首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到17条相似文献,搜索用时 161 毫秒
1.
LRRC4是一个脑组织相对特异性表达基因,是采用表达序列标签(EST)介导的定位-候选克隆策略结合5′-RACE技术从染色体7q31~32区域克隆出来的一个富亮氨酸重复(LRR)超家族的新成员.LRRC4是神经系统发育与轴突生长的功能基因.LRRC4的表达与胶质瘤的级别进展呈负相关,其表达缺失参与脑胶质瘤进展的晚期事件.LRRC4能够下调一系列神经生长因子或受体(如IGF,EGF,PDGF,CNTF,bFGF,GDNF和BDNF等)的表达,通过调控多种信号转导通路(K-Ras/c-Raf/ERK/MAPK,PI-3K/AKT/NF-κB,p70S6/PKC,STAT3以及JNK2/c-Jun/mp53等)将U251细胞阻滞在G1晚期,抑制脑胶质瘤细胞的增殖和侵袭,而且这种抑制作用依赖于它的LRR结构域.LRRC4不能诱导胶质瘤细胞的凋亡,而是诱导胶质瘤细胞向胶质样细胞分化.  相似文献   

2.
LRRC4是我室自主克隆的一个脑组织优势表达基因.前期研究结果表明,外源性LRRC4基因转染至U251细胞,可明显地抑制U251细胞的增殖、黏附、趋化和侵袭等生物学行为. 因此,LRRC4亦是一个脑胶质瘤抑制性基因.为了进一步了解LRRC4在胶质瘤发生发展中的调控作用,本研究采用荧光差异凝胶电泳(2D-DIGE)和质谱分析技术获得了LRRC4转染U251细胞的11个差异表达蛋白质,并用Western 印迹证实了U251细胞在转染LRRC4基因前后热休克蛋白27、stathmin 1和S100钙结合蛋白A11的差异表达变化. 这些差异蛋白质涉及细胞代谢、增殖、转录、信号转导等众多事件,表明LRRC4基因转染U251细胞后可能通过调控这些蛋白质的表达而参与细胞的增殖、黏附、趋化和侵袭等生物学过程.  相似文献   

3.
富亮氨酸重复超家族新成员LRRC4基因是新克隆的脑瘤相关基因,采用多聚酶链式反应(PCR)方法获得长约500bp含IgC2结构域的DNA序列,扩增产物克隆至pGEX-4T-2质粒中,构建GST融合表达质粒,在大肠杆菌中诱导表达融合蛋白,经包涵体沉淀,溶解,Glutathione-Sepharose亲和层析纯化获得融合蛋白,并以Western blot鉴定证实,通过IgC2结构域蛋白的纯化分离该结构域,为进一步研究该结构域及LRRC4基因的结构和功能奠定了基础。  相似文献   

4.
在染色体7q31-32多种肿瘤杂合性丢失(loss of heterozygosity,LOH)高频区,采用表达序列标签(expressed sequence tag,EST)介导的定位候选克隆策略获得了一个定位于人染色体7q31-32的新基因(GenBank 登录号: AF196976).该基因编码653个氨基酸,蛋白质理论pI/m:6.58/72.7 ku.它包含七个典型的LRR、一个IgC2样结构域.此外,它还包含一个N端信号肽、一个C端跨膜区.其结构特征表明它是富亮氨酸重复(leucine-rich repeat,LRR)超家族的新成员.经过人类基因组命名委员会的同意,将该基因命名为LRRC4.此外,通过序列相似性匹配还获得了定位于小鼠6号染色体的LRRC4的同源基因(GenBank 登录号: AF290542).RNA印迹和RT-PCR检测发现LRRC4在正常人脑组织相对特异表达,而在多种原发性脑瘤表达明显下调或缺失.综合考虑LRRC4基因的序列特征及表达谱,提示LRRC4基因可能在神经系统中发挥重要作用.  相似文献   

5.
目的:构建TANK结合激酶1(TBK1)相关激酶活性缺失突变体和泛素样结构域突变体真核表达载体,检测该基因相关突变体在293细胞中的表达,并利用萤光素酶报告基因实验检测其生物活性。方法:根据文献报道的突变序列及QuickChange Site-Directed Mutagenesis实验设计手册,设计合成2条针对TBK1相关激酶活性缺失突变体和泛素样结构域突变体的引物,以实验室之前构建的TBK1野生型真核表达载体为模板,构建TBK1激酶活性缺失突变体和泛素样结构域突变体真核表达载体,分别命名为pcDNA3-Flag-TBK1(KD)、pcDNA3-Flag-TBK1(ΔULD)。以LipofactAMINE2000转染试剂转染至293细胞中进行瞬时表达,利用萤光素酶实验检测2种TBK1突变体诱导β干扰素(IFN-β)转录的情况。结果:测序结果表明,TBK1相关激酶活性缺失突变体和泛素样结构域缺失突变体真核表达载体构建成功,Western印迹检测表明其在293细胞中获得有效表达;用萤光素酶报告基因实验检测,与野生型TBK1相比,其相关激酶活性缺失突变体和泛素样结构域缺失突变体诱导IFN-β转录激活的作用明显降低。结论:真核表达的TBK1相关激酶活性缺失突变体和泛素样结构域突变体具有相应的生物学活性,为研究其功能奠定了基础。  相似文献   

6.
该研究将构建的含淀粉样前体蛋白(amyloid precursor protein,APP)基因启动子的萤火虫荧光素酶报告质粒与Purα全长基因或含不同结构域的Purα缺失突变体共转染到U87MG细胞中,进行萤火虫荧光素酶活性测定,以确定Purα对APP基因表达的调控作用。同时,将Purα全长基因及含有不同结构域的Purα缺失突变体的真核表达载体分别转染至U87MG细胞,使目的蛋白过表达,通过实时定量PCR(Real-time PCR)和蛋白免疫印迹(Western blot)研究Purα不同的结构域对APP基因在转录、翻译水平的调控作用。结果显示,Purα全长蛋白及其不同结构域对APP基因表达均有不同程度的抑制作用,但至少保持N-端或C-端的结构域可能是维持Purα蛋白功能的必要条件。  相似文献   

7.
人类功能基因RNF122能够明显抑制细胞生长,导致细胞凋亡.RNF122含有RING-H2结构域.为了研究RNF122的RING结构域和凋亡的相互关系,构建了RING结构域突变体.MTT和凋亡实验发现,RNF122与细胞存活的密切关系依赖于其RING结构域.进一步的实验提示,RNF122能够负向调节ERK通路,而RING结构域突变型RNF122则能够增强ERK的磷酸化,提示RNF122可能通过ERK通路调节细胞的存活.总之,RING结构域对于RNF122发挥功能起至关重要的作用.  相似文献   

8.
目的:构建登革2型病毒非结构蛋白NS4B及其突变体Δ2K-NS4B基因的真核载体,并观察二者在哺乳动物细胞内的定位情况。方法:从登革2型病毒43株的全长cDNA克隆载体上扩增获得编码NS4B及缺失2K片段的NS4B突变体Δ2K-NS4B的基因;通过基因重组的方法分别将2段基因克隆入真核表达载体pcDNA6/V5-HisA,获得重组真核表达载体pc/D2-NS4B和pc/D2-Δ2K-NS4B;经脂质体法转染BHK-21细胞后,用RT-PCR、间接免疫荧光和Western印迹鉴定表达的蛋白。结果:重组蛋白D2-NS4B和D2-Δ2K-NS4B可在BHK-21细胞中表达,二者均定位于细胞质中,并具有较好的抗原性,能够被抗登革2型病毒NS4B的多克隆抗体特异识别。结论:重组蛋白D2-NS4B和D2-Δ2K-NS4B在哺乳动物细胞胞质中的正确表达,为深入了解NS4B在登革病毒致病过程中的生物学功能奠定了基础。  相似文献   

9.
5-Aza-CdR对胶质瘤细胞生长及LRRC4基因异常甲基化的影响   总被引:1,自引:0,他引:1  
LRRC4是一个新发现的胶质瘤抑瘤基因,它在多种胶质瘤细胞系和胶质瘤组织表达缺失或下调,前期研究结果表明胶质瘤细胞和组织中LRRC4的编码区未发生突变、缺失或重排.为了获得LRRC4作为胶质瘤抑瘤基因的进一步证据,采用去甲基化制剂5-Aza-CdR处理LRRC4表达缺失的SF126和SF767胶质瘤细胞,MSP和RT-PCR检测表明,LRRC4的启动子在表达缺失的SF126和SF767细胞存在完全的甲基化,而5-Aza-CdR能逆转LRRC4启动子的甲基化状态,恢复LRRC4的表达.MTf法测定显示,5-Aza-CdR使SF126和SF767胶质瘤细胞增殖受到明显抑制,并呈时间和剂量的依赖性.同时流式细胞仪检测显示,5-Aza-CdR使SF126和SF767胶质瘤细胞周期阻滞于G0/G1期.因此,5-Aza-CdR能抑制胶质瘤细胞SF126和SF767增殖并干扰其细胞周期,LRRC4启动子异常甲基化足其在胶质瘤细胞中表达缺失的重要机制,5-Aza-CdR能逆转LRRC4基因的甲基化,恢复LRRC4的表达,为LRRC4作为胶质瘤去甲基化治疗的靶标提供了科学依据.  相似文献   

10.
LRRC4融合蛋白的构建与表达研究   总被引:2,自引:1,他引:1  
在前期工作中,采用EST介导的定位候选克隆策略,克隆了一个在脑瘤中表达下调的脑特异表达新基因LRRC4,为进一步研究其结构与功能的关系,构建了含LRRC4基因全长编码区的pGEM-T Easy质粒,在此基础上通过亚克隆构建了LRRC4融合蛋白的绿色荧光蛋白(pEGFP-C1)表达质粒,瞬时转染哺乳动物细胞,结果发现表达的LRRC4融合蛋白定位于活细胞的细胞膜上.同时,构建了LRRC4全长和截短型原核表达pGEX-4T-2质粒,成功而高效地在大肠杆菌BL21 中表达LRRC4融合蛋白.上述工作为制备多抗,深入研究LRRC4基因的功能奠定了基础.  相似文献   

11.
12.
Our previous study has shown that LRRC4 is a novel member of the leucine-rich repeat (LRR) superfamily and has the potential to suppress brain tumor growth. In order to further analyze the functions of LRRC4 on the maintenance of normal function and suppression of tumorigenesis in the central nervous system, we investigated alterations in gene expression related to neurobiology by the Atlas array in two inducible dual-stable LRRC4-overexpressing cell lines. Seventeen of 588 genes spotted on the Atlas membrane showed altered expression levels in LRRC4 transfected U251MG Tet-on cells, which are involved in cell proliferation and cell cycle progression, tumor invasion and metastasis, and neurotransmitter synthesis and release. In addition, cell invasion assay results showed that LRRC4 can inhibit the U251MG cell migration. These studies represent the first cDNA array analysis of the effects of LRRC4 on the involvement of different neurobiological genes in U251MG glioblastoma cells and provide new insights into the function of LRRC4 in glioma.  相似文献   

13.
LRRC4是一个在脑相对特异性表达的富亮氨酸重复超家族新成员,在神经胶质瘤表达明显下调或缺失且具有抑制脑胶质瘤细胞生长的潜能. 利用 Tet-on 基因表达系统,经过两轮转染,先后将调控质粒 pTet-on 和表达质粒 pTRE-2hyg-LRRC4 转染 U251 细胞系,分别用 G418 和潮霉素 Hygromycin 进行两次筛选. 在第一轮挑取的 80 个克隆中,利用 pTRE-2hyg-luciferase 报告基因进行最佳的低背景高表达的 pTet-on 细胞克隆筛选,在通过量效关系和动力学检测筛选的最佳克隆基础上,再进行 pTRE-2hyg-LRRC4 的转染,并通过 RT-PCR 和 RNA 印迹检测,成功获得了两个具有良好诱导性 Tet 调控的 LRRC4 双稳定表达细胞系,为进一步阐明 LRRC4 在脑胶质瘤发生发展中的作用,提供有利的研究基础和理想的实验平台.  相似文献   

14.
Zhang Q  Wang J  Fan S  Wang L  Cao L  Tang K  Peng C  Li Z  Li W  Gan K  Liu Z  Li X  Shen S  Li G 《FEBS letters》2005,579(17):3674-3682
LRRC4, a novel member of LRR superfamily thought to be involved in development and tumorigenesis of the nervous tissue, has the potential to suppress tumorigenesis and cell proliferation of U251MG cells. This study aimed at revealing the correlation between expression of LRRC4 and the maintenance of normal function and tumorigenesis suppression within the central nervous system. We systematically analyzed the expression and tissue distributions of the gene in tissues. Results showed that LRRC4 expression was limited to normal adult brain, both in human and in mouse, and exhibited a development-regulated pattern, but was down-regulated in brain tumor tissues and U251MG cell line. Furthermore, dynamic alterations in gene expression associated with cell cycle progression were investigated by using Tet-on system. Results showed that LRRC4 induced a cell cycle delay at the late G1 phase, probably through the alteration of the expression of different cell cycle regulating proteins responsible for mediating G1-S progression, such as p21(Waf1/Cip1) and p27(Kip1), Cdk2 and PCNA, p-ERK1/2. These findings suggest that LRRC4 may play an important role in maintaining normal function and suppressing tumorigenesis in the central nervous system.  相似文献   

15.
Gliomas take a number of different genetic routes in the progression to glioblastoma multiforme, a highly invasive variant that is mostly unresponsive to current therapies. The alpha-chemokine stromal cell-derived factor (SDF)-1 alpha binds to the seven transmembrane G-protein-coupled CXCR-4 receptor and acts to modulate cell migration and proliferation by activating multiple signal transduction pathways. Leucine-rich repeats containing 4 (LRRC4), a putative glioma suppressive gene, inhibits glioblastoma cells tumorigenesis in vivo and cell proliferation and invasion in vitro. We also previously demonstrated that LRRC4 controlled glioblastoma cells proliferation by ERK/AKT/NF-kappa B signaling pathway. In the present study, we demonstrate that CXC chemokine receptor 4 (CXCR4) is expressed in human glioblastoma U251 cell line, and that SDF-1 alpha increases the proliferation, chemotaxis, and invasion in CXCR4+ glioblastoma U251 cells through the activation of ERK1/2 and Akt. The reintroduction of LRRC4 in U251 cells inhibits the expression of CXCR4 and SDF-1 alpha/CXCR4 axis-mediated downstream intracellular pathways such as ERK1/2 and Akt leading to proliferate, chemotactic and invasive effects. Furthermore, we provide evidence for proMMP-2 activation involvement in the SDF-1 alpha/CXCR4 axis-mediated signaling pathway. LRRC4 significantly inhibits proMMP-2 activation by SDF-1 alpha/CXCR4 axis-mediated ERK1/2 and Akt signaling pathway. Collectively, these results suggest a possible important "cross-talk" between LRRC4 and SDF-1 alpha/CXCR4 axis-mediated intracellular pathways that can link signals of cell proliferation, chemotaxis and invasion in glioblastoma, and may represent a new target for development of new therapeutic strategies in glioma.  相似文献   

16.
LRRC4 is a tumor suppressor of glioma, and it is epigenetically inactivated commonly in glioma. Our previous study has shown that induction of LRRC4 expression inhibits the proliferation of glioma cells. However, little is known about the mechanisms underlying the action of LRRC4 in glioma cells. We employed two-dimensional fluorescence differential gel electrophoresis (2-D DIGE) and MALDI -TOF/TOF-MS/MS to identify 11 differentially expressed proteins, including the significantly down-regulated STMN1 expression in the LRRC4-expressing U251 glioma cells. The levels of STMN1 expression appeared to be positively associated with the pathogenic degrees of human glioma. Furthermore, induction of LRRC4 over-expression inhibited the STMN1 expression and U251 cell proliferation in vitro, and the glioma growth in vivo. In addition, induction of LRRC4 or knockdown of STMN1 expression induced cell cycle arrest in U251 cells, which was associated with modulating the p21, cyclin D1, and cyclin B expression, and the ERK phosphorylation, and inhibiting the CDK5 and cdc2 kinase activities, but increasing the microtubulin polymerization in U251 cells. LRRC4, at least partially by down-regulating the STMN1expression, acts as a major glioma suppressor, induces cell cycle arrest and modulates the dynamic process of microtubulin, leading to the inhibition of glioma cell proliferation and growth. Potentially, modulation of LRRC4 or STMN1 expression may be useful for design of new therapies for the intervention of glioma.  相似文献   

17.
LRRC4 is a novel relatively specific gene,which displays significant down-regulation inprimary brain tumor biopsies and has the potential to suppress brain tumor growth.In this study,we inves-tigated the growth inhibitory effect of LRRC4 on tumorigencity in vivo and on cell proliferation in vitro by atetracycline-inducible expression system.Results showed that LRRC4 significantly reduced the growth andmalignant grade of xenografts arising from glioblastoma U251MG cells.Cell proliferation was markedlyinhibited after U251MG Tet-on-LRRC4 cell induction with doxycycline.Flow cytometry and Western blotanalysis demonstrated that LRRC4 mediated a delay of the cell cycle in late G_1,possibly through up-regulat-ing the expressions of p21Wafl/cip 1 and p27Kip 1 and down-regulating the expressions of cyclin-dependentkinase 2,retinoblastoma protein and epidermal growth factor receptors.Together,these findings provideclues to the function of LRRC4 as a negative regulator of cell growth and underscore a link between theabove-mentioned cyclins,cyclin-associated molecules and tumorigenicity.  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