应激凋亡,保护血管内皮细胞,其抑制内质网应激的机制主要为活化的雌激素受体激活PI3K/Akt通路。  相似文献   

4.
磷脂酰肌醇-3-激酶/蛋白激酶B/雷帕霉素靶蛋白(PI3K/Akt/mTOR)信号通路介导许多在肿瘤和自身免疫病发生中至关重要的细胞生物学过程,对于细胞增殖、细胞活化、血管生成起到重要作用.PI3 K/Akt/mTOR信号通路作为药物治疗靶点进行抗细胞增殖活化的研究近年来发展迅速.本文从PI3K/Akt/mTOR信号通路的上下游、信号通路抑制剂以及与信号通路相关的自身免疫病等方面作一综述.  相似文献   

5.
花靖棋  沈华  邓君鹏 《山东医药》2023,(36):107-111
磷脂酰肌醇3-激酶/蛋白激酶B/哺乳动物雷帕霉素靶蛋白(PI3K/Akt/mTOR)信号通路是细胞内重要的信号转导通路之一,可以调节细胞生长、分化、迁移、存活、血管生成和代谢,并参与多种恶性肿瘤的发生发展。肾癌(RCC)作为泌尿系统常见的恶性肿瘤,有较高的转移率,且对放化疗具有明显的耐受性,随着肿瘤分子生物学的发展,肾癌靶向生物治疗逐渐成为研究的热点。PI3K/Akt/mTOR信号通路抑制剂对RCC的疗效显著,以该信号通路为靶点的RCC药物主要为PI3K抑制剂,Akt抑制剂和mTOR抑制剂,这些药物可通过抑制信号通路的异常激活,阻断肿瘤细胞的增殖和生长,诱导细胞凋亡,并抑制肿瘤的侵袭和血管生成。而PI3K、mTOR双重抑制剂虽然处于临床试验阶段,但是其在RCC治疗的研究中表现出更可靠的前景性。  相似文献   

6.
综述磷脂酰肌醇3-激酶(PI3K)/蛋白激酶B(Akt)信号通路的构成、转导途径及其在高血压及相关靶器官损害中的作用机制,并阐述基于PI3K/Akt信号通路高血压的中医药治疗进展,旨在为高血压的临床诊疗提供新的思路和治疗靶点。  相似文献   

7.
PI3K/Akt通路是由磷脂酰肌醇3-激酶(phosphatidylinositol 3-kinase,PI3K)始动的生物信号转导通路,在细胞增殖、细胞周期调控、凋亡启动、血管生成等方面发挥着关键作用.此外,PI3K/Akt通路还与中枢神经系统损伤的保护机制密切相关.深入研究PI3K/Akt、下游分子及其调控机制可能为脑损伤的治疗提供新的思路和方法.  相似文献   

8.
目前,大多数肿瘤组织的恶性表型与特殊的物质代谢和能量代谢密切相关.近期研究表明,脂肪酸合酶( fatty acid synthase,FASN)介导的代谢异常在肿瘤发生、发展过程中具有重要作用[1].同时,磷脂酰肌醇3-激酶(phosphoinositide 3-kinase,PI3K)/蛋白激酶B(Protein kinase B,Akt)信号通路也与细胞代谢密切相关.因此,本研究将一种新型的PI3K抑制剂ZSTK474作用于结肠癌细胞株Caco-2,以探讨PI3K/Akt信号通路调控Caco-2细胞FASN表达的作用及其对Caco-2细胞增殖的影响.  相似文献   

9.
肝星状细胞(hepatic stellate cells,HSC)是肝纤维化的主要细胞,是细胞外基质(extracellular matrix,ECM)的重要来源.烟酰胺腺嘌呤二核苷酸磷酸氧化酶(NADPH oxidase,NOX)产生活性氧(reactive oxygen species,ROS),调控HSCs内信号转导,在肝纤维化发病中起关键作用.NOX产生的ROS可介导丝裂原活化蛋白激酶(mitogen-activated protein kinase,MAPK)和磷脂酰肌醇-3激酶/蛋白激酶(phosphatidylin-ositol-3 kinase/Akt,PI3K/Akt)信号通路激活,促进HSC增殖、抑制其凋亡,导致肝纤维化形成.抑制NOX产生ROS,阻断相应的信号通路可诱导HSC凋亡.因此,探索出以NOX为作用靶点的抗纤维化药物意义重大.  相似文献   

10.
β-Arrestins是G蛋白耦联受体信号转导通路的负调节因子,越来越多的证据表明,β-arrestins也能作用于细胞内的多种信号分子,调节胰岛素/胰岛素样生长因子-1(IGF-1)信号转导通路.在胰岛素的刺激下,β-arrestin 2能够募集蛋白激酶B(Akt)和酪氨酸激酶Src到胰岛素受体,从而调节胰岛素介导的糖代谢效应;而β-arrestin 1则与胰岛素受体底物-1(IRS-1)竞争性结合泛素连接酶Mdm2,从而减少IRS-1的泛素化和降解,促进磷脂酰肌醇3激酶(PI3K)通路的信号转导.在IGF-1介导的信号转导通路中,β-arrestin 1结合并介导了IGF-1受体(IGF-1R)的内吞,促进胞外信号调节激酶活化,正性调节丝裂原活化蛋白激酶通路.此外,β-arrestin 1与IGF-1R相耦联后,能越过信号分子IRS-1而激活PI3K,进而活化Akt,表现出对P13K途径的正性调控作用.  相似文献   

11.
12.
Growth factors are known to favor both proliferation and survival of hepatocytes. In this work, we investigated the role of 2 main signaling pathways, phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK), in these processes. First, evidence was provided that the PI3K cascade as well as the MEK/ERK cascade is a key transduction pathway controlling hepatocyte proliferation, as ascertained by arrest of DNA synthesis in the presence of LY294002, a specific PI3K inhibitor. Inhibition of FRAP/mTOR by rapamycin also abrogated DNA replication and protein synthesis induced by growth factor. We showed that expression of cyclin D1 at messenger RNA (mRNA) and protein levels was regulated by this pathway. We highlighted that 4E-BP1 phosphorylation was not activated by epidermal growth factor (EGF) but was under an insulin-regulation mechanism through a PI3K-FRAP/mTOR activation that could account for the permissive role of insulin on hepatocyte proliferation. No interference between the MEK/ERK pathway and 4E-BP1 phosphorylation was detected, whereas p70S6K phosphorylation induced by EGF was under a U0126-sensitive regulation. Last, we established that the antiapoptotic function of EGF was dependent on MEK, whereas LY294002 and rapamycin had no direct effect on cell survival. Taken together, these data highlight the regulation and the role of 2 pathways that mediate growth-related response by acting onto distinct steps. In conclusion, hepatocyte progression in late G1 phase induced by EGF generates survival signals depending on MEK activation, whereas PI3K and MEK/ERK cascades are both necessary for hepatocyte replication.  相似文献   

13.
Chemoresistance in hepatocellular carcinoma (HCC) is associated with multiple cellular responses to environmental stresses, such as nutrient deprivation and hypoxia. Nevertheless, whether ER stress resulting from nutrient deprivation and tumor hypoxia contributes to drug resistance remains unclear. Melatonin increased the efficacy of chemotherapeutic drugs in hepatocellular carcinoma in our previous studies. However, the effects of melatonin on endoplasmic reticulum (ER) stress‐induced resistance to chemotherapeutic agents in HCC have not been tested. The effect of the endoplasmic reticulum (ER) stress response during resistance of human hepatocellular carcinoma cells against doxorubicin was investigated in this study. Pretreatment of HepG2 and SMMC‐7721 cells (two human hepatocellular carcinoma cell lines) with tunicamycin, an ER stress inducer, drastically decreased the rate of apoptosis generated by doxorubicin. Interestingly, co‐pretreatment with tunicamycin and melatonin significantly increased apoptosis induced by doxorubicin. Simultaneously, the expression of phosphorylated AKT (p‐AKT) was elevated in HepG2 and SMMC‐7721 cells given tunicamycin but reduced in the presence of melatonin. Furthermore, consistent with inhibition of AKT activation by using the PI3K inhibitor LY294002, melatonin elevated the levels of CHOP (C/EBP‐homologous protein) and reduced the levels of Survivin (a member of the inhibitor of apoptosis protein family)suggesting that inhibition of the PI3K/AKT pathway by melatonin‐reversed ER stress‐induced resistance to doxorubicin in human hepatocellular carcinoma cells. These results demonstrate that melatonin attenuates ER stress‐induced resistance to doxorubicin in human hepatocellular carcinoma cells by down‐regulating the PI3K/AKT pathway, increasing the levels of CHOP and decreasing the levels of Survivin.  相似文献   

14.
In this paper the signal transduction pathways evoked by bradykinin (BK) in MCF-7 breast cancer cells were investigated. BK activation of the B(2) receptor provoked: (a) the phosphorylation of the extracellular signal-regulated kinases 1 and 2 (ERK1/2); (b) the translocation from the cytosol to the membrane of the conventional protein kinase C-alpha (PKC-alpha) and novel PKC-delta and PKC-epsilon; (c) the phosphorylation of protein kinase B (PKB/ Akt); (d) the proliferation of MCF-7 cells. The BK-induced ERK1/2 phosphorylation was completely blocked by PD98059 (an inhibitor of the mitogen-activated protein kinase kinase (MAPKK or MEK)) and by LY294002 (an inhibitor of phosphoinositide 3-kinase (PI3K)), and was reduced by GF109203X (an inhibitor of both novel and conventional PKCs); G?6976, a conventional PKCs inhibitor, did not have any effect. The BK-induced phosphorylation of PKB/Akt was blocked by LY294002 but not by PD98059. Furthermore, LY294002 inhibited the BK-provoked translocation of PKC-delta and PKC-epsilon suggesting that PI3K may be upstream to PKCs. Finally, the proliferative effects of BK were blocked by PD98059, GF109203X and LY294002. These observations demonstrate that BK acts as a proliferative agent in MCF-7 cells activating intracellular pathways involving novel PKC-delta/-epsilon, PKB/Akt and ERK1/2.  相似文献   

15.
Purpose  Ewing sarcoma cells, of which over 85% retain chimeric fusion gene EWS/Fli-1, are by and large more resistant to chemotherapeutics compared to nonneoplastic cells. The purpose of this study is to determine the role of EWS/Fli-1 fusion and its downstream targets regarding the cells’ resistance against actinomycin D (ActD), which is one of the most commonly used antitumor agents in combination chemotherapy of Ewing sarcomas. Methods  Cytotoxicity was measured by WST-8 assay. Caspase-dependent and -independent cell death was examined by fluorescence microscope. Protein expression was analyzed by western blotting. Caspase activity was determined by Caspase-Glo assay. Results  ActD-induced caspase-dependent apoptotic cell death to Ewing sarcoma TC-135 cells in a dose- and time- dependent manner. Knockdown of EWS/Fli-1 fusion by siRNA resulted in enhancement of ActD-induced apoptosis. ActD treatment activated both mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase (PI3K)/Akt pathways although in a distinctive manner. Combined administration of U0126 (MEK inhibitor) and LY294002 (PI3K inhibitor) significantly enhanced ActD-induced apoptosis in vitro and suppressed xenograft tumor growth in vivo. Conclusions  The present study demonstrated for the first time that combination of U0126 and LY294002 can augment the cytotoxicity of ActD against Ewing sarcoma cells in vitro and in vivo. Our results indicate that further study on combination of conventional chemotherapies with MEK and PI3K inhibitors may be considered for innovative treatments of Ewing sarcoma patients.  相似文献   

16.
AIM:To evaluate the expression of C-X-C motif chemokine receptor 4(CXCR4)and its signaling cascades,which were previously identified as a key factor for cancer cell progression and metastasis,in cholangiocarcinoma cell lines.METHODS:The expression of CXCR4 and its signaling cascades were determined in the cholangiocarcinoma cell lines(RMCCA1 and KKU100)by Western blotting.The invasion assays and the detection of actin polymerization were tested in these cholangiocarcinoma cells treated with CXC chemokine ligand-12(CXCL12).RESULTS:Expression of CXCR4 was detected in both cholangiocarcinoma cell lines and activation of CXCR4 with CXCL12 triggered the signaling via the extracellular signal-regulated kinase-1/2(ERK1/2)and phosphoinositide 3-kinase(PI3K)and induction of cholangiocarcinoma cell invasion,and displayed high levels of actin polymerization.Addition of CXCR4 inhibitor(AMD3100)abrogated CXCL12-induced phosphorylation of MEK1/2 and Akt in these cells.Moreover,treatment with MEK1/2 inhibitor(U0126)or PI3K inhibitor(LY294 002)also attenuated the effect of CXCL12-induced cholangiocarcinoma cell invasion.CONCLUSION:These results indicated that the activation of CXCR4 and its signaling pathways(MEK1/2 and Akt)are essential for CXCL12-induced cholangiocarcinoma cell invasion.This rises Implications on a potential role for the inhibition of CXCR4 or its signal cascades in the treatment of cholangiocarcinoma.  相似文献   

17.
Zhang J  Lodish HF 《Blood》2004,104(6):1679-1687
Oncogenic mutations in ras genes frequently occur in patients with myeloid disorders, and in these patients erythropoiesis is often affected. Previously, we showed that expression of oncogenic H-ras in purified mouse primary fetal liver erythroid progenitors blocks terminal erythroid differentiation and supports erythropoietin (Epo)-independent proliferation. As a first step in understanding the underlying molecular mechanisms we examined the signaling pathways downstream of Ras in primary erythroid cells. We found that 3 major pathways are abnormally activated by oncogenic H-ras: Raf/ERK (extracellular signal-regulated kinase), phosphatidyl inositol 3 (PI3)-kinase/Akt, and RalGEF/RalA. However, only constitutive activation of the MEK (MAPK [mitogen-activated protein kinase]/ERK kinase)/ERK pathway alone could recapitulate all of the effects of oncogenic H-ras expression in blocking erythroid differentiation and inducing Epo-independent proliferation. Although expression of a constitutively active Akt kinase (ca.Akt) in erythroid progenitors does not significantly affect erythroid differentiation in the presence of Epo, coexpression of ca.Akt together with a constitutively active MEK causes prolonged Epo-independent proliferation of erythroid progenitors in addition to a block in differentiation. Moreover, the effects of oncogenic H-ras expression on primary erythroid cells are blocked by the addition of U0126, a specific inhibitor of MEK1 and MEK2, allowing normal terminal erythroid proliferation and differentiation. Our data suggest that the interruption of constitutive MEK/ERK signaling is a potential therapeutic strategy to correct impaired erythroid differentiation in patients with myeloid disorders.  相似文献   

18.
Liang L  Jiang J  Frank SJ 《Endocrinology》2000,141(9):3328-3336
Interaction of GH with the cell-surface GH receptor (GHR) causes activation of the GHR-associated tyrosine kinase, JAK2, and consequent triggering of signaling cascades including the STAT, Ras/Raf/MEK1/MAP kinase, and insulin receptor substrate-1(IRS-1)/PI3kinase pathways. We previously showed that IRS- and GHR-deficient 32D cells that stably express the rabbit GHR and rat IRS-1 (32D-rbGHR-IRS-1) exhibited markedly enhanced GH-induced proliferation and MAP kinase (ERK1 and ERK2) activation compared with cells expressing only the GHR (32D-rbGHR). We now examine biochemical mechanism(s) by which IRS-1 augments GH-induced MAP kinase activation. Time-course experiments revealed a similarly transient (maximal at 15 min) GH-induced ERK1 and ERK2 activation in both 32D-rbGHR and 32D-rbGHR-IRS-1 cells, but, consistent with our prior findings, substantially greater activation was seen in the IRS-1-containing cells. In both cells, GH-induced MAP kinase activation was markedly blunted by the MEK1 inhibitor, PD98059, but not by the PKC inhibitor, GF109203X. Interestingly, pretreatment with the PI3K inhibitor, wortmannin (EC50 approximately 10 nM), significantly reduced GH-induced MAP kinase activation in both 32D-rbGHR and 32D-rbGHR-IRS-1 cells. This same pattern in both cells of IRS-1-dependent augmentation and IRS-1-independent wortmannin sensitivity was also observed for GH-induced activation of Akt and MEK1 (using state-specific antibody blotting for both), despite the lack of difference in GHR, JAK2, SHP-2, p85, Akt, Ras, Raf-1, MEK1, ERK1, or ERK2 abundance between the two cells. A different PI3K inhibitor, LY294002 (50 microM), substantially inhibited (roughly 72%) GH-induced MAP kinase activation in 32D-rbGHR-IRS-1 cells, but only marginally (and statistically insignificantly) inhibited GH-induced MAP kinase activation in 32D-rbGHR cells. Because GH-induced Akt activation was completely inhibited in both cells by the same concentration of LY294002, these findings indicate that the wortmannin sensitivity of both the IRS-1-independent and -dependent GH-induced MAP kinase activation may reflect the activity of another wortmannin-sensitive target(s) in addition to PI3K in mediation of GH-induced MAP kinase activation in these cells. Notably, GH-induced STAT5 tyrosine phosphorylation, unlike Akt or MAPK activation, did not differ between the cells. Finally, while GH promoted accumulation of activated Ras in both cells, both basal and GH-induced activated Ras levels were greater in cells expressing IRS-1 than in 32D-rbGHR cells. These data indicate that while GH induces tyrosine phosphorylation of STAT5 and activation of the Ras/Raf/MEK1/MAPK and PI3K pathways, IRS-1 expression augments the latter two more than the former.  相似文献   

19.
Hepatocyte growth factor (HGF) has recently been the focus of attention due to its angiogenic effects, which are similar to those of vascular endothelial growth factor (VEGF); because of these effects, HGF is considered to be a novel therapeutic agent against vascular disorders, including atherosclerotic angiopathies. Although nitric oxide (NO), which is derived from vascular endothelial cells (ECs), is also involved in angiogenesis, little is known regarding the interactions between HGF and NO. We therefore examined the effects of HGF on NO production as well as endothelial NO synthase (eNOS) phosphorylation, and investigated their mechanisms. In bovine aortic ECs, HGF induced a rapid (5 min) increase of NO production measured by diaminofluorescein-2 diacetate. Moreover, HGF rapidly (2.5 min) stimulated eNOS phosphorylation (Ser-1179) as determined by Western immunoblot analyses. Both of these effects were almost completely suppressed by the phosphoinositide 3-kinase (PI3K) inhibitor LY294002, and were partially suppressed by the mitogen-activated protein kinase (MAPK) kinase 1/2 inhibitor U0126. HGF also stimulated Akt phosphorylation (Ser-473), which was completely suppressed by LY294002 and was partially suppressed by U0126. Moreover, HGF stimulated extracellular signal-regulated kinase 1/2 phosphorylation (Thr-202/Tyr-204), which was completely suppressed by U0126 and was partially suppressed by LY294002. Taken together, these results indicate that HGF not only phosphorylates eNOS through the PI3K/Akt pathway, but also partially through the MAPK pathway, and that these two pathways may interact. Compared with VEGF, HGF was more potent in both NO production and eNOS phosphorylation. Our study thus demonstrates a novel activity of HGF-the stimulation of NO production-which occurs via eNOS phosphorylation that may in turn be mediated by cross-talk between the PI3K/Akt and MAPK pathways.  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