首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 218 毫秒
1.
肿瘤转移是一个多阶段、多途径、涉及多基因及其信号通路变化的一系列复杂过程。了解肿瘤转移相关基因的信号传导通路以及对肿瘤转移的作用机制,为寻找抑制肿瘤转移的关键靶点具有重要的意义。Ezrin高表达与肿瘤转移密切相关,它可通过改变肿瘤细胞极性及细胞运动、调节肿瘤细胞间黏附及细胞与细胞外基质黏附、参与肿瘤细胞内信号转导而影响恶性肿瘤转移。Ezrin过度表达可以破坏正常细胞内信号传递网络的平衡,其中主要涉及的为细胞信号转导相关分子(Rho)及受体酪氨酸蛋白激酶等信号传导途径。Ezrin借助于细胞内错综复杂的信号转导网络调控细胞的形态构成、黏附、吞噬、运动、血管形成等一系列的生物学过程,最终实现肿瘤细胞的侵袭和转移。本文就Ezrin蛋白的信号转导通路及其对肿瘤转移作用的研究进展做一综述。  相似文献   

2.
Ezrin是细胞骨架与细胞膜连接的特定蛋白之一,它有助于细胞内摄作用、细胞胞吐作用及跨膜信号发放的途径.研究表明,Ezrin在不同肿瘤组织中表达异常,推测它可能参与肿瘤的侵袭转移,其通过改变肿瘤细胞极性及细胞运动、调节肿瘤细胞间黏附及细胞与细胞外基质黏附、参与肿瘤细胞内信号转导而影响恶性肿瘤转移.本文主要介绍了Ezrin生物学特性、与CD44相互关系以及目前在肿瘤研究中的现状.  相似文献   

3.
埃兹蛋白:生物学特征及其在肿瘤转移中的作用   总被引:8,自引:0,他引:8  
埃兹蛋白(ezrin)是埃兹蛋白、根蛋白和膜突蛋白(ezrin-radixin-moesin,ERM)家族成员之一,主要参与上皮细胞中细胞骨架与胞膜之间的连接,具有维持细胞形态和运动、连接黏附分子及调节信号转导等功能。近年来的研究发现,埃兹蛋白在肿瘤细胞中的表达异常,提示其在肿瘤的浸润、转移机制中发挥重要作用。  相似文献   

4.
L-periaxin是外周神经系统特异表达的骨架蛋白之一,占外周神经系统髓鞘总蛋白质的16%,参与髓鞘形成和维护。埃兹蛋白(Ezrin)属于Ezrin-Radxin-Moesin(ERM)蛋白质家族,与细胞黏附、迁移、生存以及肿瘤的发生、发展相关。作者前期研究证实,L-periaxin与Ezrin通过"头对头,尾对尾"的模式相互结合。本文通过双分子荧光互补、免疫共沉淀、GST pull down、荧光共定位、海肾荧光素酶互补、荧光光谱以及分子对接等方法,揭示L-periaxin的核定位信号区(nuclear location signal,NLS)与蛋白质Ezrin的FERM(Ezrin Radixin Moesin)结构域的"头对头"相互结合,依赖Lperiaxin第3段核定位信号序列NLS3与Ezrin的FERM结构域的F3亚结构域。本结果为进一步理解Ezrin在髓鞘化过程中的作用,以及阐明调节L-periaxin蛋白功能的信号通路奠定基础。  相似文献   

5.
L-periaxin是外周神经系统特异表达的骨架蛋白之一,占外周神经系统髓鞘总蛋白质的16%,参与髓鞘形成和维护。埃兹蛋白(Ezrin)属于Ezrin-Radxin-Moesin(ERM)蛋白质家族,与细胞黏附、迁移、生存以及肿瘤的发生、发展相关。作者前期研究证实,L-periaxin与Ezrin通过"头对头,尾对尾"的模式相互结合。本文通过双分子荧光互补、免疫共沉淀、GST pull down、荧光共定位、海肾荧光素酶互补、荧光光谱以及分子对接等方法,揭示L-periaxin的核定位信号区(nuclear location signal,NLS)与蛋白质Ezrin的FERM(Ezrin Radixin Moesin)结构域的"头对头"相互结合,依赖Lperiaxin第3段核定位信号序列NLS3与Ezrin的FERM结构域的F3亚结构域。本结果为进一步理解Ezrin在髓鞘化过程中的作用,以及阐明调节L-periaxin蛋白功能的信号通路奠定基础。  相似文献   

6.
目的:骨肉瘤是一种常见的恶性骨肿瘤,恶性程度高,往往在早期就会发生远隔器官的转移,从而导致骨肉瘤的预后非常差。Orai1是一类定位于细胞膜,介导钙离子内流的受体依赖性钙通道蛋白。大量研究发现钙通道蛋白Orai1过表达于多种肿瘤细胞,并对维持肿瘤细胞粘附、侵袭、迁移等恶性表型有非常重要的作用。然而,钙通道蛋白Orai1是否参与了骨肉瘤的转移过程,目前未见相关报道。本研究的目的是探究钙通道蛋白Orai1是否在骨肉瘤转移过程中的发挥作用。方法:利用合成的靶向Orai1的小干扰RNA(Orai1 si RNA)片段,转染至人骨肉瘤细胞系Saos-2细胞。在Saos-2细胞中抑制Orai1的表达。采用细胞黏附实验、细胞划痕实验和细胞Transwell实验检测骨肉瘤细胞的黏附、迁移及侵袭等肿瘤细胞转移能力;Western-blot实验检测Saos-2细胞的中黏着斑激酶(FAK)和桩蛋白(Paxillin)的表达水平和磷酸化水平。结果:靶向Orai1 si RNA瞬时转染至骨肉瘤细胞系Saos-2细胞后,Saos-2细胞中Orai1蛋白表达水平和m RNA转录水平均显著下降。并且,在Saos-2细胞中抑制Orai1表达后,Saos-2细胞的黏附能力、迁移能力、及侵袭能力均显著下降。进一步研究发现,在Saos-2细胞中抑制Orai1表达后,Saos-2细胞的FAK和Paxillin磷酸化水平明显下降。结论:Orai1可以促进骨肉瘤细胞的黏附、迁移和侵袭,增加黏着斑的形成,从而促进骨肉瘤的转移。因此,深入研究钙通道蛋白Orai1调控骨肉瘤转移的分子机制,可为骨肉瘤转移的治疗提供新的新方向和新策略。  相似文献   

7.
热休克蛋白27(HSP27)作为热应激蛋白表达于各种生理或是环境损伤之后,保护细胞生存,其具有多种功能包括:分子伴侣,抗凋亡,参与细胞运动等。近年来发现HSP27在多种肿瘤中过度表达,参与肿瘤的发生发展,分化,耐药以及转移等方面,因而抑制HSP27成为一种新的肿瘤治疗策略。本文就相关研究进展进行综述。  相似文献   

8.
热休克蛋白27(HSP27)作为热应激蛋白表达于各种生理或是环境损伤之后,保护细胞生存,其具有多种功能包括:分子伴侣,抗凋亡,参与细胞运动等。近年来发现HSP27在多种肿瘤中过度表达,参与肿瘤的发生发展,分化,耐药以及转移等方面,因而抑制HSP27成为一种新的肿瘤治疗策略。本文就相关研究进展进行综述。  相似文献   

9.
Pan Y  Han J  Zhang Y  Li XJ 《生理科学进展》2010,41(6):413-416
波形蛋白(vimentin)是存在于间充质细胞中的一种中间丝蛋白,近些年研究显示vimentin与肿瘤发生、转移密切相关。波形蛋白调节细胞骨架蛋白、细胞粘附分子等蛋白间的相互作用,参与肿瘤细胞和肿瘤相关内皮细胞、巨噬细胞的粘附、迁移、侵袭和细胞信号转导。其高度动态的聚合解聚间的平衡和其复杂的磷酸化形式可能是vimentin参与肿瘤转移过程及细胞-细胞间相互作用的调节机制。Vimentin在肿瘤中的功能提示,其可能是抗肿瘤转移治疗药物研究的新靶点。  相似文献   

10.
Ezrin蛋白是ERM(Ezrin;Radixin;Moesin)蛋白家族成员之一。作为酪氨酸激酶的底物,Ezrin蛋白能在细胞膜蛋白与肌动蛋白骨架之间起到桥梁的作用。近年,大量研究表明Ezrin蛋白广泛参与乳腺癌细胞增殖、凋亡、黏附、侵袭转移以及新生血管发生的调节过程。上述过程不仅与Ezrin蛋白自身表达水平、亚细胞定位等改变密切相关,而且受到原发乳腺癌细胞微环境改变的影响,同时涉及胞膜粘附分子(CD44、ICAM、E-cadherin)、EGF、HGF、PDGF、E2及其相应受体所介导的多条信号通路的交叉互动。明确Ezrin蛋白在乳腺癌细胞迁移侵袭过程中的作用及机制,可为乳腺癌的防治提供潜在的药物干预靶点,并为肿瘤转移机制的阐明提供进一步的理论依据。本文就Ezrin蛋白在乳腺癌细胞迁移侵袭过程中的作用作一综述。  相似文献   

11.
Cancer metastasis is the primary cause of morbidity and mortality in cancer as it remains the most complicated, devastating, and enigmatic aspect of cancer. Several decades of extensive research have identified several key players closely associated with metastasis. Among these players, cytoskeletal linker Ezrin (the founding member of the ERM (Ezrin-Radixin-Moesin) family) was identified as a critical promoter of metastasis in pediatric cancers in the early 21st century. Ezrin was discovered 40 years ago as a aminor component of intestinal epithelial microvillus core protein, which is enriched in actin-containing cell surface structures. It controls gastric acid secretion and plays diverse physiological roles including maintaining cell polarity, regulating cell adhesion, cell motility and morphogenesis. Extensive research for more than two decades evinces that Ezrin is frequently dysregulated in several human cancers. Overexpression, altered subcellular localization and/or aberrant activation of Ezrin are closely associated with higher metastatic incidence and patient mortality, thereby justifying Ezrin as a valuable prognostic biomarker in cancer. Ezrin plays multifaceted role in multiple aspects of cancer, with its significant contribution in the complex metastatic cascade, through reorganizing the cytoskeleton and deregulating various cellular signaling pathways. Current preclinical studies using genetic and/or pharmacological approaches reveal that inactivation of Ezrin results in significant inhibition of Ezrin-mediated tumor growth and metastasis as well as increase in the sensitivity of cancer cells to various chemotherapeutic drugs. In this review, we discuss the recent advances illuminating the molecular mechanisms responsible for Ezrin dysregulation in cancer and its pleiotropic role in cancer progression and metastasis. We also highlight its potential as a prognostic biomarker and therapeutic target in various cancers. More importantly, we put forward some potential questions, which we strongly believe, will stimulate both basic and translational research to better understand Ezrin-mediated malignancy, ultimately leading to the development of Ezrin-targeted cancer therapy for the betterment of human life.  相似文献   

12.
Elucidation of how pancreatic cancer cells give rise to distant metastasis is urgently needed in order to provide not only a better understanding of the underlying molecular mechanisms, but also to identify novel targets for greatly improved molecular diagnosis and therapeutic intervention. We employed combined proteomic technologies including mass spectrometry and isobaric tags for relative and absolute quantification peptide tagging to analyze protein profiles of surgically resected human pancreatic ductal adenocarcinoma tissues. We identified a protein, dihydropyrimidinase-like 3, as highly expressed in human pancreatic ductal adenocarcinoma tissues as well as pancreatic cancer cell lines. Characterization of the roles of dihydropyrimidinase-like 3 in relation to cancer cell adhesion and migration in vitro, and metastasis in vivo was performed using a series of functional analyses, including those employing multiple reaction monitoring proteomic analysis. Furthermore, dihydropyrimidinase-like 3 was found to interact with Ezrin, which has important roles in cell adhesion, motility, and invasion, while that interaction promoted stabilization of an adhesion complex consisting of Ezrin, c-Src, focal adhesion kinase, and Talin1. We also found that exogenous expression of dihydropyrimidinase-like 3 induced activating phosphorylation of Ezrin and c-Src, leading to up-regulation of the signaling pathway. Taken together, the present results indicate successful application of combined proteomic approaches to identify a novel key player, dihydropyrimidinase-like 3, in pancreatic ductal adenocarcinoma tumorigenesis, which may serve as an important biomarker and/or drug target to improve therapeutic strategies.  相似文献   

13.
Gem is a protein of the Ras superfamily that plays a role in regulating voltage-gated Ca2+ channels and cytoskeletal reorganization. We now report that GTP-bound Gem interacts with the membrane-cytoskeleton linker protein Ezrin in its active state, and that Gem binds to active Ezrin in cells. The coexpression of Gem and Ezrin induces cell elongation accompanied by the disappearance of actin stress fibers and collapse of most focal adhesions. The same morphological effect is elicited when cells expressing Gem alone are stimulated with serum and requires the expression of ERM proteins. We show that endogenous Gem down-regulates the level of active RhoA and actin stress fibers. The effects of Gem downstream of Rho, i.e., ERM phosphorylation as well as disappearance of actin stress fibers and most focal adhesions, require the Rho-GAP partner of Gem, Gmip, a protein that is enriched in membranes under conditions in which Gem induced cell elongation. Our results suggest that Gem binds active Ezrin at the plasma membrane-cytoskeleton interface and acts via the Rho-GAP protein Gmip to down-regulate the processes dependent on the Rho pathway.  相似文献   

14.
Ou-Yang M  Liu HR  Zhang Y  Zhu X  Yang Q 《Biochimie》2011,93(5):954-961
Three closely related proteins, ezrin, radixin, and moesin (ERM), which primarily act as a linker between the plasma membrane and the cytoskeleton, are involved in many cellular functions, including regulation of actin cytoskeleton, control of cell shape, adhesion and motility, and modulation of signaling pathways. Although, ezrin is now recognized as a key component in tumor metastasis, the functional role of the radixin and moesin in tumor metastasis has not been established. In the present study, we chose highly metastatic human gastric carcinoma SGC-7901 cells, which express all of the ERM proteins as a model to examine the functional roles of these proteins in tumor metastasis. Ezrin, radixin or moesin stable knockdown SGC-7901 cell lines were established using siRNA methodology. In vitro cell migration and invasion studies showed that either ezrin, radixin or moesin specific deficiency in the cells caused the substantial reduction of the cell migration and invasion. Western blotting and immunofluorescence analysis showed that the expression of E-cadherin was also significantly increased when any member of ERM proteins was downregulated. Our results indicated that these three ERM proteins play similar roles in the SGC-7901 cell metastatic potential and their roles of upregulating the expression of E-cadherin may be important in tumor progression.  相似文献   

15.
Ezrin acts as a dynamic linkage between plasma membrane and cytoskeleton, and thus involved in many fundamental cellular functions. Yet, its potential role in human skin is virtually unknown. Here we investigate the role of Ezrin in primary skin fibroblasts, the major cells responsible extracellular matrix (ECM) production. We report that Ezrin play an important role in the maintenance of skin fibroblast size/mechanical properties and proliferation. siRNA-mediated Ezrin knockdown decreased fibroblast size and mechanical properties, and thus impaired the nuclear translocation of YAP, a protein commonly response to cell size and mechanical force. Functionally, depletion of Ezrin significantly inhibited YAP target gene expression and fibroblast proliferation. Conversely, restoration of YAP nuclear translocation by overexpression of constitutively active YAP reversed YAP target genes expression and rescued proliferation in Ezrin knockdown cells. These data reveal a novel role for Ezrin in maintenance of fibroblast size/mechanical force and regulating YAP-mediated proliferation.  相似文献   

16.
BackgroundEzrin, links the plasma membrane to the actin cytoskeleton, and plays an important role in the development and progression of human esophageal squamous cell carcinoma (ESCC). However, the roles of ezrin S66 phosphorylation in tumorigenesis of ESCC remain unclear.MethodsDistribution of ezrin in membrane and cytosol fractions was examined by analysis of detergent-soluble/-insoluble fractions and cytosol/membrane fractionation. Both immunofluorescence and live imaging were used to explore the role of ezrin S66 phosphorylation in the behavior of ezrin and actin in cell filopodia. Cell proliferation, migration and invasion of ESCC cells were investigated by proliferation and migration assays, respectively. Tumorigenesis, local invasion and metastasis were assessed in a nude mouse model of regional lymph node metastasis.ResultsEzrin S66 phosphorylation enhanced the recruitment of ezrin to the membrane in ESCC cells. Additionally, non-phosphorylatable ezrin (S66A) significantly prevented filopodia formation, as well as caused a reduction in the number, length and lifetime of filopodia. Moreover, functional experiments revealed that expression of non-phosphorylatable ezrin (S66A) markedly suppressed migration and invasion but not proliferation of ESCC cells in vitro, and attenuated local invasion and regional lymph node metastasis, but not primary tumor growth of ESCC cells in vivo.ConclusionEzrin S66 phosphorylation enhances filopodia formation, contributing to the regulation of invasion and metastasis of esophageal squamous cell carcinoma cells.  相似文献   

17.
Caspase-8 governs multiple cell responses to the microenvironmental cues. However, its integration of "death-life" signalings remains elusive. In our study, the role of caspase-8-Src is well-established as a promoter for migration or metastasis in Casp8(+)Src(+) A549/H226 cells in vivo and in vitro. In particular for nude mice models, mice implanted with Casp8(+)Src(+) A459/H226 cells remarkably increased spontaneous tumor metastatic burden with a significant survival disadvantage. Additionally, we detect that Src-mediated caspase-8 phosphorylation stimulates Src phosphorylation at Tyr-416 via the linkage of Src SH2 domain with phosph-Tyr-380 site of caspase-8. In turn, activated Src can efficiently induce epithelial-mesenchymal transition (EMT) phenotypic features to promote tumor cells metastasis. Surprisingly, RXDLL motif deletion in the DEDa of caspase-8 attenuates tumor cell migration or metastasis via impairing the recruitment of caspase-8 into the cellular periphery where activated Src is subject to caspase-8 phosphorylation. Together, a simple model is that the peripherization of caspase-8 is well-poised to facilitate Src-mediated caspase-8 phosphrylation at Tyr-380, then binding of phospho-Tyr380 of caspase-8 to Src SH2 domain may maintain Src in an active conformation to induce EMT phenotype, a key step toward cancer metastasis.  相似文献   

18.
The epithelial cells exhibit either a columnar or a flat shape dependent on extracellular stimuli or the cell-cell adhesion. Membrane-anchored ephrinA stimulates EphA receptor tyrosine kinases as a ligand in a cell-cell contact-dependent manner. The mechanism through which ephrinA1/EphA2 signal regulates the cell morphology remains elusive. We demonstrate here that ephrinA1/EphA2 signal induces compaction and enhanced polarization (columnar change) of Madin-Darby canine kidney epithelial cells by regulating Ezrin, a linker that connects plasma membrane and actin cytoskeleton. Activation of EphA2 resulted in RhoA inactivation through p190RhoGAP-A and subsequent dephosphorylation of Ezrin on Thr-567 phosphorylated by Rho kinase. Consistently, the cells expressing an active mutant of Ezrin in which Thr-567 was replaced with Asp did not change their shape in response to ephrinA1. Furthermore, depletion of Ezrin led to compaction and enhanced polarization without ephrinA1 stimulation, suggesting the role for active Ezrin in keeping the flat cell shape. Ezrin localized to apical domain irrespective of ephrinA1 stimulation, whereas phosphorylated Ezrin on the apical domain was reduced by ephrinA1 stimulation. Collectively, ephrinA1/EphA2 signal negatively regulates Ezrin and promotes the alteration of cell shape, from flat to columnar shape.  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