用户名: 密码: 验证码:
BTBD10在人脑胶质瘤中的表达及其对U251细胞株增殖和凋亡的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
脑胶质瘤是成人中枢神经系统最常见的恶性肿瘤,统计资料表明其发生率约占颅内肿瘤的40-50%,其多呈浸润性生长,与正常脑组织间常无明显分界,易侵犯重要神经结构,难以做到手术完全切除,预后差,复发率、病死率高。而药物化疗和放射治疗不能高度特异性地杀伤肿瘤细胞,并且可能产生严重的不良反应。因此,目前脑胶质瘤仍为神经外科领域的难治性疾病,而阐明其发病机制、寻找新的治疗手段成为神经外科的研究热点。
     和其他部位肿瘤一样,脑胶质瘤的发生、发展是一个非常复杂的过程,涉及多种肿瘤相关基因的表达异常或失活。胶质瘤本质上是一种多基因异常疾病,其发生是由于原癌基因的激活和抑癌基因的失活导致细胞信号传导通路的异常、细胞循环周期的改变、生命周期的延长以及细胞调亡缺陷等,从而出现细胞增殖失控及恶性转化。与脑胶质瘤的恶性进展密切相关的细胞通路主要有磷脂酰肌醇3-激醇(P13K)信号转导通路、Ras信号转导通路、Wnt信号转导通路和Notch信号转导通路等。寻找与胶质瘤发病相关的基因,在此基础上发现胶质瘤治疗的新靶点、新策略,是神经外科研究领域的主要内容之一。
     BTBD10基因含BTB/POZ功能域,位于人染色体的11p15.2区域,全长2551bp,含245-16721bp开放阅读框架,编码475氨基酸的蛋白,蛋白分子量51.7Kd。前期研究在对18例脑胶质瘤进行表达谱芯片筛选及聚类分析中发现:芯片数据提示BTBD10表达量在各级别胶质瘤中均发生下调,平均Ratio值为0.218。基因表达谱Hierarchical聚类发现与BTBD10表达谱相近的基因有:血管性肠肽(VIP)、肿瘤蛋白p53结合蛋白(TP53BP1)、p淀粉样蛋白前体(APBB1)、8氧化鸟嘌呤糖基酶(OGG1)、促性腺素诱导阻抑蛋白(GIOT1)、MADS转录增强子2C(MEF2C)。MTC为模板RT-PCR结果显示BTBD10在检测的8种正常组织中,脑组织中高表达,而心、肺、肝、肾、胰腺组织中表达量较低。Northern杂交结果显示BTBD10在正常成人脑组织中表达量高,在7例不同级别胶质瘤中表达量都下降,BTBD10在胶质瘤/正常脑组织的表达量比率与芯片数据基本相符。在肝癌、卵巢癌和肺癌中BTBD10的表达没有显著性差异。提示BTBD10基因与脑胶质瘤高度相关,有可能成为新的脑胶质瘤治疗靶基因。为进一步研究BTBD10基因在胶质瘤发生发展中的作用,本实验通过检测BTBD10在大样本胶质瘤中的表达情况,培养过表达BTBD10基因的细胞株,初步探讨BTBD10基因调节胶质瘤细胞增殖水平的可能机制。
     第一部分BTBD10在人脑胶质瘤组织中的表达及意义
     目的:探讨BTBD1OmRNA和BTBD10蛋白在脑胶质瘤中的表达及意义。
     方法:采用实时荧光定量PCR和Western blot杂交检测BTBD10mRNA和BTBD10蛋白在正常脑组织和各级别胶质瘤中的表达,分析其表达与胶质瘤临床病理特征的关系。
     结果:在正常脑组织中BTBD10mRNA和蛋白的表达较高;各级别胶质瘤中的表达均低于正常脑组织(P<0.05);在低级别胶质瘤(Ⅰ-Ⅱ级)中表达量较低,而在高级别胶质瘤(Ⅲ-Ⅳ)中表达水平更低,低级别和高级别之间的表达差异具有统计学意义(P<0.05)。BTBD1OmRNA和蛋白的表达水平与年龄、性别、病理类型等临床病理特征无明显相关(P>0.05)。
     结论:胶质瘤中BTBD10表达明显低于正常脑组织中的表达,且随病理级别的增高而降低,提示BTBD10在胶质瘤的发生发展中起重要作用,并与胶质瘤的病理分级密切相关。
     第二部分BTBD10过表达慢病毒载体的构建和鉴定
     目的:构建BTBD10慢病毒表达载体,为后续实验研究提供依据。
     方法:以BTBD10基因为模板PCR扩增目的基因,表达载体(pLV-UbC-GFP-3FLAG)酶切后进行胶回收载体片段,目的基因与载体片段同源重组后转化感受态细胞,用菌落PCR鉴定转化子,阳性克隆测序鉴定,质粒抽提,与无内毒素病毒包装质粒、膜蛋白表达质粒共转染293T细胞包装产生慢病毒颗粒,测定慢病毒滴度。
     结果:PCR扩增和测序结果均证实,插入序列完全正确,包装病毒产生病毒悬液的滴度为3.42×108TU/ml。
     结论:成功构建了携带BTBD10基因的慢病毒载体,为研究其在胶质瘤细胞中的生物学功能和以其为靶点的基因治疗奠定基础。
     第三部分BTBD10慢病毒载体转染人胶质瘤细胞株U251及其作用机制研究
     目的:了解BTBD10重组慢病毒载体转染人胶质瘤细胞株U251后,对U251细胞增殖、凋亡的影响,探讨其发挥肿瘤调控作用的机制。
     方法:应用BTBD10慢病毒载体、空载体分别转染体外培养的U251人胶质瘤细胞株,正常培养U251细胞作为空白对照。荧光定量PCR检测转染前后BTBD10和CyclinDl mRNA的表达变化,Western blot杂交检测BTBD 10、磷酸化Akt(pAkt)和CyclinD1蛋白水平变化,流式细胞仪检测细胞周期和凋亡水平,MTT法分析细胞增殖情况并绘制生长曲线。
     结果:荧光定量PCR和Western blot杂交结果显示,与未转染的U251细胞和转染空白载体的U251细胞相比,转染重组BTBD10病毒载体的U251细胞中BTBD10 mRNA和蛋白水平明显提高,差异具有统计学意义(P<0.05);Akt的磷酸化水平下调,CyclinDl mRNA和蛋白水平下调,差异具有统计学意义(P<0.05)。流式细胞仪分析各组细胞的凋亡水平,检测结果表明,与未转染的U251细胞和转染空白载体的U251细胞相比,转染BTBD10的U251细胞中凋亡细胞明显增多,早期凋亡细胞达14.64%,统计学上有差异(P<0.05):细胞周期检测结果显示,未转染的U251细胞和转染空载体的U251细胞的细胞周期和细胞状态基本相同,部分细胞处于G2/M期,转染BTBD10病毒载体的U251细胞则被明显的抑制在G0/G1期,提示增殖活性降低。MTT法检测细胞增值率,发现外源性BTBD10基因转染U251细胞后,细胞生长速度明显下降。
     结论:慢病毒表达载体对人胶质瘤细胞系U251具有很高的转染效率,感染后可显著上调靶基因BTBD10的表达和活化。BTBD10过表达后将肿瘤细胞阻滞在G0/G1期,抑制肿瘤细胞增殖,促进凋亡,可能与下调pAkt和CyclinD1水平,负调控PI3K/Akt信号通路有关。
Gliomas are the most common primary tumor of the central nervous system in adults, and accounts for approximately 40-50% of total intracranial tumors. These tumors are highly invasive, and have no obvious dividing line with normal brain tissue, often infiltrate critical neurological areas within the brain. It is difficult to achieve totally surgical resec-tion, and the recurrence and fatality rate is high. In addition, chemotherapy and radioatio-therapy can not be a high degree of specificity in killing tumor cells, and may cause serious adverse reactions. Therefore, glioma is still a refractory disease in the neurosurgical field. To elucidate its pathogenesis and to find new treatments is a research hotspot in neurosurgery.
     It has been showed that the devolpment of glioma, just the same as the tumors in other sites of the body, is a very complex process, involving expression disorder or inactivation of a variety of tumor-related genes. Glioma is essentially a multi-gene disorders. Its occurrence is due to the activation of proto-oncogenes and inactivation of tumor suppressor genes, which lead to excessive cell proliferation and malignant transformation. The malignant progression of glioma cells is closely related with several pathways, such as PI3K (phosphatidylinositol 3-kinase) signaling pathway, Ras signaling pathway, Wnt signaling pathway and Notch signaling pathway. However, up to now, the molecular mechanism of glioma is not fully elucidated. Seeking glioma pathogenesis-related genes, finding new target and new strategy for glioma therapy, plays a important role in the field of neurosurgery.
     BTBD10 contained a BTB/POZ domain, located in human chromosome 11p15.2 region. It was 2489 bp in length, containing open reading frame from 245 to 1672 bp, encoding 475 amino acid protein with a molecular weight of 51.7 Kd. Preliminary study of 18 glioma samples of cDNA microarray screening and cluster analysis found that the expression level of BTBD10 descend in all samples, with an average Ratio value of 0.218. The result of RT-PCR showed that a ubiquitous expression pattern of BTBD10 in the 8 tissues examined with high expression in adult brain, testis and small intestine and weak expression in the heart, lung, liver, kidney, pancreas, spleen, thymus. Northern blot showed that BTBD10 was highly expressed in mixed normal brain while it was sharply down-regulated in all grades of glioma tissues. RT-PCR showed that there was no significant difference in hepatocellular carcinoma, ovary cancer and lung cancer tissues. That means the BTBD10 gene is highly correlated with glioma, and it may become a new target gene in gene therapy of gliomas. For further studying the role of BTBD10 gene in glioma, we began with examining the expression of BTBD10 in a large number of glioma specimens in the present study, then increased the expression of BTBD10 in glioma cell lines, evaluated the effects of BTBD10 on glioma proliferation and apoptosis.
     Part I:Study on the expression of BTBD10 in human gliomas and its significance
     Objective:To investigate the expression of BTBD10 mRNA and protein in brain gliomas and its significance.
     Methods:The mRNA expression of BTBD10 in 52 cases of human glioma tissues and 8 cases of normal brain tissues was detected with quantitative real-time PCR. The protein expression in these samples were detected using western blot.
     Results:The expression of BTBD10 mRNA and protein is higher in normal brain tissue than in all grades of glioma tissue (P<0.05) and significantly decreased ascending the degree of glioma. The difference between the low-grade and high-grade tumors is statisticly significant(P<0.05). A statistically significant association was found between expression of BTBD10 mRNA and protein.
     Conclusion:BTBD10 mRNA and protein showed markedly lower expression in glioma than normal brain tissues, and the expression decreased with the malignancy of glioma. These results suggested that BTBD10 was significantly associated with the proliferation activity and may play an improtant role in human glioma.
     Part II:Construction and identification of lentiviral vector plasmid containing BTBD10
     Objective:To construct lentiviral vector plasmid containing BTBD10 and provide the basis for further experiments.
     Methods:The vector was obtained from pLV-UbC-GFP plasmid digested with EcoR I and Nhe I restriction enzymes. Then BTBD10 gene was inserted into the vector to construct pLV-BTBD10, which was subsequently confirmed by PCR and DNA sequencing analysis. After these identification, the resultant pLV-BTBD10 plasmid, pCD/NL-BH*DDD and pLTR-G were transfected into 293T cell to produce a replication incompetent lentivirus. Finally the titer of lentivirus was determined.
     Results:PCR analysis and DNA sequencing confirmed that the BTBD10 sequences were successfully inserted into the lentiviral vectors. The titer of concentrated virus was 3.42 x 108TU/ml.
     Conclusion:The lentiviral vector plasmid containing BTBD10 has been successfully constructed, which will provide a foundation for the further study on function of BTBD10 gene in glioma and gene therapy.
     Part III Growth inhibition of human glioma cell line U251 by transfer of lentiviral vector containing BTBD10 and its mechanism
     Objective:To investigate the effects of recombinant lentiviral vector containing BTBD10 on tumor proliferation and apoptosis of human glioma cells U251, and to explore the mechanism of regulating tumors.
     Methods:BTBD10 lentiviral vector and empty vector were transfected into cultured U251 human glioma cell lines separately. Parental U251 cells was cultured as a negative control. Real-time quantitative PCR was chosen for detection the mRNA expression of BTBD10 and CyclinDl. The protein expression of BTBD10, phosphorylated Akt(pAkt), and CyclinDl were detected using western blot. Cell apoptosis and cell cycle were analyzed by flow cytometry. MTT assay was used to detected cell viability.
     Results:The real-time quantitative PCR and western blot hybridization results showed that compared with non-transfected U251 cells and U251 cells(transfected with empty vector), the expression levels of BTBD10 mRNA and protein were significantly increased in U251 cells(transfected with BTBD10 lentiviral vector), and the difference was statistically significance (P<0.05). The expression levels of pAkt protein was decreased, and the difference was statistically significant (P<0.05). The expression levels of CyclinDl mRNA and protein were down regulated, and the difference was statistically significant (P<0.05). The quantitative analysis of apoptotic cells by FCM showed that compared with non-transfected U251 cells and U251 cells(transfected with empty vector), the apoptotic cells increased significantly in U251 cells(transfected with BTBD10 lentiviral vector) with the apoptotic rate of 14.64%. The difference was statistically significant (P<0.05). The results on cell cycle showed that in non-transfected U251 cells and U251 cells (transfected with empty vector), cell cycle and cell statement were basically the same, some cells were in the G2/M phase. U251 cells(transfected with BTBD10 lentiviral vector), were markedly inhibited in the G0/G1 phase. MTT assay showed that the group transfected with BTBD10 lentiviral vector had a much lower cell viability than other group.
     Conclusion:The lentiviral vector plasmid containing BTBD10 showed high efficiency on infecting glioma U251 cells line and significantly increased the expression and activation of BTBD10 in cells. The over-expression of BTBD10 expression was able to inhibit the glioma cell proliferation acivity and to induce cell apoptosis. The mechanism may be related to negative regulation of PI3K/Akt signaling pathways by reducing the expresssion of pAkt and CyclinDl.
引文
[1]Chen J, Xu J, Ying K, Cao G, Hu G, Wang L,Luo C,Lou M, Lu Y, et al. Molecular cloning and characterization of a novel human BTB domain-containing gene,BTBD10,which is down-regulated in glioma.Gene,2004,340:61-69.
    [2]YakataY, Nakayama T,Yoshizaki A, et al. Expression of p-STAT3 in human gastric carcinoma:si gnificant correlation in tumour invasion and prognosis. Int J Oncol.2007,30(2):437-442.
    [3]Korshunov A, Sycheva R, Golanov A. The prognostic relevance of molecular alterations in glioblastomas for patients age<50 years. Cancer.2005,104:825-32.
    [4]Ohgaki H. Epidemiology of brain tumors. Methods Mol Biol.2009,47(2):323-42.
    [5]Norden AD, Wen PY. Glioma therapy in adults. Neurologist,2006,12(8):279-292.
    [6]Reardon DA, Rich JN, Friedman HS, et al. Recent advances in the treatment of malignant astrocytoma. J Clin Oncol.2006,24(8):1253-1265.
    [7]Brandes AA,Franceschi E,Tosoni A,et al.Epidermal growth factor receptor inhibitors in neuro-oneology:hopes and disappointments. Clin Cancer Res,2008,14(4):957-960.
    [8]Carnero A,Blanco-Aparicio C,Rennet O,et al.The PTEN/PI3K/Akt signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets,2008,8(5):187-198.
    [9]Kowalczuk A, Macdonald RL,Amidei C. Quantitative imaging study of extent of surgical resection and prognosis of malignant astrocytomas. Neurosurgery,1997,41:1028.
    [10]Deimling A, Louis DN, Wiestler OD. Molecular pathways in the formation of gliomas. Glia. 1995,15:328-38.
    [11]Bredel M, Bredel C, Jufic D, Harsh GR, Vogel H, Recht LD. Functional network analysis reveals extended gliomagenesis pathway maps and three novel MYC-interacting genes in human gliomas. Cancer Res.2005,65(18):8679-8689.
    [12]Bredel M, Bredel C, Jufic D, et al. High-resolution genome-wide mapping of genetic alterations in human glial brain tumors. Cancer Res.2005,65(17):4088-4096.
    [13]Mischel PS, Cloughesy TF, Nelson SF. DNA-microarray analysis of brain cancer:molecular classification for therapy. Nat Rev Neurosci.2004,5:782-92.
    [14]Shirota Y, Kaneko S, Honda M, et al. Identification of differentially expressed genes in hepatocellular carcinoma with cDNA microarrays. Hepatology,2001,33(4):832-840.
    [15]Nutt CL, Mani DR, Betensky RA, Tamayo P, et al. Gene expression-based classification of malignant gliomas correlates better with survival than histological classification. Cancer Res. 2003,63(10):1602-1607.
    [16]陈菊祥,卢亦成,骆纯,等.人BTBD10新基因克隆及组织表达和在脑胶质瘤中的表达谱及聚类.中华神经外科杂志,2004,20(3):187-190.
    [17]Mizutani T, Yamada K, Yazawa T, et al. Cloning and characterization of gonadotropin-inducible ovarian transcription factors (GIOT1 and-2) that are novel members of the (Cys(2)-(His)(2)-type zinc finger protein family. Mol Endocrinol,2001,15:1693-1705..
    [18]Sharma A, Walters J, Gozes Y, et al. A vasoactive intestinal peptide antagonist inhibits the growth of glioblastoma cells. J Mot Neurosci,2001,17:331-339.
    [19]Weger S, Hammer E, Heilbronn R. Topors, a p53 and topoisomerase I binding protein. interacts with the adenoassociated virus (AAV-2) Rep78/68 proteins and enhances AAV-2 gene expression. J Gen Virol,2002,83.511-516.
    [20]Askanas V, Engel WK. Proposed pathogenetic cascade of inclusion-body myositis:importance of amyloid-beta, misfolded proteins. Predisposing genes, and aging. Curr Opin Rheumatol,2003,15, 737-744.
    [21]Paz-Elizur T, Ktupsky M, Blumenstein S, et al. DNA repair activity for oxidative damage and risk of lung cancer. J Natl Cancer Inst,2003,95:1312-1319.
    [22]Hosking BM, Wang SC, Chen SL, et al. SOX18 directly interacts with MEF2C in endothelial cells. Biochem Biophys Res Commun,2001,287:493-500.
    [23]Zollman S, Godt D, Prive G, et al. The BTB domain, primarily found in zinc finger proteins, defines an evolutionarily conserved family that includes several developmentally regulated genes of Drosophila Proc Natl Acad Sci USA,1994,91:10717-10721.
    [24]Bardwell, V, Treisman, R. The POZ domain:A conserved protein-protein interaction motif. Genes Dev.1994,8:1664-1667.
    [25]Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, et al. Initial sequencing and analysis of the human genome. Nature,2001,409(6822):860-921.
    [26]Melnick A, Ahmad KF, Arai S, et al. In-depth mutational analysis of the promyelocytic leukemia zinc finger BTB/POZ domain reveals motifs and residues required for biological and transcriptional functions. Mot Cell Biol.2000,20(5):6550-6567.
    [27]刘瑜,王宣春,陈凤玲,胡仁明.BTBD10基因的细胞、组织mRNA表达谱研究.中国分子心脏病学杂志.20008,8(6):347-349.
    [28]刘瑜,王宣春,陈凤玲,胡仁明.BTBD10基因的真核表达载体构建和亚细胞定位.中国分子心脏病学杂志.2009,9(1):16-18.
    [29]Mikiro N, Kohsuke K, Yuichi H, Sadakazu A, Masaaki M. A novel Akt/PKB-interacting protein promotes cell adhesion and inhibits familial amyotrophic lateral sclerosis-linked mutant SOD1-induced neuronal death via inhibition of PP2A-mediated dephosphorylation of Akt/PKB. Cell Signal.2008,20(3):493-505.
    [30]Weyer C, Bogardus C, Mott DM, et al. The natural history of insulin secretory dysfunction and insulin resistance in the pathogenesis of type 2 diabetes mellitus. J Clin Invest.1999,104:787-794.
    [31]Higuchi R, Fockler C, et al. Kinetic PCR analysis:real-time monitoring of DNA amplification reactions. Biotechnology,1999,11 (9):1026-1030.
    [32]Kel D, Chen Z, Yung, et al. A reliability test of standard-based quantitative PCR:exogenous vs endogenous standards. Mol Cell Probes,2006,14(2):127-135.
    [33]Wang X, X Li, Kual W, et al. Application of real-time polymerase chain reaction to quantitate induced expression of interleukin-1 beta mRNA in ischemic brain tolerance. Neurosci Res,2000, 59(2):238-246.
    [34]Rubinson DA, Dinon CP. A leniivirus-based system to functlonally silence genes in pirmary mammalian cells, stem cells and transgeinc mice by RNA interferenc. Nat Genet,2003,33: 401-406.
    [35]Naldini L, Blomer U. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science,1996,272:263-267.
    [36]Jacobs AH, Voges J, Kracht LW, et al. Imaging in gene therapy of patients with glioma. J Neurooncol,2003,65(3):291-305.
    [37]Alici E, Sutlu T, Sirac DilberM. Retroviral Gene Transfer into Primary Human Natural Killer Cells. Methods Mol Biol,2009,506:1271.
    [38]Sio FS, Naldini L. Short-term culture of human CD34+ cells for lentiviral gene transfer. Methods Mol Biol,2009,506:59.
    [39]Pan D. In situ (in vivo) gene transfer into murine bone marrow stem cells. Methods Mol Biol, 2009,506:159.
    [40]Yu SS, Dan K, Chono H, et al. Transient gene expression mediated by integrase-defective retroviral vectors. Biochem Biophys Res Commun,2008,368 (4):942-947.
    [41]Wang W, Tai CK, Kershaw AD, et al. Use of replication-competent retroviral vectors in an immunocompetent intracranial glioma model. Neurosurg Focus,2006,20(4):E25.
    [42]Cao H, Koehler DR, Hu J. Adenoviral vectors for gene replacement therapy. Viral Immunol,2004, 17(3):327-333..
    [43]Schagen FH, Ossevoort M, Toes RE, et al. Immune responses against adenoviral vectors and their transgene products:a review of strategies for evasion. Crit Rev Oncol Hematol,2004,50(1): 51-57.
    [44]黄黎珍,刘光泽,顾为盼.慢病毒载体法制备转基因动物研究进展.实验动物与比较医学,2007,27(1):64-66.
    [45]Delenda C. Lentiviral vectors:optimization of packaging transduction and gene expression. Gene Med,2004,6 (1):125-138.
    [46]Sumimoto H, Kawakami Y. Lentiviral vector-mediated RNAi and its use for cancer research. Future Oncol,2007,3(6):655-664.
    [47]Liehl B, Hlavaty J, Moldzio R, et al. Simian immunodeficiency virus vector pseudotypes differ in transduction efficiency and target cell specificity in brain. Gene Ther,2007,14(18):1330-1343.
    [48]Banerjen A,Li MJ, Bauer G, et al. Inhibition of HIV-1 by lentiviral vector-transduced siRNAs in T lymphocytes differentiated in SCID-hu mice and CD34+ progenitor cell-derived macrophages. Mol Ther,2003,8(1):62-69.
    [49]张经纬,冯建翔,徐荣明等.慢病毒介导碱性成纤维细胞生长因子基因转染促进半月板纤维软骨细胞的增殖与基质合成.中国组织工程研究与临床康复,2007,11(41):8267-8270.
    [50]Gerolami R, Rathviro U, Faivre J, et al. Herpes simplex virus thymidie kinase-mediated suicide gene therapy for hepatocellular using HIV-1-derived lentiviral vectors. J Hepatol,2004,40(2): 291-297.
    [51]Luo F, Yee JK, Huang SH, et al. Downregulation of human Cdc6 protein using a lentivirus RNA interference expression vector. Methods Mol Biol,2006,342:287-293.
    [52]Santhosh CV, Tamhane MC, Mukhopadhyaya R, et al. A lentiviral vector with novel multiple cloning sites:stable transgene expression in vitro and in vivo. Biochem Biophys Res Commun, 2008,4(3):546-550.
    [53]Cockrell AS, Kafri T. Gene delivery by lentivirus vectors. Mol Biotechnol,2007,36(3):184-204.
    [54]Nishitsuji H, Ikeda T, Miyoshi H, et al. Expression of small hairpin RNA by lentivirus-based vector confers efficient and stable gene-suppression of HIV-1 on human cells including primary non-dividing cells. Microbes Infect,2004,6(1):76-85.
    [55]Morris KV, Rossi JJ. Lentiviral mediated delivery of siRNAs for antiviral therapy. Gene Ther, 2006,13(6):553-558.
    [56]Hioki H, Kameda H, Nakamura H, et al. Efficient gene transduction of neurons by lentivirus with enhanced neuron-specific promoters. Gene Ther,2007,14(11):872-882.
    [57]Gerolami R, Uch R, Faivre J, et al. Herpes simplex virus thymidine kinase-mediated suicide gene therapy for hepatocellular carcinoma using HIV-1-derived lentiviral vectors. J Hepatol, 2004,40(2):291-297.
    [58]Harrich D, Ulich C, Garcia-Martinez LF,et al. Tat is required for efficient HIV-1 reverse transcri-ption. Embo J,1997,16(6):1224-1235.
    [59]Brandes AA, Franceschi E, Tosoni A, et al. Epidermal growth factor receptor inhibitors in neuro-oneology:hopes and disappointments. Clin Cancer Res,2008,14(4):957-960.
    [60]Fresno JA, Casado E, de Castro J, et al. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev,2004,30:193-204.
    [61]Carnero A, Blanco-Aparicio C, Rennet O, et al. The PTEN/PI3K/Akt signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets,2008,8:187-198.
    [62]Micklem DR, Lorens JB. RNAi screening for therapeutic targets in human malignancies. Curr Pharm Biotechnol,2007,8(6):337-43.
    [63]Simpson L, Parsons R. PTEN:life as a tumor suppressor. Exp Cell Res,2001,264(1):29-41.
    [64]Lytle RA, Jiang Z, Zheng X, et al. Retinamide-induced apoptosis in glioblastomas is associated with down-regulation of Bcl-xL and Bcl-2 proteins. J Neurooncol,2005,74(3):225-232.
    [65]Ching JC, Jones NL, Ceponis PJ, et al. Escherichia coli shiga-like toxins induce apoptosis and cleavage of poly (ADP-ribose) polymerase via in vitro activation of caspases. Infect Immun, 2002,70(8):4669-4677.
    [66]Zhou Y, Mehta KR, Choi AP, et al. DNA damage-induced inhibition of securin expression is mediated by p53. J Biol Chem,2003,278(1):462-70.
    [67]Ravitz MJ, Chen L, Lynch M, et al. C-myc repression of TSC2 contributes to control of translation initiation and myc-induced transformation. Clin Cancer Res,2007,67(23):4323-29.
    [68]Li A, Blow JJ. The origin of CDK regulation. Nature Cell Biology,2001,3:182-84.
    [69]Sandal T. Molecular aspects of the mammalian cell cycle and cancer. The Oncol,2002,7(2): 73-81.
    [70]Wang HY, Liu T, Malbon CC. Structure-function analysis of Frizzleds. J Cell Signal, 2006,18(7):934-41.
    [71]Imoto M, Doki Y, Jiang W, et al. Effects of cyclinDl overexpression on G1 progression related events. Exp Cell Res,1997,236:173-180.
    [72]Sandhu C, Slingerland J. Deregulation of cell cycle in cancer. Cancer Detect Prev,2000,24(2): 107-108.
    [73]Bahnassy AA, Zekri AR, Houssinis EL, et al. CyclinA and CyclinD, as significant prognostic markers in colorectal patients. BMC Gastroenteral,2004,4(1):22-24.
    [74]Tan PG,Xing Z, Li ZQ. Expression of CyclinDl in brain gliomas and its sighificance.Ai Zheng, 2004,23(1):63.
    [75]Sallinen SL, Sallinen PK, Kononen JT, et al. CyclinDl expression in astrocytomas is associated with cell proliferation activity and patient prognosis. J Pathol,1999,188(3):289-293.
    [76]王光良,倪虹,陈力.细胞周期检测点与肿瘤发生.国外医学肿瘤学分册,2004,3(31): 406-409.
    [77]Obaya AJ, Sedivy JM. Regulation of cyclin-Cdk activity in mammalian cells. Cell Mol Life Sci, 2002,59(7):126-142.
    [78]Hisano C, Tanaka R, Fujishima H, et al. Suppression of anoikis by vSrc but not by activated cHras in human gallbladder epithelial cells. Cell Biol Int,2003,27:415-421.
    [79]Beviglia L, Golubovskaya V, Xu L. Focal adhesion kinase N-terminus in breast carcinoma cells induces rounding, detachment and apoptosis. BiochemJ,2003,373:201-210.
    [80]Henshall DC, Araki T, Schinkier CK, et al. Activation of bcl-2-associated death protein and counter-response of Akt within cell populations during seizure-induced neuronal death. J Neurosci,2002,22(19):8458-8465.
    [81]Xin M, Deng X. Nicotine inactivation of the proapoptotic function of Bax through phosphorylation. J Biol Chem,2005,280(11):10781-10789.
    [82]Bartline B, Tostlebe H, Darmer D, et al. Shear stress-dependent expression of apoptosis-regu lating genes in endothelial cells. Biochem Biophys Res Commun,2000,278(3):740-746.
    [83]Li N, Banin S, Ouyang H, et al. ATM is required for NF-kappa B kinase activation in response to DNA double strand breaks. J Biol Chem,2001,276(12):8898-8903.
    [84]Gibson EM, Henson ES, Haney N, et al. Epidermal growth factor protects epithelial-derived cells from tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by inhibiting cytochrome release. Cancer Res,2002,62(2):488-496.
    [85]COFFEY JC,WANG JH,SMITH MJ, et al. Phosphoinositide 3 kinase accelerates postoperative tumor growth by inhibiting apoptosis and enhancing resistance to chemotherapy-induced apoptosis-novel role for an old enemy. J Biol Chem,2005,280(22):20968-20977.
    [86]Muise RC, Grimes HL, Bellacosa A, et al. CyclinD expression is controlled post-transcriptionally via a phosphatidyhnositol 3-kinase/Akt-dependent pathway. J Biol Chem,1998,273(45): 29864-29872.
    [87]Ikenoue T, Kariai F, Hikiba Y, et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res,2005,65:4562-4567.
    [88]Viglietto G, Motti ML, Bruni P, et al.. Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kipl)by PKB/Akt-mediated phosphorylation in breast cancer. Nat Med,2002,8:1136-1144.
    [89]Pantuck AJ, Seligson DB, Klatte T, et al. Prognostic relevance of the mTOR pathway in renal eell carcinoma:implications for molecular patient selection for targeted therapy. Cancer,2007, 109:2257-2267.
    [90]Ren H, Yang BF, Rainov NG. Receptor tyrosine kinases as therapeutic targets in malignant glioma. Rev Recent Clin Trials,2007,2:87-101.
    [91]Cheng CK, Fan QW, Weiss WA. PI3K signaling in glioma-animal models and therapeutic challenges. Brain Patho,2009,19:112-120.
    [92]Mandal M, Kim S, Younes MN, et al. The Akt inhibitor KP372-1 suppresses Akt activity and cell proliferation and induces apoptosis in thyroid cancer cells. Br J Cancer,2005,92(10): 1899-1905.
    [93]Koul D, Shen R, Bergh S, et al. Inhibition of Akt survival pathway by a small-molecule inhibitor in human glioblastoma. Mol Cancer Ther,2006,5(3):637-644.
    [1]Brandes AA, Franceschi E,Tosoni A,et al. Epidermal growth factor receptor inhibitors in neuro-oneology:hopes and disappointments. Clin Cancer Res,2008,14(4):957-960.
    [2]Richard M, Gunn HC. Insights into rhe PI3-K-PKB-mTOR signalling pathway from small molecules. J Chem Biol,2008,1:49-62.
    [3]Carnero A, Blanco AC, Rennet O, et al.The PTEN/PI3K/Akt signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets,2008,8(5):187-198.
    [4]Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet,2006,7:606-619.
    [5]Anderson KE, Jackson SP. Class I phosphoinositide 3-kinases. Int J Biochem Cell Biol,2003, 35:1028-1033.
    [6]Philipp V, Carsten B, Bernd N, et al. Assigning functional domains within the p110 regulatory subunit of phosphoinositide 3-kinase. J Biol Chem,2005,280(6):5121-5127.
    [7]Stephens L, Cooke FT, Walters R, et al. Characterization of a phosphatidylinositol-specific phosphoinositide 3-kinase from mammalian cells. Curr Biol,1994,4:203-214.
    [8]Wymann MP, Zvelebil M, Laffargue M. Phosphoinositide 3-kinase signaling-which way to target? Trends Pharmacol Sci,2003,24:366-376.
    [9]Tang X, Powelka AM, Soriano NA, et al. PTEN, but not SHIP2, suppresses insulin signaling through the phosphatidylinositol 3-kinase/Akt pathway in 3T3-L1 adipocytes. J Biol Chem, 2005,280:22523-22529.
    [10]Sasaoka T, Wada T, Murakami S, et al. SH-2-containing inositol phosphatase 2 predominantly regulates Akt2, and not Akt 1, phosphorylation at the plasma membrane in response to insulin in 3T3-L1 adipocytes. J Biol Chem,2004,279:14835-14843.
    [11]Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signaling controls tumour cell growth. Nature, 2006,441:424-430.
    [12]Scheid MP, Woodgett JR. Unravelling the activation mechanisms of protein kinase B/Akt. FEBS Lett,2003,546:108-112.
    [13]Sarbassov DD, Guertin DA, Ali SM, et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science,2005,307:1098-1101.
    [14]Guertin DA, Stevens DM, Thoreen CC, et al. Ablation in mice of the mTOR components raptor, rictor or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCa but not S6K1. Dev Cell,2006,11:859-871.
    [15]Vander HE, Lee S, Bandhakavi S, et al. Insulin signaling to mTOR mediated by the Akt/PKB substrate PRAS40. Nature Cell Biol,2007,9:316-323.
    [16]Pantuck AJ, Seligson DB, Klatte T, et al. Prognostic relevance of the mTOR pathway in renal eell carcinoma:implications for molecular patient selection for targeted therapy. Cancer,2007, 109:2257-2267.
    [17]Ren H, Yang BF, Rainov NG. Receptor tyrosine kinases as therpeutic targets in malignant glioma. Rev Recent Clin Trials,2007,2:87-101.
    [18]Knobbe CB, Reifenberger J, Blaschke B, et al. Hypermethylation and transcriptional downregulation of the carboxyl-terminal modulator protein gene in glioblastomas. J Nat Cancer lnst,2004,96(6):483-486.
    [19]Pim D, Massimi P, Dilworth SM, et al. Activation of the protein kinase B pathway by the HPV-16 E7 oncoprotein occurs through a mechanism involving interaction with PP2A. Oncogene,2005,24(53):7830-7838.
    [20]Shtivelman E, Sussman J, Stokoe D. A role for PI3-kinase and PKB activity in the G2/M phase of the cell cycle. Curr Biol,2002,12(11):919-924.
    [21]Muise RC, Grimes HL, Bellacosa A, et al. CyclinD expression is controlled post-transcriptionally via a phosphatidyhnositol 3-kinase/Akt-dependent pathway. J Biol Chem,1998,273(45): 29864-29872.
    [22]MAYO LD, DONNER DB. A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of mdm2 from the cyto-plasm to the nucleus. Proc Natl Acad Sci,2001,98(20):11598-115603.
    [23]XIN M, DENG X. Nicotine inactivation of the proapoptotic function of Bax through phosphorylation. J Biol Chem,2005,280(11):10781-10789.
    [24]Burgering BM, Medema RH. Decisions on life and death:FOXO forkhead transcription factors are in command when PKB/Akt is of duty. J Leukoc Biol,2003,73(6):689-701.
    [25]Jeong SJ, Pise-Masison CA, Radonovich MF, et al. Activated Akt regulates NF-κB activation, p53 inhibition an d cell survival in HTLV-1-transformed cells. Oncogene,2005,24(4): 6719-6728.
    [26]Hill MM, Hemmings BA. Inhibition of protein kinase B/Akt implications for cancer therapy. Pharmacol Ther,2002,93:243-251.
    [27]Zhang R, Xu Y, Ekman N, et al. Etk/Bmx transactivates vascular endothelial growth factor 2 and recruits phosphatidylinositol 3-kinase to mediate the tumor nec rosis factor induced angiogenic pathway. J Biol Chem,2003,278:51267-51276.
    [28]Grille SJ, Bellacosa A, Upson J, et al. The protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res,2003,63:2172-2178.
    [29]Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxia-inducible factor 1 alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/Akt/FRAP pathway in human prostate cancer cells:plications for tumor angiogenesis and therapeutics. Cancer Res, 2000,60(6):1541-1545.
    [30]Chung TW, Lee YC, Kim CH. Hepatitis B viral HBx induces matrix metalloproteinase-9 gene expression through activation Of ERK and PI3K/AKT pathways: involvement of invasive potential. Fasebeb J,2004,18:1123-1125.
    [31]Holland EC, Celestino J, Dai C, et al. Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nat Genet,2000,25(1):55-57.
    [32]Petch AK, Sohail M, Hughes MD, et al. Messenger RNA expression profiling of genes involved in epidermal growth factor receptor signalling in human cancer cells treated with scanning array-designed antisense oligonucleotides [J]. Biochem Pharmacol,2003,66(5):819-830.
    [33]Haas KD, Shalev N, Wong M, et al. Protein kinaseB(PKB/Akt) activity is elevated in glioblastorna cells due to mutation of the tumor suppressor PTENz/MMAC. Curr Biol,1998, 8(21):1195-1198.
    [34]Kubiatowski T, Jang T, Laehyankar MB, et al. Association of increased phosphatidylinositol 3-kinase signaling with increased invasiveness and gelatinase activity in malignant gliomas. J Neurosurg,2001,95(3):480-488.
    [35]Ermoian RP, Fumiss CS, Lambom KR, et al. Dysregulation of PTEN and protein kinase B is associated with glioma histology and patient survival. Clin Cancer Res,2002,8(5):1100-1106.
    [36]Ralph PE, Tania K, Kathleen RL, et al. Signal transduction molecules in gliomas of all grades. J Neuroomcol,2009,91:19-26.
    [37]Shingu T, Yamada K, Hara N, et al. Synergistic augmentation of antimicrotubule agent-induced cytotoxicity by a phosphoinositide 3-kinase inhibitor in human malignant glioma cells. Cancer Res,2003,63(14):4044-4047.
    [38]Cheng CK, Fan QW, Weiss WA. PI3K Signaling in Glioma-Animal Models and Therapeutic Challenges. Brain Pathology,2009,19:112-120.
    [39]Koul D, Shen R, Bergh S, et al. Inhibition of Akt survival pathway by a small-molecule inhibitor in human glioblastoma. Mol Cancer Ther,2006,5(3):637-644.
    [40]康春生,浦佩玉,李捷,等.反义及显性调节AKT2 RNA对胶质瘤细胞增殖抑制作用的体外研究.癌症,2004,23(11):1267-1272.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700