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 156 毫秒
1.
Dbl家族鸟苷交换因子(GEFs)是Rho家族蛋白发生恶性转化的主要调控单位,它通过使Rho蛋白从无活性的GDP形式转换为GTP形式的Rho蛋白而发挥作用,参与细胞骨架重排,细胞的生长和活力。P92GEF是一GEFs家族分子。本研究通过Real time PCR对P92GEF在人体48种正常组织中的表达情况进行了测定;GST-pulldown技术对P92GEF的体内GEF活性进行了检测;双荧光素酶报告基因检测技术对下游小分子进行转录因子活性检测;应用免疫荧光双染标记法完成了高表达P92GEF对正常细胞骨架形态的影响;在细胞表型实验中分别使用CCK8法、Transwell法及软琼脂克隆形成实验检测了高表达P92GEF对细胞增殖侵袭迁移及体外成瘤能力的影响。研究结果显示P92GEF有841个氨基酸,具有典型的Dbl家族分子结构域,在肺组织中表达量最高,能够促使正常成纤维细胞中的应力纤维(stress fiber)增多,P92GEF转染的NIH3T3细胞可以独立生长和形成继发性病灶,同时促使细胞增殖,侵袭及克隆形成能力增强,体外转录因子活性检测发现该基因可能与JAK/STAT通路有关。因此,P92GEF是一个典型的鸟苷交换因子家族分子,能激活Rho家族分子Rho A,具有明显的癌基因特征。  相似文献   

2.
对宿主细胞的侵袭是细胞内菌致病的关键步骤。细菌侵袭过程与Rho、Rac和Cdc42等Rho家族成员介导的肌动蛋白细胞骨架重排有关,涉及“拉链(zipper)”与“触发(trigger)”两种主要机制以及“拉链-触发(zipper-trigger)”双重机制。研究细菌侵袭过程中Rho家族的作用及相应细胞骨架事件,有助于揭示细菌侵袭的分子机制。  相似文献   

3.
RHO蛋白家族与细胞极性   总被引:2,自引:0,他引:2  
细胞的极性形成对细胞发育、分化及其功能的发挥起着举足轻重的作用,细胞极性的丧失与肿瘤的发生发展密切相关.小G蛋白Rho家族是肌动蛋白细胞骨架重新组装的主要调节因子之一,在协调细胞极性化和正常的形态形成过程中起重要作用.现就Rho蛋白家族与细胞极性及二者的关系作一综述.  相似文献   

4.
连环蛋白P120可在细胞连接处与E-钙黏蛋白结合形成连环蛋白-钙黏蛋白复合体,调控钙黏蛋白介导的细胞黏附作用;在胞质内可与Rho家族GTP酶相互作用调节细胞骨架的运动;在细胞核内可与核转录因子NF-κB和转录抑制因子Kaiso结合,影响炎性反应和细胞增殖.P120对细胞黏附、细胞动力、炎性反应和细胞增殖的影响使其与损伤修复和肿瘤的发生、发展密切相关.深入研究P120的作用及其相关机制对于进一步研究损伤后修复及肿瘤预防和治疗具有深远的意义.  相似文献   

5.
摘要 目的:探究长链非编码RNA LINC01006对前列腺癌(prostate cancer, PCa)细胞增殖和侵袭能力的影响。方法:体外培养人前列腺正常上皮细胞系RWPE-1,人PCa细胞系LNCaP、22Rv1、PC3、C4-2b,应用实时定量PCR(qRT-PCR)技术检测上述细胞LINC01006的表达;分别通过转染小干扰RNA(siRNA)或过表达LINC01006的慢病毒载体,在LNCaP和PC3细胞中敲减LINC01006或稳定过表达LINC01006;应用CCK8、克隆形成实验检测LINC01006对PCa细胞增殖能力的影响;应用Transwell侵袭实验检测LINC01006对PCa细胞侵袭能力的影响;通过网站预测LINC01006的转录调控因子及其结合位点。结果:相较于正常前列腺上皮细胞系RWPE-1,PCa细胞系LNCaP、22Rv1、C4-2b和PC3中LINC01006表达明显升高(P<0.05)。敲减LINC01006后的PCa细胞系LNCaP和PC3的增殖和侵袭能力被显著抑制(P<0.05),过表达LINC1006则明显促进PCa细胞系LNCaP和PC3的增殖、侵袭能力(P<0.05)。通过PROMO网站预测可见AR是LINC01006的潜在转录调控因子,通过Cistrome DB数据库发现LINC01006上游启动子区域存在AR富集;敲减、抑制AR后LNCaP细胞中LINC01006表达明显升高(P<0.05)。结论:LINC01006在PCa细胞系中呈高表达,促进PCa细胞的增殖和侵袭,其受到AR负向调控,可能在PCa发生发展和去势抵抗形成过程中发挥作用。  相似文献   

6.
细胞形态的形成与维持与其执行的生物学功能息息相关,其中微丝作为细胞骨架,是细胞形态的决定性因素之一。小G蛋白Rho家族是控制微丝的重要信号分子。然而Rho蛋白的不同种类,以及上游的RhoGEF和RhoGAP如何协同调控细胞形态,目前还缺乏系统研究。该研究以果蝇的羊浆膜细胞为模型,通过组织特异性表达RNAi,对Rho家族及其上游信号分子进行遗传筛选。结果显示, Rho1并未参与到羊浆膜细胞形态发生的过程中,同属Rho亚家族的RhoL、RhoBTB, Rac亚家族的Mtl,以及Cdc42是Rho活性的主要来源。通过敲降果蝇的26个RhoGEF和22个RhoGAP,进一步发现了若干重要的羊浆膜发育调控因子,其中在羊浆膜组织中敲降RhoGAP19D导致的胚胎死亡率接近100%。该研究通过RNAi遗传筛选,鉴定出一系列在羊浆膜发育过程中影响微丝的关键因子,为下一步的研究提供线索。  相似文献   

7.
Rho家族小分子鸟苷三磷酸酶(small GTPases of Rho family,Rho GTPases)是调节细胞许多生理病理活动的关键分子开关,参与细胞骨架、基因转录、细胞周期进程、细胞黏附的调控及多条信号通路的调节。骨髓间充质干细胞(bone marrow-derived mesenchymal stem cells,MSCs)是一类具有自我更新和多向分化潜能的特殊细胞,通过增殖、迁移、分化等途径参与机体损伤组织的修复过程。研究表明,Rho GT-Pases在MSCs迁移、分化等过程中起着重要的调节作用。  相似文献   

8.
Rab蛋白是真核生物中保守的小GTP酶家族. Rab蛋白在细胞中普遍表达,它的活性在细胞内受到严格的调控:在活性形式Rab-GTP和无活性形式Rab-GDP之间转换,这是由鸟嘌呤核苷酸交换因子(GEFs)和GTP酶活化蛋白(GAPs)调控的,并在囊泡转运的调控中起分子开关的作用.在囊泡运输中, Rab蛋白与不同的下游效应分子相互作用,参与从供体膜选择货物、出芽形成囊泡、调控囊泡沿细胞骨架运动、囊泡与受体膜锚定融合.当Rab蛋白功能受损导致囊泡转运途径障碍时,则会表现出不同的疾病,包括神经退行性疾病、癌症等.本文将对近年来Rab的蛋白结构和功能、参与囊泡运输的分子机制、Rab蛋白的循环调控以及其异常导致的疾病进行综述.  相似文献   

9.
摘要 目的:探讨低氧诱导因子基因沉默对胃癌细胞BGC-823微血管生成和血管内皮生长因子表达的影响。方法:对数生长期的BGC-823细胞株分为三组-实验组、对照组与空白组,分别转染200 ng/mL shRNA-HIF-1α、200 ng/mL shRNA-NC与等体积的磷酸盐缓冲液。采用CCK法检测细胞增殖,流式细胞仪检测细胞凋亡,Transwell小室实验检测细胞迁移与侵袭,Western blot检测蛋白表达,qRT-PCR检测基因表达。结果:细胞转染后24 h与48 h,实验组的HIF-1α相对表达量、细胞增殖指数、细胞迁移与侵袭指数显著低于空白组和对照组(P<0.05),细胞凋亡指数显著高于空白组和对照组(P<0.05)。细胞转染后48 h,与对照组和空白组相比,实验组的血管内皮生长因子(vascular endothelial growth factor,VEGF) 蛋白相对表达水平显著降低(P<0.05)。结论:沉默HIF-1α表达能抑制胃癌细胞BGC-823微血管生成和VEGF表达,从而抑制胃癌细胞增殖、转移与侵袭,促进细胞凋亡。  相似文献   

10.
Rho小G蛋白家族是Ras超家族成员之一,人类Rho小G蛋白包括20个成员,研究最清楚的有RhoA、Rac1和Cdc42。Rho小G蛋白参与了诸如细胞骨架调节、细胞移动、细胞增殖、细胞周期调控等重要的生物学过程。在这些生物学过程的调节中,Rho小G蛋白的下游效应蛋白质如蛋白激酶(p21-activated kinase,PAK)、ROCK(Rho-kinase)、PKN(protein kinase novel)和MRCK(myotonin-related Cdc42-binding kinase)发挥了不可或缺的作用。迄今研究发现,PAK可调节细胞骨架动力学和细胞运动,另外,PAK通过MAPK(mitogen-activated protein kinases)参与转录、细胞凋亡和幸存通路及细胞周期进程;ROCK与肌动蛋白应力纤维介导黏附复合物的形成及与细胞周期进程的调节有关;哺乳动物的PKN与RhoA/B/C相互作用介导细胞骨架调节;MRCK与细胞骨架重排、细胞核转动、微管组织中心再定位、细胞移动和癌细胞侵袭等有关。该文简要介绍Rho小G蛋白下游激酶PAK、ROCK、PKN和MRCK的结构及其在细胞骨架调节中的功能,重点总结它们在真核细胞周期调控中的作用,尤其是在癌细胞周期进程中所发挥的作用,为寻找癌症治疗的新靶点提供理论依据。  相似文献   

11.
12.
Beck SC  Meyer TF 《FEBS letters》2000,480(2-3):287-292
The target Rho GTPases of many guanine nucleotide exchange factors (GEFs) of the Dbl family remain to be identified. Here we report a new method: the yeast exchange assay (YEA), a rapid qualitative test to perform a wide range screen for GEF specificity. In this assay based on the two-hybrid system, a wild type GTPase binds to its effector only after activation by a specific GEF. We validated the YEA by activating GTPases by previously reported GEFs. We further established that a novel GEF, GEF337, activates RhoA in the YEA. GEF337 promoted nucleotide exchange on RhoA in vitro and promoted F-actin stress fiber assembly in fibroblasts, characteristic of RhoA activation.  相似文献   

13.
XPLN,a guanine nucleotide exchange factor for RhoA and RhoB,but not RhoC   总被引:3,自引:0,他引:3  
Rho proteins cycle between an inactive, GDP-bound state and an active, GTP-bound state. Activation of these GTPases is mediated by guanine nucleotide exchange factors (GEFs), which promote GDP to GTP exchange. In this study we have characterized XPLN, a Rho family GEF. Like other Rho GEFs, XPLN contains a tandem Dbl homology and pleckstrin homology domain topography, but lacks homology with other known functional domains or motifs. XPLN protein is expressed in the brain, skeletal muscle, heart, kidney, platelets, and macrophage and neuronal cell lines. In vitro, XPLN stimulates guanine nucleotide exchange on RhoA and RhoB, but not RhoC, RhoG, Rac1, or Cdc42. Consistent with these data, XPLN preferentially associates with RhoA and RhoB. The specificity of XPLN for RhoA and RhoB, but not RhoC, is surprising given that they share over 85% sequence identity. We determined that the inability of XPLN to exchange RhoC is mediated by isoleucine 43 in RhoC, a position occupied by valine in RhoA and RhoB. When expressed in cells, XPLN activates RhoA and RhoB, but not RhoC, and stimulates the assembly of stress fibers and focal adhesions in a Rho kinase-dependent manner. We also found that XPLN possesses transforming activity, as determined by focus formation assays. In conclusion, here we describe a Rho family GEF that can discriminate between the closely related RhoA, RhoB, and RhoC, possibly giving insight to the divergent functions of these three proteins.  相似文献   

14.
15.
Vav and Vav2 are members of the Dbl family of proteins that act as guanine nucleotide exchange factors (GEFs) for Rho family proteins. Whereas Vav expression is restricted to cells of hematopoietic origin, Vav2 is widely expressed. Although Vav and Vav2 share highly related structural similarities and high sequence identity in their Dbl homology domains, it has been reported that they are active GEFs with distinct substrate specificities toward Rho family members. Whereas Vav displayed GEF activity for Rac1, Cdc42, RhoA, and RhoG, Vav2 was reported to exhibit GEF activity for RhoA, RhoB, and RhoG but not for Rac1 or Cdc42. Consistent with their distinct substrate targets, it was found that constitutively activated versions of Vav and Vav2 caused distinct transformed phenotypes when expressed in NIH 3T3 cells. In contrast to the previous findings, we found that Vav2 can act as a potent GEF for Cdc42, Rac1, and RhoA in vitro. Furthermore, we found that NH(2)-terminally truncated and activated Vav and Vav2 caused indistinguishable transforming actions in NIH 3T3 cells that required Cdc42, Rac1, and RhoA function. In addition, like Vav and Rac1, we found that Vav2 activated the Jun NH(2)-terminal kinase cascade and also caused the formation of lamellipodia and membrane ruffles in NIH 3T3 cells. Finally, Vav2-transformed NIH 3T3 cells showed up-regulated levels of Rac-GTP. We conclude that Vav2 and Vav share overlapping downstream targets and are activators of multiple Rho family proteins. Therefore, Vav2 may mediate the same cellular consequences in nonhematopoietic cells as Vav does in hematopoietic cells.  相似文献   

16.
17.
Signals triggered by diverse receptors modulate the activity of Rho family proteins, although the regulatory mechanism remains largely unknown. On the basis of their biochemical activity as guanine nucleotide exchange factors (GEFs), Dbl family proteins are believed to be implicated in the regulation of Rho family GTP-binding proteins in response to a variety of extracellular stimuli. Here we show that GEF activity of full-length proto-Dbl is enhanced upon tyrosine phosphorylation. When transiently coexpressed with the activated form of the non-receptor tyrosine kinase ACK1, a downstream target of Cdc42, Dbl became tyrosine-phosphorylated. In vitro GEF activity of Dbl toward Rho and Cdc42 was augmented following tyrosine phosphorylation. Moreover, accumulation of the GTP-bound form of Rho and Rac within the cell paralleled ACK-1-dependent tyrosine phosphorylation of Dbl. Consistently, activation of c-Jun N-terminal kinase downstream of Rho family GTP-binding proteins was also enhanced when Dbl was tyrosine-phosphorylated. Collectively, these findings suggest that the tyrosine kinase ACK1 may act as a regulator of Dbl, which in turn activates Rho family proteins.  相似文献   

18.
CDC42 and FGD1 Cause Distinct Signaling and Transforming Activities   总被引:8,自引:2,他引:6       下载免费PDF全文
Activated forms of different Rho family members (CDC42, Rac1, RhoA, RhoB, and RhoG) have been shown to transform NIH 3T3 cells as well as contribute to Ras transformation. Rho family guanine nucleotide exchange factors (GEFs) (also known as Dbl family proteins) that activate CDC42, Rac1, and RhoA also demonstrate oncogenic potential. The faciogenital dysplasia gene product, FGD1, is a Dbl family member that has recently been shown to function as a CDC42-specific GEF. Mutations within the FGD1 locus cosegregate with faciogenital dysplasia, a multisystemic disorder resulting in extensive growth impairments throughout the skeletal and urogenital systems. Here we demonstrate that FGD1 expression is sufficient to cause tumorigenic transformation of NIH 3T3 fibroblasts. Although both FGD1 and constitutively activated CDC42 cooperated with Raf and showed synergistic focus-forming activity, both quantitative and qualitative differences in their functions were seen. FGD1 and CDC42 also activated common nuclear signaling pathways. However, whereas both showed comparable activation of c-Jun, CDC42 showed stronger activation of serum response factor and FGD1 was consistently a better activator of Elk-1. Although coexpression of FGD1 with specific inhibitors of CDC42 function demonstrated the dependence of FGD1 signaling activity on CDC42 function, FGD1 signaling activities were not always consistent with the direct or exclusive stimulation of CDC42 function. In summary, FGD1 and CDC42 signaling and transformation are distinct, thus suggesting that FGD1 may be mediating some of its biological activities through non-CDC42 targets.  相似文献   

19.
The Dbl family of guanine nucleotide exchange factors (GEFs) is made up of a vast array of members that participate in the activation of the Rho family of small GTPases. Dbl-family proteins promote the exchange of guanosine diphosphate/guanosine triphosphate (GDP/GTP) in their target molecules, resulting in the activation of a variety of signaling pathways involved in diverse cellular events, such as actin-cytoskeleton remodeling, cellular invasion, cell movement, and other functions. It has been reported that members of the Dbl family have important roles in several cellular events in Entamoeba histolytica. These include activation of the actin cytoskeleton, cytokinesis, capping, uroid formation, cellular proliferation, erythrophagocytosis, cell migration, and chemotaxis. Here, we report the identification and testing of inhibitors of the E. histolytica guanine nucleotide exchange factor 1 (EhGEF1) protein (the research compounds 2BYRF, 2BY05, 2BYT6, 2BYLX, and 2BYPD), which decreased the in vitro ability of the protein to exchange GDP/GTP at its target GTPases, EhRacG and EhRho1, by 14.9-85.2%. Interestingly, the drug 1,1'-(1,2-phenylene)-bis-(1H-pyrrole-2,5-dione), which completely inhibits the GEF activity of the Trio protein in human cells, decreases the GEF activity of the EhGEF1 protein on the EhRacG and EhRho1 GTPases by 55.7% and 3.2%, respectively. The identification and evaluation of such inhibitors opens up the possibility of obtaining a new pharmacological tool to study the function of amoeba GEF proteins, their roles in various Rho GTPase-mediated signaling pathways, and the repercussions of modulating their activities with respect to several mechanisms related to E. histolytica pathogenesis.  相似文献   

20.
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