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 218 毫秒
1.
TNF受体家族介导的细胞凋亡信号转导   总被引:4,自引:0,他引:4       下载免费PDF全文
肿瘤坏死因子(TNF)家族是一类多功能的细胞因子,具有诱导细胞凋亡、抗病毒、免疫调节等多种生物学活性.其中一些成员可以通过和细胞膜上相应受体(即TNF受体家族成员)结合,启动细胞内的凋亡机制,而诱导细胞凋亡.一些蛋白质(如TRADD、FADD、RIP、RAIDD等)参与这些信号传递过程.越来越多的TNF家族成员、TNF受体以及与细胞凋亡相关的Caspase蛋白酶家族成员被人们发现.  相似文献   

2.
肿瘤坏死因子α(tumor necrosis factor alpha,TNFα)诱导的L929细胞死亡是研究细胞程序性坏死的重要模型,但也有报道称,TNFα处理后的L929细胞发生了凋亡。该研究以所在实验室保存的L929细胞(L929-A)和从商业化细胞库购买的L929细胞(L929-N)为模型,进一步鉴定了TNFα诱导的L929细胞死亡类型与调控机制。结果发现,TNFα处理后的L929-A细胞中出现了凋亡特征,且阻断胱冬肽酶(caspase)信号通路可显著抑制TNFα诱导的L929-A细胞死亡,但却促进TNFα诱导的L929-N细胞死亡。此外,受体相互作用蛋白1(receptor-interacting protein 1,RIP1)在TNFα诱导的两种L929细胞死亡过程中都具有关键性的调控作用,表明TNFα处理后的L929-A细胞发生了RIP1依赖的细胞凋亡,而L929-N细胞发生了程序性坏死(necroptosis)。同时,启动细胞程序性坏死的关键蛋白RIP3(receptor-interacting protein 3)在L929-N细胞中表达水平显著高于L929-A细胞,因此,RIP3的这种差异表达可能是决定两种L929细胞在TNFα处理后发生不同类型细胞程序性死亡的重要原因。  相似文献   

3.
细胞凋亡信号的传递途径及其调控   总被引:8,自引:0,他引:8  
细胞凋亡是不同于细胞坏死的一种细胞死亡方式。在形态上表现为细胞皱缩、染色体边缘化、核碎裂、凋亡小体形成等。目前公认的细胞凋亡指标包括形态学上光镜、电镜观察,DNA电泳的阶梯状条带,流式细胞术测定亚G_1峰等。但这些关于凋亡的研究仅限于明确是否存在凋亡,而无法解释凋亡的发生机制。近年来,随着分子生物学突飞猛进的发展,关于细胞凋亡分子机制的研究有了很大的突破,尤其是FasL和TNF诱导的细胞凋亡。这种细胞凋亡可在刺激后1—2h内发生,而且去除细胞核后,胞浆的凋亡改变可在无核的情况下发生。  相似文献   

4.
目的实验应用Hsp90过表达系统观察Hsp90抑制TNFα诱导的细胞凋亡,探讨其可能的作用机制。方法采用电穿孔技术建立稳定过表达Hsp90的细胞克隆,应用激光共聚焦显微镜和流式细胞仪观察TNFα和放线菌酮诱导的细胞凋亡,应用比色分析和Western blotting方法检测caspase的变化。结果1)在稳定过表达Hsp90的NIH3T3细胞中,Hsp90能够抑制TNFα诱导的细胞凋亡。2)在凋亡信号转导通路中,Hsp90作用于caspase-8下游、caspase-3上游。结论1)Hsp90能够在细胞凋亡信号转导通路中发挥负性调节因子的作用。2)Hsp90抑制剂作为抗肿瘤药物的机制之一,可能是通过促进caspase-3活化而促进肿瘤细胞凋亡。  相似文献   

5.
研制了基因工程靶向融合蛋白XE-TNFαm2.其中,XE为HIV/SIV辅助受体CXCR4的第二胞外域;TNFαm2是经突变改型的TNFα,其毒副作用已降低18倍,已用于临床治疗恶性肿瘤.已有的研究表明XE-TNFαm2的功能之一是杀灭受HIV/SIV感染的细胞、但不杀伤未受HIV/SIV感染的正常细胞.探讨XE-TNFαm2可否引起细胞的凋亡,以阐明其杀伤细胞作用的可能机制.结果表明,XE-TNFαm2只能引起受HIV/SIV感染细胞的凋亡,但不能引起未受HIV/SIV感染的正常细胞的凋亡.这一结果表明XE-TNFαm2是一种可特异杀灭受HIV/SIV感染细胞的准确靶向融合蛋白,其毒副作用己最小化.  相似文献   

6.
TNF相关的凋亡诱导配体(TNF-related apoptosis-induicing ligand,TRAIL),属于TNF超家族成员,与Apo-1L(FasL)有较高的同源性,又称为Apo-2L。TRAIL有两类受体,一类是死亡受体,如DR4和DR5,TRAIL与DR4或DR5结合可以诱导细胞凋亡;另一类是“诱骗”受体,如DcR1、DcR2,可以竞争性地与TRAIL结合,逃避或抑制TRAIL诱导的正常细胞损伤。TRAIL及其受体的发现为肿瘤的治疗提供了一个新的方向。  相似文献   

7.
肿瘤坏死因子信号传导的分子机理   总被引:8,自引:0,他引:8  
肿瘤坏死因子(tumor necrosis factor,TNF)是一种具有多种生物学效应的细胞因子,其生物学效应包括促进细胞生长、分化、凋亡及炎症诱发等.TNF的生物学效应都是通过细胞表面的两种TNF受体引发的.TNF的信号传导通路主要包括细胞凋亡及转录因子NF-kB和JNK蛋白激酶的激活.这3条信号传导通路之间及各通路内部含有各种调节机制,使TNF的各种生物学功能协调发挥出来.从1994年到现在,对肿瘤坏死因子信号传导通路的分子机理研究取得了一系列突破性进展,在细胞信号传导研究领域中树立了成功的典范.  相似文献   

8.
凋亡信号调节激酶1对细胞凋亡的调节作用   总被引:1,自引:0,他引:1  
细胞存活与凋亡之间的平衡是多细胞生物正常发育与稳态的关键。细胞凋亡是受多种因素高度调控的细胞程序性死亡过程。凋亡信号调节激酶 1 (ASK1 )是促分裂原活化蛋白激酶激酶激酶家族的成员之一 ,它分别激活SER1 JNK和MKK3 MKK6 p38途径 ,在细胞因子及应激诱导细胞凋亡过程中起着关键作用。TNF受体和Fas信号转导系统在抗凋亡与促凋亡过程中发挥重要作用 ,其中包括TNF受体Ⅰ相关死亡域蛋白 (TRADD)、Fas相关死亡域蛋白 (FADD)等多种蛋白因子。细胞色素C是线粒体依赖性死亡信号 ,受Bcl 2家族蛋白的调节。反应性氧化合物的氧化激活使硫氧还蛋白 (Trx)氧化 ,并与ASK1分离 ,从而激活ASK1造成细胞凋亡。总之 ,许多促凋亡与抗凋亡因子组成复杂的、相互拮抗的机制。在信号转导的各种不同的关卡上 ,这些因子的平衡作用最终决定细胞的生与死。  相似文献   

9.
探讨了粘着斑激酶(focal adhesion kinase,FAK)在TNF-α和环己亚胺协同诱导SMMC-7721细胞凋亡中的作用,利用转染FAK反义质粒来特异性降低SMMC-7721细胞的FAK含量,用Western杂交的方法来检测蛋白激酶B(PKB)的蛋白含量,以及用流式细胞仪的方法来检测细胞的凋亡。研究发现TNF-α本身并不能诱导SMMC-7721细胞发生凋亡,而只有当用环己亚胺和TNF-α协同处理时才发生凋亡。在TNF-α和环己亚胺协同诱导的凋亡过程中,PKB蛋白含量的降低,提示PKB的蛋白水平与凋亡的发生密切相关。当用FAK反义质粒降低SMMC-7721细胞的FAK含量(约降低了60%)后,细胞对TNF-α和环己亚胺协同诱导的凋亡的敏感性在用低剂量TNF-α处理的情况下增加,而在高剂量TNF-α和处理的情况下降低,相应地,在低剂量TNF-α和环己亚胺处理的情况下,FAK反义质粒转染株细胞的PKB含量比对照的PKB含量低;而在高剂量TNF-α和环己亚胺处理的情况下,FAK反义质粒转染株细胞的PKB含量比对照的要高。结果提示,FAK对TNF-α和环己亚胺协同诱导SMMC-7721凋亡的过程有一双相作用,并且该过程可能与PKB有关。  相似文献   

10.
Fas与FasL的分子生物学研究进展   总被引:1,自引:0,他引:1  
包兆胜 《生命的化学》2002,22(2):127-129
Fas是细胞表面诱导凋亡的分子,是I型膜蛋白,属TNF受体家族成员。而Fas配体(FasL)是Ⅱ型膜蛋白,属TNF家族成员。Fas与FasL结合,可向细胞传递死亡信号,引发细胞凋亡。Fas在人体中表达及功能正常与否,对人体免疫调控起重要作用。  相似文献   

11.
肿瘤坏死因子相关的凋亡诱导配体 (TRAIL)能选择性诱导肿瘤细胞凋亡 .为利用基因工程技术获得重组TRAIL蛋白可溶性片段 (sTRAIL) ,设计 1对引物 .利用PCR技术特异性扩增出sTRAIL的cDNA ,克隆于质粒pGEM 3Zf( )的EcoRⅠ和PstⅠ位点 .经测序证明序列正确后克隆于表达质粒pBV2 2 0的EcoRⅠ和PstⅠ位点 ,转化大肠杆菌DH5α .转化菌株经温度诱导 ,SDS PAGE检测和Western印迹鉴定 ,获得重组sTRAIL的高水平非融合表达菌株 .表达量占菌体总蛋白的 2 0 % .对其表达产物进行了初步纯化 ,SDS PAGE结果显示纯度可达 90 %以上 .用L92 9细胞测定其生物学活性表明 ,重组蛋白在体外能明显诱导肿瘤细胞凋亡  相似文献   

12.
The extracellular portion (amino acids 95–281 or 114–281) of the human tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) was genetically linked to the C terminus of the fluoresce-enhanced green fluorescent protein variant (EGFP) to generate two versions of EGFP–sTRAIL fusion proteins, designated EGFP–sTR95 and EGFP–sTR114, respectively. The two versions of EGFP–sTRAIL fusion proteins both induce extensive apoptosis in lymphoid as well as nonlymphoid tumor cell lines. In addition, the two versions of fusion proteins retain similar fluorescence spectra to those of EGFP and have shown the specific binding to TRAIL receptor-positive cells; thus, the stained cells could be analyzed with flow cytometry. Hence, the two versions of fusion proteins represent a readily obtainable source of biologically active sTRAIL that may prove useful in exploit fully the characteristics of both the soluble TRAIL and its receptor system.  相似文献   

13.
Epidermal growth factor receptor (EGFR) signaling inhibition by monoclonal antibodies and EGFR-specific tyrosine kinase inhibitors has shown clinical efficacy in cancer by restoring susceptibility of tumor cells to therapeutic apoptosis induction. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anti-cancer agent with tumor-selective apoptotic activity. Here we present a novel approach that combines EGFR-signaling inhibition with target cell-restricted apoptosis induction using a TRAIL fusion protein with engineered specificity for EGFR. This fusion protein, scFv425:sTRAIL, comprises the EGFR-blocking antibody fragment scFv425 genetically fused to soluble TRAIL (sTRAIL). Treatment with scFv425:sTRAIL resulted in the specific accretion to the cell surface of EGFR-positive cells only. EGFR-specific binding rapidly induced a dephosphorylation of EGFR and down-stream mitogenic signaling, which was accompanied by cFLIP(L) down-regulation and Bad dephosphorylation. EGFR-specific binding converted soluble scFv425:sTRAIL into a membrane-bound form of TRAIL that cross-linked agonistic TRAIL receptors in a paracrine manner, resulting in potent apoptosis induction in a series of EGFR-positive tumor cell lines. Co-treatment of EGFR-positive tumor cells with the EGFR-tyrosine kinase inhibitor Iressa resulted in a potent synergistic pro-apoptotic effect, caused by the specific down-regulation of c-FLIP. Furthermore, in mixed culture experiments binding (L)of scFv425:sTRAIL to EGFR-positive target cells conveyed a potent apoptotic effect toward EGFR-negative bystander tumor cells. The favorable characteristics of scFv425:sTRAIL, alone and in combination with Iressa, as well as its potent anti-tumor bystander activity indicate its potential value for treatment of EGFR-expressing cancers.  相似文献   

14.
BACKGROUND: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces cell death in various tumor cells, but relatively spares normal cells. Recombinant adeno-associated virus (rAAV) vectors have a number of advantages including in vivo long-term gene expression. Here, we assessed the biological activity of a novel, secreted form of TRAIL (sTRAIL) for cancer gene therapy using a rAAV2 vector. METHODS: A plasmid and rAAV2 vectors were constructed encoding sTRAIL composed of a leader sequence, the isoleucine zipper, and the active domain of TRAIL (aa 95-281). The functionality of sTRAIL was validated by cell viability, FACS analysis, caspase-3 activity, and TUNEL staining. rAAV-sTRAIL was injected intratumorally to nude mice bearing human A549 lung tumor cells. Nude mice received A549 tumor cells after intravenous delivery of rAAV-sTRAIL. The antitumor effect was then evaluated by measuring tumor regression and occurrence in the experimental animal. RESULTS: sTRAIL was released from cells transfected with the sTRAIL expression construct or transduced with rAAV-sTRAIL, and induced apoptosis in cancer cells, but spared normal fibroblast cells. Secreted sTRAIL formed oligomers including trimers with intersubunit disulfide. Purified sTRAIL exerted much lower cytotoxicity on primary human hepatocytes compared to recombinant TRAIL. Intratumoral delivery of rAAV-sTRAIL significantly inhibited growth of A549 tumors established in nude mice. A number of apoptotic tumor cells were detected by TUNEL staining in mice treated with rAAV-sTRAIL. Systemic pretreatment with rAAV-sTRAIL significantly inhibited tumor formation in nude mice. CONCLUSION: The results suggest that rAAV-sTRAIL may be useful for local or systemic cancer gene therapy for treating TRAIL-sensitive tumors.  相似文献   

15.
Mesenchymal stem cells (MSCs) are able to infiltrate tumor tissues and thereby effectively deliver gene therapeutic payloads. Here, we engineered murine MSCs (mMSCs) to express a secreted form of the TNF-related apoptosis-inducing ligand (TRAIL), which is a potent inducer of apoptosis in tumor cells, and tested these MSCs, termed MSC.sTRAIL, in combination with conventional chemotherapeutic drug treatment in colon cancer models. When we pretreated human colorectal cancer HCT116 cells with low doses of 5-fluorouracil (5-FU) and added MSC.sTRAIL, we found significantly increased apoptosis as compared with single-agent treatment. Moreover, HCT116 xenografts, which were cotreated with 5-FU and systemically delivered MSC.sTRAIL, went into remission. Noteworthy, this effect was protein 53 (p53) independent and was mediated by TRAIL-receptor 2 (TRAIL-R2) upregulation, demonstrating the applicability of this approach in p53-defective tumors. Consequently, when we generated MSCs that secreted TRAIL-R2-specific variants of soluble TRAIL (sTRAIL), we found that such engineered MSCs, labeled MSC.sTRAILDR5, had enhanced antitumor activity in combination with 5-FU when compared with MSC.sTRAIL. In contrast, TRAIL-resistant pancreatic carcinoma PancTu1 cells responded better to MSC.sTRAILDR4 when the antiapoptotic protein XIAP (X-linked inhibitor of apoptosis protein) was silenced concomitantly. Taken together, our results demonstrate that TRAIL-receptor selective variants can potentially enhance the therapeutic efficacy of MSC-delivered TRAIL as part of individualized and tumor-specific combination treatments.  相似文献   

16.
The extracellular domain of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) may function as a soluble cytokine to selectively kill various cancer cells without toxicity to most normal cells. We used a high-biosafety plasmid pVAX1 as a vector and constructed a recombinant plasmid expressing the extracellular domain (95-281 aa) of human TRAIL fused with signal peptides of human IgGgamma, designated as pVAX-sT. Transduction of human BEL7402 liver cancer cells with pVAX-sT led to high levels of sTRAIL protein in the cell culture media and induced apoptosis. The therapeutic potential of pVAX-sT was then evaluated in the BEL7402 transplanted naked mouse model. Subsequent intratumoral administration of naked pVAX-sT resulted in the expression of soluble TRAIL in the sera and the tumor site, as well as effective suppression of tumor growth, with no toxicity to liver. In conclusion, the successful inhibition of liver cancer growth and the absence of detectable toxicity suggest that pVAX-sT could be useful in the gene therapy of liver cancer.  相似文献   

17.
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in malignant cells, including gliomas, and is currently in anticancer clinical trials. However, the full-length and tagged forms of TRAIL, unlike the untagged ligand (soluble TRAIL (sTRAIL)), exhibits toxicity against normal cells. Here, we report the generation and testing of an adenovirus (AdsTRAIL) that expresses untagged sTRAIL in an intracranial xenograft model and a human glioma organotypic slice culture model. AdsTRAIL efficiently induced apoptosis in glioma cell lines, including those resistant to sTRAIL, but not in normal human astrocytes (NHAs). It inhibited anchorage-independent glioma growth and exerted a bystander effect in transwell assays. Intratumoral injections of AdsTRAIL in a rodent intracranial glioma model resulted in reduced tumor growth and improved survival compared with Ad-enhanced green fluorescent protein (EGFP)- or vehicle-treated controls without toxicity. Human glioma organotypic slices treated with AdsTRAIL demonstrated apoptosis induction and caspase activation.  相似文献   

18.
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a new member of the TNF superfamily. Here, a recombinant form of the extracellular domain of the TRAIL (sTRAIL) was expressed in Escherichia coli BL21(DE3) under the control of a T7 promoter. The resulting insoluble bodies were separated from cellular debris by centrifugation and solubilized with 8 M urea. A rapid and simple on-column refolding procedure was developed. It was applied and then the refolded sTRAIL was purified by anion-exchange chromatography. The purified final product was >98% pure by SDS-PAGE stained with Coomassie brilliant blue R-250. Mass spectroscopic analysis indicated the protein to be 19.2 kDa, which equalled the theoretically expected mass. N-terminal sequencing of refolding sTRAIL showed the sequence which corresponded to the designed protein. The renatured protein displayed its immunoreactivity with the antibodies to TRAIL protein by Western blotting. The purified sTRAIL had a strong cytotoxic activity against human cervical cancer HeLa cells with ED50 about 1.5 mg/L. Circular dichroism and fluorescence spectrum analysis showed that the refolded sTRAIL had a structure similar to that of native protein with beta-sheet secondary structure. This efficient procedure of sTRAIL renaturation may be useful for the mass production of this therapeutically important protein.  相似文献   

19.
肿瘤坏死因子相关凋亡诱导配体(TNF-related apoptosis inducing ligand, TRAIL) 是TNF超家族中的成员,能够广泛诱导肿瘤细胞凋亡,对正常细胞无明显毒副作用. TRAIL已成为肿瘤治疗领域的研究热点.人脑胶质瘤是神经系统肿瘤中最常见类型, 占颅内肿瘤50%~60%,5年存活率为20%~30%. 本研究探讨可溶性TRAIL蛋白对人脑胶质瘤细胞(U251)的抑制作用. 由大肠杆菌表达系统表达的TRAIL多为包涵体,为获得可溶性的蛋白,将hTRAIL95~281功能区基因片段插入到pHisSUMO表达载体,经IPTG低温诱导表达,Ni-NTA Agarose纯化后获得可溶性SUMO-hTRAIL,经SUMO ProteaseⅠ切去SUMO融合标签后获得成熟可溶hTRAIL蛋白. 以U251细胞为靶细胞,通过MTT法检测TRAIL对肿瘤细胞的抑制作用.结果证明,TRAIL对U251细胞的抑制呈剂量依赖关系,最大抑制率为53.9%.流式细胞仪检测TRAIL诱导U251细胞凋亡实验中,对照组细胞存活率为92.2±0.8%,实验组细胞存活率为35.5±1.2%,证明重组蛋白具有生物学活性,并在体外能明显诱导U251肿瘤细胞发生死亡.本研究结果为TRAIL蛋白在临床上应用于肿瘤治疗奠定了基础.  相似文献   

20.
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), as an anticancer protein with tumor-selective apoptotic activity, has been examined for use in clinical application. Melittin, an antibacterial peptide isolated from the bee Apis mellifera, has shown strong cytotoxicity to both tumor and normal cells. To ameliorate the cytotoxicity of melittin on cells and enhance the activity of TRAIL on cancer cells, we constructed a novel fusion protein, sTRAIL–melittin, containing a small ubiquitin-related modifier (SUMO) tag and expressed this fusion protein in Escherichia coli. Data showed that expression of the soluble fusion protein with the SUMO tag was approximately 85 % of total target protein which was much higher than that without the SUMO tag (approximately 10 %); sTRAIL–melittin was easily purified using Ni-NTA affinity chromatography and the tag was removed easily using SUMO-specific protease. To assay anticancer activity and side effects, methyl thiazolyl tetrazolium, hemolytic, and apoptosis assays were employed. Results demonstrated that sTRAIL–melittin had cytotoxic and apoptotic activity in K562 leukemia cells and HepG2 liver carcinoma cells, while it had only a minimal effect on erythrocytes and normal HEK293 cells. This indicates that the cytotoxicity of sTRAIL–melittin in normal cells was low and the anticancer activity of the fusion protein in tumor cells was significantly enhanced compared with sTRAIL (P?<?0.01). Furthermore, we found that sTRAIL–melittin also showed antibacterial activity to Staphylococcus aureus due to the presence of the melittin domain. Therefore, TRAIL fused with an antibacterial peptide may be a promising novel TRAIL-based anticancer treatment strategy.  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