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 265 毫秒
1.
该文探讨了乳腺癌细胞中表皮生长因子(EGF)介导的MEK非依赖性ERK激活通路。Western blot检测EGF刺激下,siRNA抑制MEK1/2后的T47D细胞的p-ERK水平,以验证T47D细胞中存在EGF介导的MEK非依赖性ERK激活的通路。接着使用可能参与MEK非依赖性ERK激活的激酶的小分子抑制剂抑制相关激酶(AC、PKC、Src、PI3K、PDK1和Akt)活性后,检测T47D细胞EGF介导ERK的磷酸化水平。siRNA抑制MEK1/2表达后,T47D细胞在EGF刺激后的仍保留部分p-ERK,即在T47D细胞中,存在EGF介导的MEK非依赖性的ERK磷酸化通路。小分子抑制剂抑制AC、PKC、Src对MEK非依赖性ERK激活途径影响不大。而使用小分子抑制剂抑制PI3K、PDK1和Akt后,ERK的磷酸化水平显著降低,提示PI3K/Akt通路下游的激酶参与T47D中EGF介导的MEK非依赖性ERK激活途径。siRNA干扰PI3K/Akt通路下游PBK/TOPK后并使用U0126抑制MEK功能后,几乎检测不到p-ERK,提示PBK/TOPK参与T47D细胞中EGF介导的MEK非依赖性ERK激活途径。乳腺癌抗雌激素药物耐药株T47D细胞存在EGF介导的MEK非依赖性ERK激活途径,且该途径受PI3K/Akt下游的PBK/TOPK调控。  相似文献   

2.
基于细胞Raf/MEK/ERK信号通路与病毒复制的关系,应用Western印迹检测 p-ERK1/2蛋白的表达、用终点滴定法测定病毒增殖量(TCID50),以及观察感染细胞的细胞病变效应(CPE)等,揭示单纯疱疹病毒Ⅱ型(HSV-2)复制与 ERK通路的关系. 结果表明,HSV-2的复制可引起细胞ERK通路的活化;用U0126预先抑制ERK通路的活化,或用特异性siRNA敲减MEK1/2基因的表达可显著地抑制病毒复制.提示ERK信号通路以及MEK1/2蛋白对HSV-2的复制具有重要的作用.该研究对进一步阐明细胞ERK通路各激酶蛋白在病毒复制中的作用机制、寻找抗病毒作用靶标等奠定了良好的基础.  相似文献   

3.
目的:用低血清培养液来模拟肾脏供血不足的营养不良状态,研究低浓度哇巴因对低血清培养下OK细胞(负鼠肾小管上皮细胞)增殖的影响。方法:用低浓度哇巴因(1-30n M)处理0.2%血清培养下OK细胞,MTT实验和Brdu掺入法检测哇巴因对OK细胞增殖的影响;Western blot检测Akt和ERK1/2的磷酸化水平;用LY294002和PD98059分别抑制PI3K/Akt和ERK1/2蛋白激酶活性,观察抑制PI3K/Akt和ERK1/2对哇巴因促进OK细胞增殖的影响。结果:低浓度哇巴因(1-30n M)促进OK细胞的增值,上调OK细胞中Akt和ERK1/2磷酸化水平。用LY294002和PD98059特异抑制Akt和ERK1/2的活化能够抑制哇巴因的促增殖作用。结论:低浓度哇巴因(1-10n M)能够促进OK细胞的增值,PI3K/Akt和ERK1/2信号通路参与哇巴因对OK细胞促增殖作用的调节。  相似文献   

4.
探讨MEK/ERK1/2信号通路在Cyclosporin A(CsA)诱导滋养细胞表达titin中的作用。应用RT-PCR、Western blot检测CsA诱导的滋养细胞titin的表达水平,Western blot检测CsA作用于滋养细胞后ERK1/2的活化程度,并观察MEK特异性抑制剂U0126对其mRNA转录的影响。发现CsA以时间和剂量依赖方式诱导titin表达,并刺激滋养细胞ERK1/2的活化,U0126以剂量依赖方式抑制CsA诱导的titin表达。结果表明CsA通过活化MEK/ERK1/2信号通路诱导滋养细胞titin 的表达,改变其生物学行为,从而有利于胚胎着床及早期发育。  相似文献   

5.
为了探讨在人永生化支气管上皮细胞BEP2D细胞中,Smad4分子对 ERK/MAPK通路的作用,我们用RNA干扰的方法分别设计了两对Smad4 siRNA,并使BEP2D细胞中Smad4靶向沉默,用Western印迹分析了细胞内ERK激酶和MEK激酶磷酸化水平的变化.结果发现,当Smad4表达沉默后,ERK激酶磷酸化水平未变,MEK激酶磷酸化水平有所降低;再加TGF-β1诱导后ERK激酶和MEK激酶磷酸化水平均显著降低至基础水平以下.结果表明在BEP2D细胞中,Smad4的缺失抑制TGF-β1对ERK/MAPK通路的活化,故提出TGF β活化ERK/MAPK通路需要Smad4存在的假设.  相似文献   

6.
为了探讨雌激素对发育期大鼠海马NMDA受体活性的快速影响,对出生后18d的雄性大鼠进行苯甲酸雌二醇皮下注射,1h后用WesternBlot检测海马NMDA受体NR1和NR2B亚基、雌激素β受体、ERK1/2蛋白的表达,以及NR2B和ERK1/2的磷酸化水平;并通过海马内给予雌激素受体拮抗剂ICI182,780和MEK1/2抑制剂U0126预处理,进一步分析苯甲酸雌二醇影响NR2B和ERK1/2磷酸化的作用机制。结果显示,苯甲酸雌二醇不影响NR1、NR2B、ERβ和ERK1/2的表达,但能快速增强NR2B和ERK1/2的磷酸化水平。雌激素受体拮抗剂ICI182,780和MEK1/2抑制剂U0126均能明显抑制苯甲酸雌二醇诱导的NR2B和ERK1/2磷酸化水平的增加。以上结果提示,雌激素可能通过雌激素受体的非基因组机制激活ERK/MAPK信号转导通路,快速诱导NMDA受体NR2B亚基磷酸化,激活NMDA受体。  相似文献   

7.
摘要 目的:分析富含半胱氨酸的酸性分泌蛋白类似蛋白1(SPARCL1)对非小细胞肺癌(NSCLC)细胞增殖、凋亡、侵袭的影响,并探讨分裂原活化抑制剂(MEK)/细胞外调节蛋白激酶(ERK)通路在其中发挥的作用。方法:收集2019年9月~2021年6月期间本院接受手术治疗的84例NSCLC患者癌组织与相应癌旁组织,实时定量逆转录聚合酶链反应(qRT-PCR)法测定并比较各组织以及正常肺上皮细胞HBEpiC、NSCLC细胞A549、HCC827、H1299、H292中SPARCL1 信使RNA(mRNA)表达水平,选取A549、HCC827培养并分组,分为对照组、NC siRNA组、SPARCL1 siRNA组、U0126组(MEK/ERK特异性抑制剂)、SPARCL1 siRNA加U0126组,细胞计数法(CCK8)以及平板克隆法测定A549、HCC827细胞增殖,流式细胞仪测定A549、HCC827细胞凋亡,Transwell小室法测定A549、HCC827细胞侵袭能力,蛋白质印迹法(western blot)检测SPARCL1、p-MEK、MEK、p-ERK1/2、ERK1/2蛋白表达。结果:SPARCL1在NSCLC组织中mRNA表达水平低于癌旁组织(P<0.05);与HBEpiC细胞相比,NSCLC细胞A549、HCC827、H1299、H292细胞中SPARCL1 mRNA表达水平降低(P<0.05);与对照组相比,SPARCL1 siRNA组A549、HCC827细胞SPARCL1 mRNA表达水平与蛋白表达、凋亡率降低(P<0.05),OD450、克隆形成数、侵袭细胞数、p-MEK/MEK、p-ERK1/2/ERK1/2蛋白表达升高(P<0.05),U0126组A549、HCC827细胞SPARCL1 mRNA表达水平与蛋白表达、凋亡率升高(P<0.05),OD450、克隆形成数、侵袭细胞数、p-MEK/MEK、p-ERK1/2/ERK1/2蛋白表达降低(P<0.05);与SPARCL1 siRNA组相比,SPARCL1 siRNA加U0126组A549、HCC827细胞SPARCL1 mRNA表达水平与蛋白表达、凋亡率升高(P<0.05),OD450、克隆形成数、侵袭细胞数、p-MEK/MEK、p-ERK1/2/ERK1/2蛋白表达降低(P<0.05)。结论:SPARCL1可能通过调控MEK/ERK通路影响NSCLC A549、HCC827细胞增殖、侵袭与凋亡。  相似文献   

8.
目的:研究κ-阿片受体(κ-OR)激动剂U50488H在高浓度葡萄糖(25.5mmol/L)诱导的心肌细胞肥大中的作用及可能的信号转导通路。方法:以原代培养的新生大鼠心肌细胞为模型,应用25.5mmol/L的高浓度葡萄糖诱导心肌肥大,用Lowry法检测心肌细胞蛋白含量;用消化分离法及计算机图像分析系统检测心肌细胞体积;用Western蛋白印迹法测定细胞外信号调节激酶(ERK)磷酸化水平。结果:25.5mmol/L的高浓度葡萄糖使心肌细胞蛋白含量和体积明显增加,1μmol/L的U50488H能抑制高糖诱导的心肌肥大,使ERK磷酸化水平降低,与10μmol/L的ERK抑制剂U0126对心肌肥大的抑制程度相近,统计结果没有显著性差异。结论:U50488H抑制高糖诱导的心肌肥大与ERK信号有关。  相似文献   

9.
目的在细胞学水平明确IFN-α和胞外信号调节激酶(extracellular signal-regulated kinase, ERK)通路抑制剂U0126联合用药对肠道病毒A71(EV-A71)感染的作用及其可能机制。方法利用病毒致细胞病变效应、病毒终点滴定实验以及Western blot,确定IFN-α和U0126联合用药对EV-A71抗病毒效果,对细胞干扰素(interferon, IFN)受体及其下游信号通路重要蛋白水平、ERK通路活性的影响。结果 IFN-α和U0126联合用药能有效发挥抗EV-A71增殖的作用(P0.01),同时也能有效抑制ERK通路磷酸化活性、阻断EV-A71 2A~(pro)介导的I型干扰素受体1(interferon alpha receptor 1, IFNAR1)表达水平下调(P0.001),并上调IFN信号通路重要分子eIF2α磷酸化(P0.001)。此外,利用ERK抑制剂(U0126和sorafenib)或特异性siRNA分别阻断ERK磷酸化活性后,可显著阻断肠道病毒2A~(pro)介导的eIF4GI切割和IFNAR1表达下调的作用,同时受染细胞EV-A71复制也显著下降。结论 IFN-α和U0126联合用药可通过有效地抑制ERK通路,抑制2A~(pro)依赖的切割eIF4GI和下调IFNAR1表达的作用,使得外源IFN-α能更有效与细胞膜上IFNAR结合,有效激活IFN抗病毒信号通路,从而发挥IFN抗EV-A71蛋白翻译及增殖作用。  相似文献   

10.
近来发现新的蛋白激酶C δ亚型(PKCδ)的磷酸化激活与帕金森病(PD)中神经元的缺失有关.我们的前期研究发现, PKCδ 磷酸化参与6-羟基多巴胺引起的多巴胺能细胞的毒性作用,但其具体机制尚不清楚.本研究以TUNEL检测发现,6-羟基多巴胺引起较显著的细胞凋亡,PKC δ 抑制剂 Rottlerin 可减轻6-羟基多巴胺诱导的凋亡,总PKC抑制剂Bis、钙依赖性PKC(α和β)抑制剂Go6976对6-羟基多巴胺诱导的凋亡无影响.采用Western免疫印迹杂交实验发现,6-羟基多巴胺持续性激活 ERK 1/2 和PKC δ, Rottlerin既可抑制PKCδ的磷酸化激活,又可抑制ERK的磷酸化激活,而 MEK 抑制剂 U0126 仅能抑制 ERK 1/2 磷酸化激活,对 PKC δ 磷酸化却无显著影响.这说明 PKCδ 是6-羟基多巴胺持续性激活 ERK 1/2 的上游激酶.本研究结果提示,在凋亡过程中PKCδ仍然是ERK1/2激活的上游激酶,阻断PKCδ磷酸化可阻断ERK1/2持续激活.Rottlerin正是由于阻断PKCδ的激活,进一步阻断ERK1/2持续激活,减轻多巴胺能细胞的凋亡.因此, Rottlerin 可能对防治帕金森病患者神经元缺失有一定作用.  相似文献   

11.
1,7-Bis(4-hydroxyphenyl)-1,4-heptadien-3-one (EB30) is a diarylheptanoid-like compound isolated from Viscum coloratum. This curcumin analog exhibits significant cytotoxic activity against HeLa, SGC-7901, and MCF-7 cells. However, little is known about the anticancer effects and mechanisms of EB30 in human lung cancer. The current study reports that EB30 significantly reduced the cell viability of A549 and NCI-H292 human lung cancer cells. Further examination revealed that EB30 not only induced cell cycle arrest and promoted the generation of reactive oxygen species (ROS) but also induced cell apoptosis through the intrinsic and extrinsic signaling pathways. Furthermore, EB30 upregulated the expression levels of p-ERK1/2 and p-P90RSK, whereas downregulating the phosphorylation of Akt and P70RSK. Cell viability was further inhibited by the combination of EB30 with LY294002 (a specific PI3K inhibitor) or U0126 (a MEK inhibitor). The current study indicates that EB30 is a potential anticancer agent that induces cell apoptosis via suppression of the PI3K/Akt pathway and activation of the ERK1/2 pathway.  相似文献   

12.
13.
Primary cortical neurones exposed to an oxidative insult in the form of hydrogen peroxide (H(2)O(2)) for 30 min showed a concentration-dependent increase in oxidative stress followed by a delayed NMDA receptor-dependent cell death measured 24 h later. Extracellular signal-regulated protein kinase (ERK1/2), c-jun N-terminal kinase (JNK) and the kinase Akt/PKB may regulate neuronal viability in response to oxidative insults. Using phospho-specific antibodies, a 15-min stimulation of neurones with H(2)O(2) (100 microm - 1 mm) produced a concentration-dependent phosphorylation of ERK1/2 and Akt/PKB that was partly dependent on extracellular Ca(2+) and phosphatidylinositol 3-kinase (PI3-K). Higher concentrations of H(2)O(2) (1 mm) also stimulated a phosphorylation of JNK which was totally dependent on extracellular Ca(2+) but not PI3-K. H(2)O(2)-induced phosphorylation of ERK1/2, Akt/PKB or JNK were unaffected by the NMDA channel blocker MK801. Blocking ERK1/2 activation with the upstream inhibitor U0126 (10 microm) enhanced H(2)O(2)-induced (100-300 microm range) neurotoxicity and inhibited H(2)O(2)-mediated phosphorylation of the cyclic AMP regulatory binding protein (CREB), suggesting that ERK1/2 signals to survival under these conditions. At higher concentrations (mm), H(2)O(2)-stimulated a phosphorylation of c-jun. It is likely, therefore, that subjecting neurones to moderate oxidative-stress recruits pro-survival signals to CREB but during severe oxidative stress pro-death signals through JNK and c-jun are dominant.  相似文献   

14.
Cerebral ischemia increases neural progenitor cell proliferation and neurogenesis. However, the precise molecular mechanism is poorly understood. The present study was undertaken to determine roles of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt and their signaling pathways in neural progenitor cells exposed to hypoxia/reoxygenation (H/R), an in vitro model of ischemia/reperfusion. Neural progenitor cells were isolated from postnatal mouse brain. ERK and Akt were transiently activated during the early phase of reoxygenation following 4-h of hypoxia. The ERK activation was inhibited by U0126, a specific inhibitor of MEK, but not by LY294002, a specific inhibitor of PI3K, whereas the Akt activation was blocked by LY294002, but not by U0126. Reoxygenation following 4-h hypoxia stimulated cell proliferation, which was dependent on ERK and Akt activation. Inhibitors of growth factor receptor (AG1478) and Src (PP2) and the antioxidant N-acetylcysteine did not affect activation of ERK and Akt, while the Ras and Raf inhibitors inhibited activation of ERK, but not Akt. PKC inhibitors inhibited both ERK and Akt activation. Taken together, these results suggest that H/R induces activation of MEK/ERK and PI3K/Akt survival signaling pathways through a PKC-dependent mechanism. These pathways may be responsible for the repair process during ischemia/reperfusion.  相似文献   

15.
Proteinase-activated receptor-2 (PAR2) plays pro-inflammatory roles in many organs including the gastrointestinal (GI) tract. To clarify the downstream pro-inflammatory signaling of PAR2 in the GI tract, we examined interleukin-8 (IL-8) release and the underlying cellular signaling following PAR2 stimulation in human colorectal cancer-derived HCT-15 cells and human gastric adenocarcinoma-derived MKN-45 cells. A PAR2-activating peptide, but not a PAR2-inactive scrambled peptide or a PAR1- activating peptide, caused IL-8 release in these GI epithelial cells. The PAR2-triggered IL-8 release was suppressed by inhibitors of MEK (U0126) or PI3-kinase (LY294002), and PAR2 stimulation indeed activated the downstream kinases, ERK and Akt. U0126 blocked the phosphorylation of ERK, but not Akt, and LY294002 blocked the phosphorylation of Akt, but not ERK. Together, PAR2 triggers IL-8 release via two independent signaling pathways, MEK/ERK and PI3-kinase/Akt, suggesting a role of PAR2 as a pro-inflammatory receptor in the GI tract.  相似文献   

16.
In Jurkat T lymphocytes, hydrogen peroxide (H(2)O(2)) potentiates the phosphorylation level of extracellular signal-regulated kinase 1 and 2 (ERK1/2) caused by T cell receptor (TCR) stimulation with anti-CD3 and anti-CD28 or anti-CD3 alone. Submillimolar concentrations of H(2)O(2)-induced phosphorylation of ERK1/2 and MAP/ERK kinase 1 and 2 (MEK1/2) without antigenic stimulation. H(2)O(2) also induced the electrophoretic mobility shift of Lck from 56 to 60 kDa. The MEK inhibitor, PD98059 attenuated ERK1/2 and MEK1/2 phosphorylation, as well as the migration shift of Lck induced by H(2)O(2). The phospholipase C (PLC) inhibitor, U73122, and EGTA reduced the phosphorylation of both ERK1/2 and MEK1/2 induced by H(2)O(2). Interestingly, an increase of intracellular cAMP level with forskolin or 8-(4-chlorophenylthio)-cAMP augmented ERK1/2 phosphorylation by H(2)O(2), while inhibiting MEK1/2 phosphorylation by H(2)O(2). These results demonstrate an alternative pathway that results in augmentation of ERK1/2 phosphorylation without concomitant MEK1/2 phosphorylation in T cells.  相似文献   

17.
We have previously shown the importance of LTB4 in human pancreatic cancer. LTB4 receptor antagonists block growth and induce apoptosis in pancreatic cancer cells both in vitro and in vivo. Therefore, we investigated the effect of LTB4 on proliferation of human pancreatic cancer cells and the mechanisms involved. LTB4 stimulated DNA synthesis and proliferation of both PANC-1 and AsPC-1 human pancreatic cancer cells, as measured by thymidine incorporation and cell number. LTB4 stimulated rapid and transient activation of MEK and ERK1/2 kinases. The MEK inhibitors, PD98059 and U0126, blocked LTB4-stimulated ERK1/2 activation and cell proliferation. LTB4 also stimulated phosphorylation of p38 MAPK; however, the p38 MAPK inhibitor, SB203580, failed to block LTB4-stimulated growth. The activity of JNK/SAPK was not affected by LTB4 treatment. Phosphorylation of Akt was also induced by LTB4 and this effect was blocked by the PI-3 kinase inhibitor wortmannin, which also partially blocked LTB4-stimulated cell proliferation. In conclusion, LTB4 stimulates proliferation of human pancreatic cancer cells through MEK/ERK and PI-3 kinase/Akt pathways, while p38 MPAK and JNK/SAPK are not involved.  相似文献   

18.
The MEK1-ERK1/2 signaling pathway has been implicated in the regulation of renal epithelial cell proliferation, epithelial-to-mesenchymal transition and the induction of an invasive cell phenotype. Much less information is available about the MEK5-ERK5 module and its role in renal epithelial cell proliferation and differentiation. In the present study we have investigated the regulation of these two families of extracellular signal-regulated kinases in epidermal growth factor (EGF)-stimulated human kidney-2 (HK-2) cells and a possible interaction between ERK1/2 and ERK5. Here we report that 5 ng/ml EGF led to a strong stimulation of HK-2 cell proliferation, which was largely U0126-sensitive. Both synthetic MEK1/2 inhibitors U0126 and Cl-1040, when used at 10 and 1 microM, respectively, inhibited basal and EGF-induced ERK1/2 phosphorylation but not ERK5 phosphorylation. Long-term inhibition of MEK1/2-ERK1/2 signaling and/or vanadate-sensitive protein phosphatases enhanced and prolonged EGF-induced ERK5 phosphorylation, while transient expression of an adenoviral constitutively active MEK1 (Ad-caMEK1) construct completely blocked EGF-induced ERK5 phosphorylation. Expression of Ad-caMEK1 in HK-2 cells resulted in the upregulation of the dual-specificity phosphatases MKP-3/DUSP6, MKP-1/DUSP1, and DUSP5. The EGF-mediated time-dependent induction of MKP-3, MKP-1 and DUSP5 mRNA levels was U0126-sensitive at a concentration, which blocked EGF-mediated ERK1/2 phosphorylation but not ERK5 phosphorylation. Furthermore, U0126 inhibited EGF-induced MKP-3 and MKP-1 protein expression. Both MKP-3 and MKP-1 co-immunoprecipitated with ERK5 in unstimulated as well as in EGF-stimulated HK-2 cells. These results suggest the existence of an ERK1/2-driven negative feed-back regulation of ERK5 signaling in EGF-stimulated HK-2 cells, which is mediated by MKP-3, DUSP5 and/or MKP-1.  相似文献   

19.
Avocado (Persea americana) fruits are consumed as part of the human diet and extracts have shown growth inhibitory effects in various types of human cancer cells, although the effectiveness of individual components and their underlying mechanism are poorly understood. Using activity-guided fractionation of the flesh of avocado fruits, a chloroform-soluble extract (D003) was identified that exhibited high efficacy towards premalignant and malignant human oral cancer cell lines. From this extract, two aliphatic acetogenins of previously known structure were isolated, compounds 1 [(2S,4S)-2,4-dihydroxyheptadec-16-enyl acetate] and 2 [(2S,4S)-2,4-dihydroxyheptadec-16-ynyl acetate]. In this study, we show for the first time that the growth inhibitory efficacy of this chloroform extract is due to blocking the phosphorylation of EGFR (Tyr1173), c-RAF (Ser338), and ERK1/2 (Thr202/Tyr204) in the EGFR/RAS/RAF/MEK/ERK1/2 cancer pathway. Compounds 1 and 2 both inhibited phosphorylation of c-RAF (Ser338) and ERK1/2 (Thr202/Tyr204). Compound 2, but not compound 1, prevented EGF-induced activation of the EGFR (Tyr1173). When compounds 1 and 2 were combined they synergistically inhibited c-RAF (Ser338) and ERK1/2 (Thr202/Tyr204) phosphorylation, and human oral cancer cell proliferation. The present data suggest that the potential anticancer activity of avocado fruits is due to a combination of specific aliphatic acetogenins that target two key components of the EGFR/RAS/RAF/MEK/ERK1/2 cancer pathway.  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