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 78 毫秒
1.
EB病毒LMP1及其CTAR1、CTAR2导入人HNE2鼻咽癌细胞的研究   总被引:4,自引:2,他引:2  
用电穿孔转染法,建立稳定表达野生型LMP1及其不同突变体的鼻咽癌细胞系,并以这些细胞系为材料,用MTT法检测增殖期活细胞,观察LMP1及其不同的结构域对鼻咽癌细胞生长的影响。结果得到了LMP1及其三种突变体、空白载体表达的鼻咽癌细胞系;HNE2-LMP1(野生型)、HNE2-LMP1△185-351(CTAR1缺失型)、HNE2-LMP1(1-231)(CTAR2缺失型)、HNE2-LMP1(1-185)(羧基端胸浆区缺失型)、HNE2-pSG5(空载体型)。进一步证实HNE2-LMP1、HNE2-LMP1(1-231)、HNE2-LMP1△185-351平均吸光度(A)比值高于对照组HNE2-pSG5及HNE2(P<0.01)。这提示:EB病毒LMP1及其LMP1(1-231)和LMP1△185-351在体外明显促进HNE2细胞增殖。结果表明EB病毒LMP1可能在鼻咽癌中发挥着重要的作用。  相似文献   

2.
为了探讨EB病毒潜伏膜蛋白1(LMP1)的致瘤机制,对鼻咽癌中LMP1激活重要的核转录因子NF-κB机制进行了研究.首先,采用免疫共沉淀-蛋白质印迹在稳定表达LMP1的鼻咽癌细胞系HNE2-LMP1中证实LMP1与TRAF1,2,3结合形成免疫共沉淀复合物,进一步以野生型LMP1及其三种突变体的鼻咽癌细胞系LMP1(野生型,wt)、HNE2-LMP1 del187~351(CTAR1缺失型)、HNE2-LMP1(1~231)(CTAR2缺失型)、HNE2-LMP1(1~187)(羧基端胞浆区缺失型)、HNE2-pSG5(空白载体型)为材料,结合NF-κB报道基因质粒(pGL2-NF-κB-luc)的荧光素酶活性表达分析NF-κB的活性,证实:较之母细胞, 野生型LMP1活化NF-κB达13.8倍, LMP1(1~187)几乎不活化NF-κB,LMP1(1~231)活化NF-κB达4.9倍, LMP1(del187~351)活化NF-κB达9.1倍;TRAF1过表达升高LMP1(wt)及LMP1(1~231)介导的NF-κB活性,而对LMP1(del 187~351)活化NF-κB无影响;TRAF3过表达或TRAF3负显性突变体抑制LMP1(wt)及LMP1(1~231)介导的NF-κB活性,而不影响LMP1(del 187~351)活化NF-κB; TRAF2过表达升高LMP1(wt)、LMP1 (1~231)及LMP1(del 187~351)介导的NF-κB活性.这些结果表明:鼻咽癌中LMP1通过TRAF1、TRAF2或TRAF3调控NF-κB,TRAF1和TRAF3主要通过CTAR1发挥作用,TRAF2的作用主要是通过CTAR1和CTAR2介导的.  相似文献   

3.
EB病毒潜伏膜蛋白1通过结合TRAFs调控NF-κB   总被引:2,自引:1,他引:1  
为了探讨EB病毒潜伏膜蛋白1(LMP1)的致瘤机制,对鼻咽癌中LMP1激活重要的核转录因子NF-κB机制进行了研究,首先,采用免疫共沉淀-蛋白质印迹在稳定表达LMP1的鼻咽癌细胞系HNE2-LMP1中证实LMP1与TRAF1,2,3结合形成免疫共沉淀复合物,进一步以野生型LMP1及其三种突变体的鼻咽癌细胞系LMP1(野生型, wt),HNE2-LMP1 del187-351(CTAR1缺失型),HNE2-LMP1(1-231),(CTAR2缺失型),HNE2-LMP1(1-187)(羰基端胞浆区缺失型),HNE2-pSG5(空白载体型)为材料,结合NF-κB报道基因质粒(pG12-NF-B-luc)的荧光素酶活性表达分析NF-κB的活性,证实:较之母细胞,野生型LMP1活化NF-B达13.8倍,LMP1(1-187)几乎不活化NF-kb,LMP1(1-231)活化NF-kB 达4.9倍,LMP1(del187-351)活化NFκB达9.1倍,TRAF1过表达升高LMP1( wt)及LMP1(1-231)介导的NF-κB活性,而对LMP1(del187-351)活化NFκB无影响,TRAF3过表达或TRAF3负显性突变体抑,制LMP1(wt)及LMP1(1-231)介导的NF-κB活性而不影响LMP1(del187-351)活化NF-κB,TRAF2过表达升高LMP1(wt),LMP1(1-231)及LMP1(del 187-351)介导的NF-kB活性,这些结果表明:鼻咽癌中LMP1通过TRAF1,TRAF2或TRAF3调控NF-kB,TRAF1和TRAF3主要通过CTAR1发挥作用,TRAF2的作用主要是通过CTAR1和CTAR2介导的。  相似文献   

4.
EB病毒潜伏膜蛋白1通过TRAF/TRADD激活JNK信号途径   总被引:12,自引:3,他引:9  
为了探讨在鼻咽癌细胞中EB病毒编码的潜伏膜蛋白1(LMP1)激活c-Jun氨基端激酶(JNK)信号途径的分子机制,利用可调控表达LMP1的鼻咽癌细胞系L7,蛋白质印迹检测,发现LMP1能够促进JNK的活化;利用稳定表达LMP1的鼻咽癌细胞系HNE2-LMP1及其三种突变体HNE2-LMP1ΔCTAR1、HNE2-LMP1ΔCTAR2、HNE2-LMP1ΔCTAR1,2及LMP1阴性的HNE2为材料,采用蛋白质印迹和报告基因法分析JNK和活化蛋白1(AP1)活化情况,结果显示HNE2-LMP1和HNE2-LMP1ΔCTAR1中磷酸化JNK蛋白表达量和AP1活性都无显著差异,而与HNE2-LMP1ΔCTAR2、HNE2-LMP1ΔCTAR1,2、阴性对照HNE2及空白载体转染细胞的JNK蛋白表达和AP1活性具有显著差异;进一步比较转染TRAF、TRADD显性负性突变体鼻咽癌细胞系HNE2-LMP1中磷酸化的JNK量和AP1活性,结果显示:TRAF-DN和TRADD-DN的导入使活化的JNK蛋白和AP-1活性显著降低,二者间无显著差异,提示TRAF和TRADD可能参与了LMP1对JNK和AP-1的活化.以上结果提示在鼻咽癌细胞系中LMP1功能结构域CTAR2通过结合TRAF/TRADD激活JNK从而活化重要的转录因子AP1.  相似文献   

5.
EB病毒LMP1-CTAR3对NP69细胞增殖和蛋白质表达的影响   总被引:1,自引:0,他引:1  
为了探讨EB病毒潜伏性膜蛋白1(LMP1)第三个功能活性区域(CTAR3)在鼻咽上皮细胞NP69中的转化作用机制,采用逆病毒感染的方法,将浓缩的逆病毒RV-LMP1和RV-LMP1△232~351分别感染鼻咽上皮细胞NP69,建立NP69-LMP1与NP69-LMP1△232~351稳定表达细胞系.通过绘制生长曲线、平皿克隆形成试验和软琼脂集落形成试验比较野生型和突变型LMP1对NP69细胞增殖的影响,运用蛋白质组学方法鉴定NP69-LMP1与NP69-LMP1△232~351细胞间的差异表达蛋白,选用实时荧光定量RT-PCR与Western blot对其中部分蛋白质点差异表达进行验证.结果发现:a.突变型LMP1△232~351促NP69细胞增殖的能力较野生型LMP1明显降低(n=3,P<0.05);b.鉴定了LMP1-CTAR3在NP69细胞中参与调节的16个蛋白质(表达上调的蛋白质8个,下调的8个).c.实时荧光定量RT-PCR和Western blot证实了部分上述蛋白质的差异表达.以上结果说明,LMP1-CTAR3是其发挥促细胞增殖的重要活性部位,可能通过参与调节G蛋白和异柠檬酸脱氢酶等蛋白质的表达而起作用.  相似文献   

6.
为了观察潜伏性膜蛋白1(LMP1)羧基端活性区3(CTAR3)对鼻咽癌干细胞SP18迁移与侵袭的影响,本研究通过建立稳定表达LMP1及CTAR3突变型LMP1(LMP1△252-351)的SP18细胞系(即SP18-LMP1和SP18-LMP1△252-351),观察LMP1-CTAR3缺失突变后对SP18细胞增殖、迁移与侵袭的影响.采用基因芯片分析SP18-LMP1和SP18-LMP1△252-351间的差异表达基因,并验证基因的表达,用生物信息学分析差异表达基因间的相互关系.结果显示:a.SP-LMP1△252-351细胞生长速度较SP-LMP1细胞明显变缓,克隆形成和迁移与侵袭能力降低(n=3,P0.05);b.鉴定出LMP1羧基端CTAR3影响SP18细胞迁移与侵袭的18个基因(其中表达上调基因13个,下调基因5个),经荧光定量PCR验证与基因芯片检测结果基本一致.c.13个差异基因间相互联系,网络节点联系最多的基因是FN1、MMP14、THBS1、ITGA2、IL1B和IL6基因.结果提示,LMP1羧基端CTAR3可能通过调节FN1、MMP14、THBS1、ITGA2、IL1B和IL6基因的表达,发挥其促鼻咽癌干细胞SP18细胞迁移与侵袭的功能.  相似文献   

7.
在EB病毒潜伏膜蛋白LMP1介导的信号传导通路中,TRAFs作为LMP1活化的第一位信号分子,可能扮演着重要的分子开关角色。令人关注的是,在上皮性肿瘤NPC的发生中,EB病毒LMP1能否激活重要的TRAFs信号分子?究竟激活何种TRAFs信号分子,激活的机制何在?将LMP1cDNA导入LMP1表达阴性的HNE2中,建立稳定表达LMP1的鼻咽癌细胞系HNE2-LMP1。以此为材料,应用差异RT-PCR和Western blotting法证实,无论在RNA水平,还是蛋白水平上,TRAF1在HNE2-LMP1中表达较HNE2强,而TRAF2及TRAF3在HNE2-LMP1与HNE2细胞中表达无明显差异;进一步用免疫共沉淀-Western blotting证实LMP1可使TRAF1、TRAF2、TRAF3磷酸化而被活化。这些结果提示在鼻咽癌中,LMP1可能诱导TRAF1表达,而对TRAF2及TRAF3并不影响,但LMP1可磷酸化TRAF1、TRAF2、TRAF3而使其功能性活化。q  相似文献   

8.
探讨了EB病毒编码的潜伏膜蛋白1(LMP1)是否通过STAT3调控诱导血管内皮细胞生长因子(VEGF)的表达.利用蛋白质印迹的方法对HNE2、HNE2-LMP1以及瞬时转染STAT3显性负性突变体STAT3β的HNE2-LMP1细胞中VEGF含量进行检测,发现LMP1可以上调VEGF的表达,而STAT3β可以抑制VEGF的上调;利用LMP1可控表达细胞系tet-on-LMP1-HNE2进行LMP1时间和剂量诱导表达研究,发现VEGF可以随LMP1的动态表达而表达;将VEGF野生型报告基因和VEGF潜在的STAT3转录因子突变体报告基因与LMP1表达载体分别共转染研究发现,LMP1可以激活VEGF的转录,这种转录通过VEGF启动子区STAT3转录因子的结合位点发挥作用;电泳迁移率变动分析(EMSA)确证了STAT3的这种DNA位点的特异性活性.结果表明:EB病毒编码的LMP1 在鼻咽癌细胞中可以增加VEGF的转录和表达,并能通过VEGF启动子区STAT3转录因子结合位点发挥作用.  相似文献   

9.
LPLUNC1在正常的鼻咽组织及人胚鼻咽组织中高表达,而在71%的鼻咽癌中表达下调或缺失,是与鼻咽癌的发生发展密切相关的新基因.通过研究LPLUNC1基因对鼻咽癌细胞系HNE1的影响,进一步确定其与鼻咽癌发生发展的关系.将LPLUNC1基因全长cDNA克隆入pcDNA3.1( )真核表达载体中,通过脂质体介导稳定转染入LPLUNC1低表达鼻咽癌细胞系HNE1中,通过RT-PCR及RNA印迹筛选LPLUNC1高表达的细胞株,并利用细胞生长曲线、MTT、BrdU掺入、流式细胞仪检测、软琼脂集落形成实验及裸鼠成瘤等实验,研究了LPLUNC1对鼻咽癌细胞系HNE1细胞生长、增殖的影响.结果发现,稳定转染LPLUNC1的HNE1细胞的生长速度明显减慢,在MTT与BrdU掺入实验发现LPLUNC1可明显地抑制鼻咽癌细胞的增殖,并且通过流式细胞仪检测也发现,LPLUNC1基因可明显延缓HNE1细胞的细胞周期进程,使G0/G1期细胞增多而S期细胞相对减少.进一步通过软琼脂集落形成及裸鼠成瘤实验发现,LPLUNC1稳定转染后的HNE1细胞集落形成率与集落的大小均小于空白载体细胞,同时能明显地抑制HNE1细胞的体外成瘤.结果表明,LPLUNC1基因能明显抑制鼻咽癌细胞HNE1的生长增殖,是鼻咽癌发生发展中的重要候选抑瘤基因之一.  相似文献   

10.
鼻咽癌细胞中EB病毒编码的潜伏膜蛋白1活化cyclinD1的表达   总被引:20,自引:1,他引:19  
为了探讨EB病毒编码的潜伏膜蛋白1(EBV-LMP1)促进细胞增殖,参与EBV相关疾病致瘤的分子机制,研究了LMP1在鼻咽癌细胞中调节cyclinD1表达,进而影响细胞周期行进及细胞恶性表型改变,并初步确定了LMP1发挥该功能的结构域.利用已建株的Tet-on-LMP1-HNE2鼻咽癌细胞系,蛋白质印迹实验分析LMP1诱导cyclinD1蛋白质表达的表达动力学,包括时间效应及剂量效应;利用三种LMP1功能区缺失的突变体及野生型LMP1,以载体型细胞为对照,确定LMP1活化cyclinD1表达的结构域.同时结合基因诱导表达及反义寡聚核酸技术阻断基因表达的实验方法,进一步确定LMP1上调的cyclinD1功能,即对细胞周期行进及细胞恶性表型的影响.结果表明LMP1确实可以诱导cyclinD1的表达(2~4倍),且诱导具有时间依赖性及剂量依赖性;利用三种LMP1功能区缺失的突变体及野生型LMP1,以载体型细胞为对照,结合报道基因分析法,确定与空白载体细胞系比较,野生型LMP1从转录水平可反式激活cyclinD1报道基因活性约11.2倍,其中CTAR1及CTAR2均可活化cyclinD1表达,但以CTAR2为主,与野生型LMP1诱导cyclinD1反式激活活性比较,CTAR1缺失导致cyclinD1报道基因活性下降23.6%,CTAR2缺失导致cyclinD1活性下降约80.7%,C端均缺失时cyclinD1活性只有野生型的17.7%.流式细胞仪分析显示,强力霉素诱导后cyclinD1高表达的细胞停留于G0/G1期明显减少,较未经诱导的细胞,从66.42%减至56.55%,而进入S期及G2/M期的细胞明显增多.在稳定表达LMP1的细胞中,与导入正义LMP1比较,导入反义LMP1 PS-ODNs及反义cylinD1,可以使细胞软琼脂集落形成率明显降低(从30.48%分别降至15.21%,21.76%).EBV-LMP1可以活化cyclinD1的表达,且发挥这种功能的结构域以CTAR2为主,活化的cyclinD1参与细胞周期行进,抑制LMP1及cyclinD1的表达均可导致细胞软琼脂集落形成率降低.  相似文献   

11.
The Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) gene is considered the EBV oncogene as it is necessary for EBV-mediated transformation of B lymphocytes and itself transforms rodent fibroblasts. LMP1 activates the NF-kappaB, phosphatidylinositol 3-kinase (PI3K)-Akt, mitogen-activated protein kinase, and Jun N-terminal protein kinase signaling pathways through its two signaling domains, carboxyl-terminal activating regions 1 and 2 (CTAR1 and CTAR2). CTAR1 and CTAR2 induce signal transduction pathways through their direct (CTAR1) or indirect (CTAR2) recruitment of tumor necrosis factor receptor-associated factors (TRAFs). CTAR1 is necessary for LMP1-mediated transformation as well as activation of PI3K signaling and induction of cell cycle markers associated with G(1)/S transition. In this study, activation of PI3K-Akt signaling and deregulation of cell cycle markers were mapped to the TRAF-binding domain within CTAR1 and to the residues between CTAR1 and CTAR2. LMP1 CTAR1 also activated the MEK1/2-extracellular signal-regulated kinase 1/2 signaling pathway, and this activation was necessary for LMP1-induced transformation of Rat-1 fibroblasts. Dominant-negative forms of TRAF2 and TRAF3 inhibited but did not fully block LMP1-mediated transformation. These findings identify a new signaling pathway that is uniquely activated by the TRAF-binding domain of LMP1 and is required for transformation.  相似文献   

12.
13.
14.
Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) transforms rodent fibroblasts and is expressed in most EBV-associated malignancies. LMP1 (transformation effector site 2 [TES2]/C-terminal activation region 2 [CTAR2]) activates NF-κB, p38, Jun N-terminal protein kinase (JNK), extracellular signal-regulated kinase (ERK), and interferon regulatory factor 7 (IRF7) pathways. We have investigated LMP1 TES2 genome-wide RNA effects at 4 time points after LMP1 TES2 expression in HEK-293 cells. By using a false discovery rate (FDR) of <0.001 after correction for multiple hypotheses, LMP1 TES2 caused >2-fold changes in 1,916 mRNAs; 1,479 RNAs were upregulated and 437 were downregulated. In contrast to tumor necrosis factor alpha (TNF-α) stimulation, which transiently upregulates many target genes, LMP1 TES2 maintained most RNA effects through the time course, despite robust and sustained induction of negative feedback regulators, such as IκBα and A20. LMP1 TES2-regulated RNAs encode many NF-κB signaling proteins and secondary interacting proteins. Consequently, many LMP1 TES2-regulated RNAs encode proteins that form an extensive interactome. Gene set enrichment analyses found LMP1 TES2-upregulated genes to be significantly enriched for pathways in cancer, B- and T-cell receptor signaling, and Toll-like receptor signaling. Surprisingly, LMP1 TES2 and IκBα superrepressor coexpression decreased LMP1 TES2 RNA effects to only 5 RNAs, with FDRs of <0.001-fold and >2-fold changes. Thus, canonical NF-κB activation is critical for almost all LMP1 TES2 RNA effects in HEK-293 cells and a more significant therapeutic target than previously appreciated.  相似文献   

15.
Epstein-Barr virus (EBV) latently infects most of the human population and is strongly associated with lymphoproliferative disorders. EBV encodes several latency proteins affecting B cell proliferation and survival, including latent membrane protein 2A (LMP2A) and the EBV oncoprotein LMP1. LMP1 and LMP2A signaling mimics CD40 and BCR signaling, respectively, and has been proposed to alter B cell functions including the ability of latently-infected B cells to access and transit the germinal center. In addition, several studies suggested a role for LMP2A modulation of LMP1 signaling in cell lines by alteration of TRAFs, signaling molecules used by LMP1. In this study, we investigated whether LMP1 and LMP2A co-expression in a transgenic mouse model alters B cell maturation and the response to antigen, and whether LMP2A modulates LMP1 function. Naïve LMP1/2A mice had similar lymphocyte populations and antibody production by flow cytometry and ELISA compared to controls. In the response to antigen, LMP2A expression in LMP1/2A animals rescued the impairment in germinal center generation promoted by LMP1. LMP1/2A animals produced high-affinity, class-switched antibody and plasma cells at levels similar to controls. In vitro, LMP1 upregulated activation markers and promoted B cell hyperproliferation, and co-expression of LMP2A restored a wild-type phenotype. By RT-PCR and immunoblot, LMP1 B cells demonstrated TRAF2 levels four-fold higher than non-transgenic controls, and co-expression of LMP2A restored TRAF2 levels to wild-type levels. No difference in TRAF3 levels was detected. While modulation of other TRAF family members remains to be assessed, normalization of the LMP1-induced B cell phenotype through LMP2A modulation of TRAF2 may be a pathway by which LMP2A controls B cell function. These findings identify an advance in the understanding of how Epstein-Barr virus can access the germinal center in vivo, a site critical for both the genesis of immunological memory and of virus-associated tumors.  相似文献   

16.
Latent membrane protein 2A (LMP2A) and LMP2B are viral proteins expressed during Epstein-Barr virus (EBV) latency in EBV-infected B cells both in cell culture and in vivo. LMP2A has important roles in modulating B-cell receptor (BCR) signal transduction by associating with the cellular tyrosine kinases Lyn and Syk via specific phosphotyrosine motifs found within the LMP2A N-terminal tail domain. LMP2A has been shown to alter normal BCR signal transduction in B cells by reducing levels of Lyn and by blocking tyrosine phosphorylation and calcium mobilization following BCR cross-linking. Although little is currently known about the function of LMP2B in B cells, the similarity in structure between LMP2A and LMP2B suggests that they may localize to the same cellular compartments. To investigate the function of LMP2B, B-cell lines expressing LMP2A, LMP2B, LMP2A/LMP2B, and the relevant vector controls were analyzed. As was previously shown, cells expressing LMP2A had a dramatic block in normal BCR signal transduction as measured by calcium mobilization and tyrosine phosphorylation. There was no effect on BCR signal transduction in cells expressing LMP2B. Interestingly, when LMP2B was expressed in conjunction with LMP2A, there was a restoration of normal BCR signal transduction upon BCR cross-linking. The expression of LMP2B did not alter the cellular localization of LMP2A but did bind to and prevent the phosphorylation of LMP2A. A restoration of Lyn levels, but not a change in LMP2A levels, was also observed in cells coexpressing LMP2B with LMP2A. From these results, we conclude that LMP2B modulates LMP2A activity.  相似文献   

17.
Several lines of evidence are compatible with the hypothesis that Epstein-Barr virus (EBV) nuclear antigen 2 (EBNA-2) or leader protein (EBNA-LP) affects expression of the EBV latent infection membrane protein LMP1. We now demonstrate the following. (i) Acute transfection and expression of EBNA-2 under control of simian virus 40 or Moloney murine leukemia virus promoters resulted in increased LMP1 expression in P3HR-1-infected Burkitt's lymphoma cells and the P3HR-1 or Daudi cell line. (ii) Transfection and expression of EBNA-LP alone had no effect on LMP1 expression and did not act synergistically with EBNA-2 to affect LMP1 expression. (iii) LMP1 expression in Daudi and P3HR-1-infected cells was controlled at the mRNA level, and EBNA-2 expression in Daudi cells increased LMP1 mRNA. (iv) No other EBV genes were required for EBNA-2 transactivation of LMP1 since cotransfection of recombinant EBNA-2 expression vectors and genomic LMP1 DNA fragments enhanced LMP1 expression in the EBV-negative B-lymphoma cell lines BJAB, Louckes, and BL30. (v) An EBNA-2-responsive element was found within the -512 to +40 LMP1 DNA since this DNA linked to a chloramphenicol acetyltransferase reporter gene was transactivated by cotransfection with an EBNA-2 expression vector. (vi) The EBV type 2 EBNA-2 transactivated LMP1 as well as the EBV type 1 EBNA-2. (vii) Two deletions within the EBNA-2 gene which rendered EBV transformation incompetent did not transactivate LMP1, whereas a transformation-competent EBNA-2 deletion mutant did transactivate LMP1. LMP1 is a potent effector of B-lymphocyte activation and can act synergistically with EBNA-2 to induce cellular CD23 gene expression. Thus, EBNA-2 transactivation of LMP1 amplifies the biological impact of EBNA-2 and underscores its central role in EBV-induced growth transformation.  相似文献   

18.
Latent membrane protein 2A (LMP2A) is expressed in latent Epstein-Barr virus (EBV) infection. We have demonstrated that Nedd4 family ubiquitin-protein ligases (E3s), AIP4, WWP2/AIP2, and Nedd4, bind specifically to two PY motifs present within the LMP2A amino-terminal domain. In this study, LMP2A PY motif mutant viruses were constructed to investigate the role of the LMP2A PY motifs. AIP4 was found to specifically associate with the LMP2A PY motifs in EBV-transformed lymphoblastoid cell lines (LCLs), extending our original observation to EBV-infected cells. Mutation of both of the LMP2A PY motifs resulted in an absence of binding of AIP4 to LMP2A, which resulted in an increase in the expression of Lyn and the constitutive hyperphosphorylation of LMP2A and an unknown 120-kDa protein. In addition, there was a modest increase in the constitutive phosphorylation of Syk and an unidentified 60-kDa protein. These results indicate that the PY motifs contained within LMP2A are important in regulating phosphorylation in EBV-infected LCLs, likely through the regulation of Lyn activity by specifically targeting the degradation of Lyn by ubiquination by Nedd4 family E3s. Despite differences between PY motif mutant LCLs and wild-type LCLs, the PY motif mutants still exhibited a block in B-cell receptor (BCR) signal transduction as measured by the induction of tyrosine phosphorylation and BZLF1 expression following BCR activation. EBV-transformed LCLs with mutations in the PY motifs were not different from wild-type LCLs in serum-dependent cell growth. Protein stability of LMP1, which colocalizes with LMP2A, was not affected by the LMP2A-associated E3s.  相似文献   

19.
Recent cDNA cloning and sequencing of two Epstein-Barr virus (EBV)-specific mRNAs from latently infected cultures revealed that these RNAs are encoded across the fused terminal repeats of the viral genome and that they are likely to encode two nearly identical proteins with the same transmembrane domains. The smaller predicted protein (LMP2B) lacks 119 amino-terminal amino acids found in the larger one (LMP2A). To test whether these proteins are expressed in latently infected lymphocytes, antibodies to the LMP2 proteins were derived by immunizing rabbits with TrpE-LMP2A fusion proteins. Affinity-purified LMP2-specific antibodies recognized 54- and 40-kilodalton proteins, corresponding to LMP2A and LMP2B, in immunoblots of rodent fibroblasts stably transfected with eucaryotic expression plasmids containing either the LMP2A or LMP2B cDNA. Similar-size proteins were also identified in immunoblots of latently infected lymphocytes. LMP2A localized to membranes in cellular fractionation studies. In immunofluorescent studies, LMP2 localized in the plasma membrane of EBV-infected lymphocytes, with the majority of reactivity confined to the region of the LMP1 patch. This reactivity was detected in almost all lymphoblastoid cells latently infected with EBV.  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