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 125 毫秒
1.
线粒体丝氨酸蛋白酶Omi/HtrA2与细胞凋亡   总被引:4,自引:0,他引:4  
Wang XL  Wang J  Lv XP 《生理科学进展》2006,37(3):285-288
Omi/HtrA2是一种线粒体丝氨酸蛋白酶,具有修复、降解线粒体中折叠错误的蛋白质的作用,并可以通过破坏caspase与X染色体连锁凋亡抑制蛋白(XIAP)之间的相互作用和直接利用其自身具有的蛋白酶活性引起细胞凋亡。本文介绍了Omi/HtrA2的结构、生物学作用、参与细胞凋亡的机制及其在某些疾病中的作用。  相似文献   

2.
线粒体促凋亡因子Omi/HtrA2在乳腺癌中的表达及意义   总被引:1,自引:0,他引:1  
目的本文旨在通过检测促凋亡分子Omi/HtrA2在乳腺良恶性肿瘤组织中的表达,探讨其与乳腺癌发病的关系及临床意义.方法采用免疫组化EnVinsion system法检测63例乳腺癌及37例乳腺纤维腺瘤组织中Omi/HtrA2的表达情况,同时检测这些组织中Caspase-3蛋白的表达.结果 Omi/HtrA2在乳腺纤维腺瘤中表达显著低于乳腺癌中的表达(P<0.05),而Caspase-3则在乳腺纤维腺瘤中表达显著高于乳腺癌中的表达(P<0.05),但二者的表达与乳腺癌的临床分期均无明显相关性(P>0.05).结论线粒体促凋亡蛋白Omi/HtrA2的在乳腺癌中高表达与其发生发展有关.  相似文献   

3.
目的:p66Shc在线粒体内积累和HtrA2/Omi的功能缺陷都能导致线粒体损伤,诱导细胞凋亡.探讨在线粒体中HtrA2对p66Shc的调控作用.方法:构建p66Shc和成熟型HtrA2的真核表达质粒,共转染HEK293T细胞,免疫印迹法(Western blot)检测p66Shc蛋白;构建原核表达质粒,大肠杆菌纯化蛋白,体外切割实验,SDS-PAGE分离后考马斯亮蓝染色检测;提取HtrA2功能缺陷小鼠( mnd2)大脑组织的线粒体,检测线粒体内p66Shc的蛋白水平.结果:细胞实验和体外实验证明HtrA2可以切割p66Shc,且在mnd2小鼠大脑中,线粒体内p66Shc的蛋白水平明显升高(P<0.05).结论:p66Shc是HtrA2的直接底物,且HtrA2参与调节线粒体中p66Shc的蛋白水平,揭示了HtrA2发挥神经保护功能新的可能机制.  相似文献   

4.
HtrA2/Omi是一种线粒体丝氨酸蛋白酶, 在哺乳动物细胞中具有双重功能, 即诱导细胞凋亡和参与维持线粒体活性的动态平衡. PDZ结构域是最重要的蛋白质相互作用结构域之一, 参与多种生物学过程, 如细胞信号转导、蛋白质降解、细胞骨架组织等. 最近研究发现, HtrA2/Omi蛋白的PDZ结构域与配体的相互作用, 可以调节HtrA2/Omi蛋白自身的水解酶活性.以HtrA2/Omi PDZ结构域为研究对象, 用酵母双杂交系统验证性筛选PDZ结构域结合配体文库, 快速研究该结构域的结合特性, 并在人类全蛋白质组范围内预测并发现该结构域新的相互作用蛋白, 最后分析这些新的相互作用所能够形成的最小相互作用网络来评估其可信度. 研究结果揭示了HtrA2/Omi PDZ结构域新的结合特性, 即: 不仅能够结合已报道的II类PDZ配体而且还可以结合I类和III类PDZ配体, 并且配体-3位氨基酸具有一定范围内的可变性. 而且, 发现了7个新的HtrA2/Omi PDZ结构域相互作用蛋白, 为进一步阐明HtrA2/Omi蛋白的生物学功能提供了重要线索. 同时证明了验证性筛选目的结构域结合配体文库, 这一结构域结合特性研究新策略的实用性和高效性.  相似文献   

5.
本研究探讨了外源性C2-神经酰胺诱导入结肠癌HT-29细胞凋亡中,线粒体膜间隙凋亡蛋白的释放机制.不同浓度C2-神经酰胺作用HT-29细胞,流式细胞仪检测线粒体膜电位(△ψm),线粒体/细胞液分离试剂盒分离亚细胞成分,聚丙烯酰胺凝胶电泳检测细胞色素C(Cytc)、高温必需蛋白A2(HtrA2)、线粒体源性半胱天冬氨酸蛋白酶第二活化因子(Smac)、凋亡抑制蛋白(XtAP)和半胱天冬氨酸蛋白酶-3(Caspase-3)蛋白表达水平.实验结果显示25和50μmol/L C2-神经酰胺作用细胞6h,△ψm即开始下降(P〈0.05),且环孢霉素能通过调节线粒体膜通透性转换孔抑制△ψm的下降.C2-神经酰胺对Cyt c,HtrA2和Smac总蛋白表达没有明显影响,但能诱导Cyt c,HtrA2和Smac从线粒体释放入细胞液中,并下调XIAP蛋白的表达及活化Caspase-3.在Caspase抑制剂存在下,C2-神经酰胺仍能诱导Cyt c和HtrA2从线粒体释放,但不能诱导Smac释放.因此认为C2-神经酰胺能通过线粒体凋亡通路诱导HT-29细胞凋亡,C2-神经酰胺诱导Cytc和HtrA2从线粒体的释放是Caspase非依赖性的,而Smac释放是Caspase依赖性的.  相似文献   

6.
目的:观察Nur77通过线粒体转位对缺氧/复氧(H/R)诱导的心肌细胞凋亡的影响。方法:原代培养l-2天SD大鼠心肌细胞,建立H/R模型。随机分为正常对照组、H/R组、Nur77组,采用免疫荧光检测横纹肌肌动蛋白(α-actin)鉴定心肌细胞;采用TUNEL染色法及Caspase-3酶活性检测心肌细胞凋亡情况;采用Western blot检测细胞核及线粒体Nur77蛋白表达、线粒体及胞浆Omi/HtrA2蛋白表达。结果:H/R组细胞核中Nur77蛋白表达明显低于正常对照组;而在线粒体中则相反。Nur77组线粒体中的Omi/HtrA2蛋白表达明显低于正常对照组;而在胞浆中则相反。结论:在心肌细胞H/R损伤时,Nur77线粒体转位促使Omi/HtrA2蛋白从线粒体释放入胞浆,从而导致心肌细胞凋亡。  相似文献   

7.
线粒体促凋亡因子Omi/HtrA2在肝癌组织中表达的研究   总被引:4,自引:0,他引:4  
目的探讨丝氨酸蛋白酶Omi/HtrA2在肝癌组织、癌旁组织与正常肝组织中的表达及意义。方法应用免疫组化SABC法检测43例肝癌、30例癌旁组织及10例正常肝组织中Omi/HtrA2的表达。结果29例(67·44%)肝癌中Omi/HtrA2蛋白表达阳性,30例癌旁组织和10正常肝组织没有或只有少量很弱的表达。肝癌高分化组中Omi/HtrA2蛋白的表达明显高于中、低分化组(P<0·01)。另外,Omi/HtrA2表达与肿瘤大小及临床分期相关,但Omi/HtrA2表达与肝硬化、有无癌栓、HBsAg和AFP无关。结论肝细胞癌可能需要Omi/HtrA2的表达来促进凋亡,Omi/HtrA2的表达对肝癌的发展有重要作用。  相似文献   

8.
线粒体在控制细胞死亡中的作用   总被引:3,自引:0,他引:3  
细胞死亡由细胞坏死或细胞凋亡所致。细胞坏死时 ,细胞质膜形成疱 (突起 ) ,疱破裂释放细胞内容物 ;细胞凋亡时 ,细胞内容物不释放到细胞外。细胞坏死时 ,细胞内ATP耗竭 ;凋亡时 ,细胞需利用ATP完成凋亡过程。1.线粒体外膜释放凋亡活性物质细胞凋亡过程中 ,原先位于线粒体膜间隙的某些与凋亡有关的活性物质释放到胞液中 ,这些物质包括细胞色素c(Cytc)、凋亡诱导因子 (apoptosis inducingfactor ,AIF)、线粒体胱天蛋白酶 (caspase)2 ,3,9、hsp10、hsp6 0、Bcl 2家族成员等。细胞受到凋…  相似文献   

9.
活性氧、线粒体通透性转换与细胞凋亡   总被引:2,自引:0,他引:2  
线粒体是真核细胞中非常重要的细胞器,细胞中的活性氧等自由基主要来源于此,线粒体膜的通透性转换(mitochondrial permeability transition,MPT)及其孔道(mitochondrialpermeability transition pore,MPTP)更是在内源性细胞凋亡中发挥了关键作用。持续性的线粒体膜通透性转换在凋亡的效应阶段起决定性作用,可介导细胞色素c等促凋亡因子从线粒体释放到胞浆中,进一步激活下游的信号通路,导致细胞不可逆地走向凋亡。瞬时性的线粒体膜通透性转换及其偶联的线粒体局部的活性氧爆发同样具有促凋亡的作用。线粒体通透性孔道的开放释放出大量活性氧,这些活性氧又能够进一步激活该孔道,以正反馈的形式进一步加剧孔道的打开,放大凋亡信号。活性氧、线粒体通透性转换与细胞凋亡之间具有密不可分的联系,本文根据已知的研究结果集中讨论了这三者的关系,并着重论述了该领域中的最新发现和成果。  相似文献   

10.
线粒体分裂、融合与细胞凋亡   总被引:2,自引:0,他引:2  
线粒体是高度动态变化的细胞器,其在细胞内不断分裂、融合并形成网状结构。线粒体的分裂和融合是由多种蛋白质精确调控完成的。Drp1/Dnm1p,Fis1/Fis1p,Caf4p和Mdv1p参与线粒体分裂的调控;Mfn1/2/Fzo1p控制线粒体外膜的融合,而Mgm1p/OPA1则参与线粒体内膜的融合。在细胞凋亡过程中线粒体片段化,网状结构被破坏,线粒体嵴发生重构,抑制这一过程可以部分抑制细胞色素c的释放和细胞凋亡。线粒体形态对于细胞维持正常生理代谢和机体发育起着重要的作用,一旦出现障碍会导致严重的疾病。  相似文献   

11.
Mitochondria,the killer organelles and their weapons   总被引:35,自引:0,他引:35  
Apoptosis is a cell-autonomous mode of death that is activated to eradicate superfluous, damaged, mutated, or aged cells. In addition to their role as the cell's powerhouse, mitochondria play a central role in the control of apoptosis. Thus, numerous pro-apoptotic molecules act on mitochondria and provoke the permeabilization of mitochondrial membranes. Soluble proteins contained in the mitochondrial intermembrane space are released through the outer membrane and participate in the organized destruction of the cell. Several among these lethal proteins can activate caspases, a class of cysteine proteases specifically activated in apoptosis, whereas others act in a caspase-independent fashion, by acting as nucleases (e.g., endonuclease G), nuclease activators (e.g., apoptosis-inducing factor), or serine proteases (e.g., Omi/HtrA2). In addition, mitochondria can generate reactive oxygen species, following uncoupling and/or inhibition of the respiratory chain. The diversity of mitochondrial factors participating in apoptosis emphasizes the central role of these organelles in apoptosis control and unravels novel mechanisms of cell death execution.  相似文献   

12.
Vaux DL  Silke J 《Cell》2003,115(3):251-253
Mammalian mitochondrial HtrA2/Omi was originally described as an apoptosis inducer, but rather than having extra cells, mice with mutant HtrA2/Omi suffer from a neurodegenerative disease due to progressive mitochondrial damage. This suggests that instead of promoting cell death by antagonizing inhibitor of apoptosis (IAP) proteins, the primary function of HtrA2/Omi is to handle misfolded proteins in the mitochondria.  相似文献   

13.
Mitochondria are 'life-essential' organelles for the production of metabolic energy in the form of ATP. Paradoxically mitochondria also play a key role in controlling the pathways that lead to cell death. This latter role of mitochondria is more than just a 'loss of function' resulting in an energy deficit but is an active process involving different mitochondrial proteins. Cytochrome c was the first characterised mitochondrial factor shown to be released from the mitochondrial intermembrane space and to be actively implicated in apoptotic cell death. Since then, other mitochondrial proteins, such as AIF, Smac/DIABLO, endonuclease G and Omi/HtrA2, were found to undergo release during apoptosis and have been implicated in various aspects of the cell death process. Members of the Bcl-2 protein family control the integrity and response of mitochondria to apoptotic signals. The molecular mechanism by which mitochondrial intermembrane space proteins are released and the regulation of mitochondrial homeostasis by Bcl-2 proteins is still elusive. This review summarises and evaluates the current knowledge concerning the complex role of released mitochondrial proteins in the apoptotic process.  相似文献   

14.
Loss of Omi/HtrA2 function leads to nerve cell loss in mouse models and has been linked to neurodegeneration in Parkinson's and Huntington's disease. Omi/HtrA2 is a serine protease released as a pro-apoptotic factor from the mitochondrial intermembrane space into the cytosol. Under physiological conditions, Omi/HtrA2 is thought to be involved in protection against cellular stress, but the cytological and molecular mechanisms are not clear. Omi/HtrA2 deficiency caused an accumulation of reactive oxygen species and reduced mitochondrial membrane potential. In Omi/HtrA2 knockout mouse embryonic fibroblasts, as well as in Omi/HtrA2 silenced human HeLa cells and Drosophila S2R+ cells, we found elongated mitochondria by live cell imaging. Electron microscopy confirmed the mitochondrial morphology alterations and showed abnormal cristae structure. Examining the levels of proteins involved in mitochondrial fusion, we found a selective up-regulation of more soluble OPA1 protein. Complementation of knockout cells with wild-type Omi/HtrA2 but not with the protease mutant [S306A]Omi/HtrA2 reversed the mitochondrial elongation phenotype and OPA1 alterations. Finally, co-immunoprecipitation showed direct interaction of Omi/HtrA2 with endogenous OPA1. Thus, we show for the first time a direct effect of loss of Omi/HtrA2 on mitochondrial morphology and demonstrate a novel role of this mitochondrial serine protease in the modulation of OPA1. Our results underscore a critical role of impaired mitochondrial dynamics in neurodegenerative disorders.  相似文献   

15.
The inhibitor-of-apoptosis proteins (IAPs) play a critical role in the regulation of apoptosis by binding and inhibiting caspases. Reaper family proteins and Smac/DIABLO use a conserved amino-terminal sequence to bind to IAPs in flies and mammals, respectively, blocking their ability to inhibit caspases and thus promoting apoptosis. Here we have identified the serine protease Omi/HtrA2 as a second mammalian XIAP-binding protein with a Reaper-like motif. This protease autoprocesses to form a protein with amino-terminal homology to Smac/DIABLO and Reaper family proteins. Full-length Omi/HtrA2 is localized to mitochondria but fails to interact with XIAP. Mitochondria also contain processed Omi/HtrA2, which, following apoptotic insult, translocates to the cytosol, where it interacts with XIAP. Overexpression of Omi/HtrA2 sensitizes cells to apoptosis, and its removal by RNA interference reduces cell death. Omi/HtrA2 thus extends the set of mammalian proteins with Reaper-like function that are released from the mitochondria during apoptosis.  相似文献   

16.
The mature serine protease Omi/HtrA2 is released from the mitochondria into the cytosol during apoptosis. Suppression of Omi/HtrA2 by RNA interference in human cell lines reduces cell death in response to TRAIL and etoposide. In contrast, ectopic expression of mature wildtype Omi/HtrA2, but not an active site mutant, induces potent caspase activation and apoptosis. In vitro assays demonstrated that Omi/HtrA2 could degrade inhibitor of apoptosis proteins (IAPs). Consistent with this observation, increased expression of Omi/HtrA2 in cells increases degradation of XIAP, while suppression of Omi/HtrA2 by RNA interference has an opposite effect. Combined, our data demonstrate that IAPs are substrates for Omi/HtrA2, and their degradation could be a mechanism by which the mitochondrially released Omi/HtrA2 activates caspases during apoptosis.  相似文献   

17.
Omi/HtrA2 is a mitochondrial serine protease that is released into the cytosol during apoptosis and promotes cytochrome c (Cyt c)dependent caspase activation by neutralizing inhibitor of apoptosis proteins (IAPs) via its IAP-binding motif. The protease activity of Omi/HtrA2 also contributes to the progression of both apoptosis and caspase-independent cell death. In this study, we found that wild-type Omi/HtrA2 is more effective at caspase activation than a catalytically inactive mutant of Omi/HtrA2 in response to apoptotic stimuli, such as UV irradiation or tumor necrosis factor. Although similar levels of Omi/HtrA2 expression, XIAP-binding activity, and Omi/HtrA2 mitochondrial release were observed among cells transfected with catalytically inactive and wild-type Omi/HtrA2 protein, XIAP protein expression after UV irradiation was significantly reduced in cells transfected with wild-type Omi/HtrA2. Recombinant Omi/HtrA2 was observed to catalytically cleave IAPs and to inactivate XIAP in vitro, suggesting that the protease activity of Omi/HtrA2 might be responsible for its IAP-inhibiting activity. Extramitochondrial expression of Omi/HtrA2 indirectly induced permeabilization of the outer mitochondrial membrane and subsequent Cyt c-dependent caspase activation in HeLa cells. These results indicate that protease activity of Omi/HtrA2 promotes caspase activation through multiple pathways.  相似文献   

18.
Omi/HtrA2 is a nuclear encoded mitochondrial serine protease with dual and opposite functions that depend entirely on its subcellular localization. During apoptosis, Omi/HtrA2 is released into the cytoplasm where it participates in cell death. While confined in the inter-membrane space of the mitochondria, Omi/HtrA2 has a pro-survival function that may involve the regulation of protein quality control (PQC) and mitochondrial homeostasis. Loss of Omi/HtrA2's protease activity causes the neuromuscular disorder of the mnd2 (motor neuron degeneration 2) mutant mice. These mice develop multiple defects including neurodegeneration with parkinsonian features. Loss of Omi/HtrA2 in non-neuronal tissues has also been shown to cause premature aging. The normal function of Omi/HtrA2 in the mitochondria and how its deregulation causes neurodegeneration or premature aging are unknown. Here we report that the mitochondrial Mulan E3 ubiquitin ligase is a specific substrate of Omi/HtrA2. During exposure to H2O2, Omi/HtrA2 degrades Mulan, and this regulation is lost in cells that carry the inactive protease. Furthermore, we show accumulation of Mulan protein in various tissues of mnd2 mice as well as in Omi/HtrA2(−/−) mouse embryonic fibroblasts (MEFs). This causes a significant decrease of mitofusin 2 (Mfn2) protein, and increased mitophagy. Our work describes a new stress-signaling pathway that is initiated in the mitochondria and involves the regulation of Mulan by Omi/HtrA2 protease. Deregulation of this pathway, as it occurs in mnd2 mutant mice, causes mitochondrial dysfunction and mitophagy, and could be responsible for the motor neuron disease and the premature aging phenotype observed in these animals.  相似文献   

19.
Omi/HtrA2 is a pro-apoptotic mitochondrial serine protease involved in both forms of apoptosis, caspase-dependent as well as caspase-independent cell death. However, the impact of Omi/HtrA2 in the apoptotic cell machinery that takes place in vivo under pathological conditions such as cerebral ischemia remains unknown. The present study was monitored in order to examine whether Omi/HtrA2 plays a decisive role in apoptosis observed after focal cerebral ischemia in rats. Male adult rats were subjected to 90min of focal cerebral ischemia followed by reperfusion and treated with vehicle or ucf-101, a novel and specific Omi/HtrA2 inhibitor, prior reperfusion. Focal cerebral ischemia/reperfusion induced a mitochondrial up-regulation of Omi/HtrA2 and significantly increased cytosolic accumulation of Omi/HtrA2. Furthermore, ischemia led to activation of caspase-3 and degradation X-linked inhibitor of apoptosis protein (XIAP). Treatment of animals prior ischemia with ucf-101, the specific inhibitor of Omi/HtrA2, was able to (1) reduce the number of TUNEL-positive cells, to (2) attenuate the XIAP-breakdown and to (3) reduce the infarct size. This study shows for the first time that focal cerebral ischemia in rats results in Omi/HtrA2 translocation from the mitochondria to the cytosol, where it participates in neuronal cell death. Blocking the proteolytic activity of Omi/HtrA2 with specific inhibitors, such as the ucf-101, could be a novel way to afford neuroprotection and minimize cellular damage in cerebral ischemia/reperfusion.  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