依赖p53的癌基因Wip1在人脑胶质瘤细胞恶性增殖机制中的作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的胶质瘤通常伴有p53的突变。Wip1基因抑制并负反馈调节p53功能而表现出癌基因的特性。本研究拟探讨Wip1基因在脑胶质瘤组织中的表达及其与p53/p14ARF信号通路失活的关系。
     方法实时定量PCR及Western Blot法检测8例正常脑组织及52例不同病理分级的脑胶质瘤组织中Wip1基因的表达,直接测序法检测p53基因的突变情况,逆转录PCR及甲基化PCR检测p14ARF的表达情况,统计学分析其相关性。
     结果52例胶质瘤样本中p53/p14ARF信号通路失活30例(57.7%)。22例无p53或p14ARF改变的样本中,11例伴有Wip1 mRNA过表达;而p53/p14ARF信号通路失活30例中,仅1例(3.3%)伴有Wip1 mRNA过表达。Wip1在脑胶质瘤中存在着选择性的过表达,过表达的Wip1主要与p53野生型相关而与p14ARF的甲基化状态及转录子表达无关。
     结论Wip1在脑胶质瘤中存在选择性的过表达;Wip1基因在脑胶质瘤中的过表达与p53/p14ARF信号通路的失活有关,Wip1基因可能抑制p53/p14ARF信号通路。
     目的构建人Wip1基因的RNA干扰(RNAi)慢病毒载体,测定病毒滴度,并对不同靶点进行筛选,挑选干扰效率最高的靶点。
     方法根据人Wip1基因序列,设计三对Wip1基因特异性RNAi靶序列,经退火制备双链DNA oligo,连接到pFU-GW-iRNA载体,转化后经PCR鉴定,阳性克隆测序证实,慢病毒包装转染293T细胞,获得病毒上清并测定其滴度。将病毒上清感染人胶质瘤细胞株U251和U-87MG,实时定量PCR及Western blot鉴定RNA干扰效率,筛选出基因沉默效率最高的慢病毒载体。
     结果PCR扩增和测序结果表明成功构建Wip1基因慢病毒干扰载体,慢病毒载体包装获得的病毒上清滴度在3-8×108TU/ml。可以有效地沉默U251细胞及U-87MG细胞中Wip1基因的表达,构建的RNA干扰慢病毒载体感染U251细胞后Wip1基因的mRNA表达量同对照组相比较分别为36.3%、32.9%、23.8%;在U-87MG中则分别为48.8%、36.7%和23.7%。Western blot结果显示Wip1蛋白表达量在感染细胞7d后显著下降。
     结论成功构建人Wip1基因的RNA干扰慢病毒载体并筛选出最佳靶点,为进一步研究Wip1基因在胶质瘤细胞中的作用提供可靠的实验平台。
     目的研究Wip1基因在人脑胶质瘤细胞恶性增殖中的作用及探讨其可能的作用机制。
     方法Wip1基因RNA干扰慢病毒载体感染胶质瘤细胞U251及U-87MG。CCK-8法(cell counting kit-8)及克隆形成实验检测细胞的增殖能力;流式细胞术Annexin V-APC/PI双标法及Hoechst 33258染色检测细胞的凋亡情况;流式细胞术PI单染法检测细胞周期分布;Transwell实验检测细胞的侵袭性;实时定量PCR法检测Wip1基因沉默后差异性基因的mRNA表达;Western blot检测Wip1可能作用途径的基因的蛋白表达。SPSS 13.0软件分析统计学差异。
     结果胶质瘤中Wip1基因的表达被有效沉默。Wip1基因沉默的胶质瘤细胞增殖变慢,在U-87MG细胞中更加明显;转染慢病毒载体的U-87MG和U251细胞在4d时凋亡无明显变化,但7d和10d时出现明显的凋亡,且凋亡率随着时间的延长而逐渐增加,U-87MG细胞的凋亡率为52.2%和67.1%,U251则分别为26.1%和38.2%。U-87MG在慢病毒感染后10d与U251细胞相比,凋亡率明显要高(P<0.05);Hoechst 33258染色显示Wip1基因沉默后的胶质瘤细胞中可以观察到典型的细胞凋亡的形态学改变如核浓缩、核碎裂;细胞周期分析显示Wip1基因沉默对U251细胞周期无明显影响,而U-87MG细胞则在10d时出现明显的S期增多和Gl、G2期细胞的减少;U-87MG细胞在Wip1基因沉默后出现明显的体外侵袭力下降。Wip1基因沉默后,U251细胞PIK3R1基因出现上调,伴有CDKN2A和p14ARF基因的下调,而在U-87MG细胞中他们的表达均上调;Western blot结果显示两种胶质瘤细胞中的p38MAPK的表达均上调,而Akt的表达均下调,但是p-Akt(Ser473)却只在U-87MG细胞中存在上调。在U-87MG细胞中p53及p-p53 (Ser15)表达均上调,但在U251细胞中无明显变化。
     结论Wip1对胶质瘤细胞的增殖、凋亡和侵袭具有重要的作用,并且这种作用主要与其调节p53的功能有关。
Objective Patients with gliomas often have mutations in the p53 gene. Wild-type p53-induced phosphatase 1 (Wip1 or PPM1D) encodes a negative regulator of p53. Wip1 over-expression inhibits p53 function and reduces selection for p53 mutations during cancer progression. Wip1 may have oncogenic function. To clarify the correlation of Wip1 with p53/p14ARF pathway disruption in glioma, the expression of Wip1 and p53/p14ARF pathway alterations have been investigated.
     Methods Tumor samples of 52 patients with primary glioams and 8 samples of normal brain tissues were examined for p53 mutations, p14ARF expression, and Wip1 expression. Direct sequencing of region from exons 5 to 8 of the p53 gene was performed on the genomic DNA in each tumor. The DNA methylation states of the CpG islands of the p14ARF gene were determined by methylation-specific PCR. All tumor specimens were analyzed for expression of Wip1 by real-time quantitative RCR, western blot and immunohistochemical staining.
     Results Disruption of the p53/p14ARF pathway was detected in 57.7%(30 in 52) of samples from patients with gliomas. Among the 22 cases without p53 and p14ARF alterations,11 (50%) had Wip1 mRNA over-expression. In tumors with p53 or p14ARF disruptions, Wip1 mRNA was over-expressed in only 1 case out of 30 (3.3%). Higher levels of Wip1 were associated with wild-type p53 but not with lower levels of expression of p14ARF or aberrant promoter hypermethylation of the p14ARF gene.
     Conclusion Wip1 is selectively over-expressed in tumors without alterations in p53 or p14ARF.Wip1 may inhibit the P53/p14ARF pathway.
     Objective To construct the lentiviral expression vector for RNA interference (RNAi) of Wip1 gene in glioma cells and select the optimal target sequence of Wip1 gene that is the most effective for RNAi.
     Methods Three double-stranded oligo DNAs were designed and synthesized according to the sequence of Wip1 gene and cloned into the pFU-GW-iRNA vector. After verification of the positive clones by PCR and sequence analysis, the verified plasmids were transfected into 293 T cells, the lenti virus was produced and the titer of virus was determined. The lentivirus was used to infect the glioma cell lines U251 and U-87MG. Real-time quantitative PCR and Western blot were performed to determine Wip1 expression levels in the virus infected glioma cells and the optimal interfering target was selected.
     Results Three recombinant lentiviral vector expressing shRNAs against Wip1 gene were obtained and confirmed by DNA sequencing. The titer of virus was 3-8×10 8TU/ml. Wip1 mRNA and protein expressions in U251 and U-87MG cells after infected with lentiviral vector were decreased remarkably. The mRNA expression in U251 cells was 36.3%,32.9% and 23.8% respectively, compared with the control group. And in U-87MG was 48.8%,36.7% and 23.7%. Western blot showed that Wip1 protein expression decreased 7 d after the infection significantly.
     Conclusion The lentiviral shRNA expression vector targeting human Wip1 gene capable of stable Wip1 gene silencing in glioma cells has been successfully constructed, which provides a basis for further study of mechanisms that Wip1 gene acts for malignant proliferation of glioma cells.
     Objective To detect the roles that Wip1 plays in the malignant growth of glioma cells and explore the possible mechanisms.
     Methods Glioma cells U251 and U-87MG were infected with Wip1 RNAi lentiviral vector. The proliferation activities of cells with Wip1 silencing were detected by cell counting kit-8 and clone formation experiments. Cell apoptosis was determined by flow cytometry AnnexinⅤ-APC/PI double labeling method and Hoechst 33258 staining. Cell cycle distributions were analyzed by flow cytometry PI staining. Cell invasiveness in glioma cells was determined by Transwell invasion assay. Real-time quantitative PCR was used to identify the mRNA expressions of differentially expressed genes after Wip1 silencing. Western blot was performed to detect the expressions of passageway proteins that Wipl may effect in. SPSS 13.0 software was used to analyze the significant difference.
     Results The expressions of Wip1gene in glioma cells were silenced effectively. Cells with Wip1 silencing, especially U-87MG cells, had reduced proliferation ability compared with mock cells. The number of apoptotic cells determined at 4 d after infection with Wip1 RNAi-R3 and NC lentiviral were not of the difference between them. At 7 d and 10 d, both U-87MG and U251 cells after Wip1 silencing showed significantly increased apoptotic cells compared with the mock cells. And the amount of apoptotic cells increased as the time periods extend. The apoptotic cells accounted for 52.2% and 67.1% of all the U-87MG cells with Wip1 silencing, while accounting for 26.1% and 38.2% in U251 cells. And U-87MG cells were more apoptotic than U251 cells 10 days after the RNAi-R3 lentiviral infection (P<0.05). Both U-87MG and U251 cells after Wip1 silencing had undergone extensive DNA strand breakage and nuclei-shrunk, characteristic changes of apoptosis. No significant changes of cell cycle distributions analyzed by flow cytometry were detected in U251 cells after Wip1 silencing. But increased S phase and decreased G1 and G2 phases were detected in U-87MG 10 d after the infection. U-87MG cells showed decreased invasiveness after Wip1 silencing. After Wip1 silencing, PIK3R1 gene mRNA expression was upregulated, while CDKN2A and p14ARF were downregulated in U251 cells. But in U-87MG cells, they all showed increased tendencies. Western blot showed that protein expression of p38MAPK in both U251 and U-87MG cells was increased, but expression of Akt was downregulated. Expression of p-Akt(Ser473)was only increased in U-87MG cells. The expressions of p53 and p-p53 (Ser 15) were both upregulated in U-87MG cell but remained unchanged in U251 cells.
     Conclusion Wip1 plays a important role in proliferation, apoptosis and invasiveness in glioma cells, and the effects are of much relevance with the fact that Wip1 could inhibit the function of p53.
引文
1. Dehdashti AR, Hegi ME, Regli L, et al. New trends in the medical management of glioblastoma multiforme:the role of temozolomide chemotherapy. Neurosurg Focus, 2006,20(4):E6.
    2. Pietsch EC, Sykes SM, McMahon SB, et al. The p53 family and programmed cell death. Oncogene,2008,27(50):6507-6521.
    3. Soussi T, Wiman KG. Shaping genetic alterations in human cancer:the p53 mutation paradigm. Cancer Cell,2007,12(4):303-312.
    4. Kato H, Kato S, Kumabe T, et al. Functional evaluation of p53 and PTEN gene mutations in gliomas. Clin Cancer Res,2000,6(10):3937-3943.
    5. Yue WY, Yu SH, Zhao SG, et al. Molecular markers relating to malignant progression in Grade Ⅱ astrocytoma. J Neurosurg,2009,110(4):709-714.
    6. Li J, Yang Y, Peng Y, et al. Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet,2002,31(2):133-134.
    7. Bulavin DV, Phillips C, Nannenga B, et al. Inactivation of the Wipl phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet,2004,36(4):343-350.
    8. Rauta J, Alarmo EL, Kauraniemi P, et al. The serine-threonine protein phosphatase PPM ID is frequently activated through amplification in aggressive primary breast tumours. Breast Cancer Res Treat,2006,95(3):257-263.
    9. Saito-Ohara F, Imoto I, Inoue J, et al. PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res,2003,63(8):1876-1883.
    10. Hirasawa A, Saito-Ohara F, Inoue J, et al. Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM ID and APPBP2 as likely amplification targets. Clin Cancer Res,2003,9(6):1995-2004.
    11. Castellino RC, De Bortoli M, Lu X, et al. Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D. J Neurooncol,2008,86(3):245-256.
    12. Lu X, Nguyen TA, Zhang X, et al. The Wipl phosphatase and Mdm2:cracking the "Wip" on p53 stability. Cell Cycle,2008,7(2):164-168.
    13. Bulavin DV, Demidov ON, Saito S, et al. Amplification of PPM 1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet,2002,31(2):210-215.
    14. Marino S. Medulloblastoma:developmental mechanisms out of control. Trends Mol Med,2005,11(1):17-22.
    15. Fiscella M, Zhang H, Fan S, et al. Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA,1997,94(12):6048-6053.
    16. Parssinen J, Alarmo EL, Khan S, et al. Identification of differentially expressed genes after PPM1D silencing in breast cancer. Cancer Lett,2008,259(1):61-70.
    17. Parssinen J, Alarmo EL, Karhu R, et al. PPM1D silencing by RNA interference inhibits proliferation and induces apoptosis in breast cancer cell lines with wild-type p53. Cancer Genet Cytogenet,2008,182(1):33-39.
    18. Lu X, Ma O, Nguyen TA, et al. The Wipl Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell,2007,12(4):342-354.
    19. Oliva-Trastoy M, Berthonaud V, Chevalier A, et al. The Wip1 phosphatase (PPM1D) antagonizes activation of the Chk2 tumour suppressor kinase. Oncogene,2007, 26(10):1449-1458.
    20. Rossi M, Demidov ON, Anderson CW, et al. Induction of PPM1D following DNA-damaging treatments through a conserved p53 response element coincides with a shift in the use of transcription initiation sites. Nucleic Acids Res,2008, 36(22):7168-7180.
    1. Zheng H, Ying H, Yan H, et al. p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation. Nature,2008,455(7216):1129-1133.
    2. Fulci G, Ishii N, Van Meir EG. p53 and brain tumors:from gene mutations to gene therapy. Brain Pathol,1998,8(4):599-613.
    3. Harris SL, Levine AJ. The p53 pathway:positive and negative feedback loops. Oncogene,2005,24(17):2899-2908.
    4. Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53:ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell,1998,92(6):725-734.
    5. Kato H, Kato S, Kumabe T, et al. Functional evaluation of p53 and PTEN gene mutations in gliomas. Clin Cancer Res,2000,6(10):3937-3943.
    6. Fiscella M, Zhang H, Fan S, et al. Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA,1997,94(12):6048-6053.
    7. Saito-Ohara F, Imoto I, Inoue J, et al. PPM ID is a potential target for 17q gain in neuroblastoma. Cancer Res,2003,63(8):1876-1883.
    8. Hirasawa A, Saito-Ohara F, Inoue J, et al. Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM ID and APPBP2 as likely amplification targets. Clin Cancer Res,2003,9(6):1995-2004.
    9. Castellino RC, De Bortoli M, Lu X, et al. Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D. J Neurooncol,2008,86(3):245-256.
    10. Lu X, Nguyen TA, Zhang X, et al. The Wip1 phosphatase and Mdm2:cracking the "Wip" on p53 stability. Cell Cycle,2008,7(2):164-168.
    11. Holstege H, Joosse SA, van Oostrom CT, et al. High incidence of protein-truncating TP53 mutations in BRCAl-related breast cancer. Cancer Res,2009,69(8):3625-3633.
    12. Esteller M, Tortola S, Toyota M, et al. Hypermethylation-associated inactivation of p14(ARF) is independent of p16(INK4a) methylation and p53 mutational status. Cancer Res,2000,60(1):129-133.
    13. Taylor WR, Stark GR. Regulation of the G2/M transition by p53. Oncogene,2001, 20(15):1803-1815.
    14. Watanabe T, Katayama Y, Yoshino A, et al. Deregulation of the TP53/p14ARF tumor suppressor pathway in low-grade diffuse astrocytomas and its influence on clinical course. Clin Cancer Res,2003,9(13):4884-4890.
    15.Sun Y. p53 and its downstream proteins as molecular targets of cancer. Mol Carcinog, 2006,45(6):409-415.
    16. Soussi T, Wiman KG. Shaping genetic alterations in human cancer:the p53 mutation paradigm. Cancer Cell,2007,12(4):303-312.
    17. Fulci G, Labuhn M, Maier D, et al. p53 gene mutation and ink4a-arf deletion appear to be two mutually exclusive events in human glioblastoma. Oncogene,2000, 19(33):3816-3822.
    18. Cairns P, Polascik TJ, Eby Y, et al. Frequency of homozygous deletion at p16/CDKN2 in primary human tumours. Nat Genet,1995, 11(2):210-212.
    19. Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science,2008,321(5897):1807-1812.
    20. Amatya VJ, Takeshima Y, Inai K. Methylation of pl4(ARF) gene in meningiomas and its correlation to the p53 expression and mutation. Mod Pathol,2004,17(6):705-710.
    21. Choi J, Nannenga B, Demidov ON, et al. Mice deficient for the wild-type p53-induced phosphatase gene (Wipl) exhibit defects in reproductive organs, immune function, and cell cycle control. Mol Cell Biol,2002,22(4):1094-1105.
    22. Yu E, Ahn YS, Jang SJ, et al. Overexpression of the wipl gene abrogates the p38 MAPK/p53/Wipl pathway and silences p16 expression in human breast cancers. Breast Cancer Res Treat,2007,101(3):269-278.
    23. Rossi M, Demidov ON, Anderson CW, et al. Induction of PPM1D following DNA-damaging treatments through a conserved p53 response element coincides with a shift in the use of transcription initiation sites. Nucleic Acids Res,2008, 36(22):7168-7180.
    24. Bulavin DV, Phillips C, Nannenga B, et al. Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet,2004,36(4):343-350.
    25. Lu X, Nannenga B, Donehower LA. PPM1D dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints. Genes Dev,2005,19(10):1162-1174.
    1. Saito-Ohara F, Imoto I, Inoue J, et al. PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res,2003,63(8):1876-1883.
    2. Hirasawa A, Saito-Ohara F, Inoue J, et al. Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification targets. Clin Cancer Res,2003,9(6):1995-2004.
    3. Castellino RC, De Bortoli M, Lu X, et al. Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D. J Neurooncol,2008,86(3):245-256.
    4. Qin XF, An DS, Chen IS, et al. Inhibiting HIV-1 infection in human T cells by lenti viral-mediated delivery of small interfering RNA against CCR5. Proc Natl Acad Sci USA,2003,100(1):183-188.
    5. Cenciarelli C, Budoni M, Mercanti D, et al. In vitro analysis of mouse neural stem cells genetically modified to stably express human NGF by a novel multigenic viral expression system. Neurol Res,2006,28(5):505-512.
    6. Meunier A, Latremoliere A, Dominguez E, et al. Lentiviral-mediated targeted NF-kappaB blockade in dorsal spinal cord glia attenuates sciatic nerve injury-induced neuropathic pain in the rat. Mol Ther,2007,15(4):687-697.
    7. Yarosh W, Barrientos T, Esmailpour T, et al. TBX3 is overexpressed in breast cancer and represses p14 ARF by interacting with histone deacetylases. Cancer Res,2008, 68(3):693-699.
    8. Fiscella M, Zhang H, Fan S, et al. Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA,1997,94(12):6048-6053.
    9. Li J, Yang Y, Peng Y, et al. Oncogenic properties of PPM ID located within a breast cancer amplification epicenter at 17q23. Nat Genet,2002,31(2):133-134.
    10. Lu X, Nguyen TA, Moon SH, et al. The type 2C phosphatase Wipl:an oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev,2008,27(2):123-135.
    11. Lu X, Ma O, Nguyen TA, et al. The Wip1 Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell,2007,12(4):342-354.
    12. Gartel AL, Kandel ES. RNA interference in cancer. Biomol Eng,2006,23(1):17-34.
    13. Wang X, Tang R, Xue Z, et al. Lentivector-mediated RNAi efficiently downregulates expression of murine TNF-alpha gene in vitro and in vivo. J Huazhong Univ Sci Technolog Med Sci,2009,29(1):112-117.
    14. Li M, Rossi JJ. Lentiviral vector delivery of siRNA and shRNA encoding genes into cultured and primary hematopoietic cells. Methods Mol Biol,2008,433287-299.
    1. Fiscella M, Zhang H, Fan S, et al. Wipl, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad SciUSA,1997,94(12):6048-6053.
    2. Choi J, Nannenga B, Demidov ON, et al. Mice deficient for the wild-type p53-induced phosphatase gene (Wipl) exhibit defects in reproductive organs, immune function, and cell cycle control. Mol Cell Biol,2002,22(4):1094-1105.
    3. Li J, Yang Y, Peng Y, et al. Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet,2002,31(2):133-134.
    4. Lu X, Ma O, Nguyen TA, et al. The Wip1 Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell,2007,12(4):342-354.
    5. Parssinen J, Alarmo EL, Khan S, et al. Identification of differentially expressed genes after PPM1D silencing in breast cancer. Cancer Lett,2008,259(1):61-70.
    6. Saito-Ohara F, Imoto I, Inoue J, et al. PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res,2003,63(8):1876-1883.
    7. Hirasawa A, Saito-Ohara F, Inoue J, et al. Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM ID and
    APPBP2 as likely amplification targets. Clin Cancer Res,2003,9(6):1995-2004.
    8. Castellino RC, De Bortoli M, Lu X, et al. Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D. J Neurooncol,2008,86(3):245-256.
    9. Lu X, Nguyen TA, Moon SH, et al. The type 2C phosphatase Wip1:an oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev,2008,27(2):123-135.
    10. Bulavin DV, Demidov ON, Saito S, et al. Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet,2002,31(2):210-215.
    11. Barlund M, Nupponen NN, Karhu R, et al. Molecular cytogenetic mapping of 24 CEPH YACs and 24 gene-specific large insert probes to chromosome 17. Cytogenet Cell Genet,1998,82(3-4):189-191.
    12. Taylor WR, Stark GR. Regulation of the G2/M transition by p53. Oncogene,2001, 20(15):1803-1815.
    13. Parssinen J, Alarmo EL, Karhu R, et al. PPM1D silencing by RNA interference inhibits proliferation and induces apoptosis in breast cancer cell lines with wild-type p53. Cancer Genet Cytogenet,2008,182(1):33-39.
    14. Batista LF, Roos WP, Christmann M, et al. Differential sensitivity of malignant glioma cells to methylating and chloroethylating anticancer drugs:p53 determines the switch by regulating xpc, ddb2, and DNA double-strand breaks. Cancer Res,2007, 67(24):11886-11895.
    15. Tanaka K, Sasayama T, Mizukawa K, et al. Specific mTOR inhibitor rapamycin enhances cytotoxicity induced by alkylating agent 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea (ACNU) in human U251 malignant glioma cells. J Neurooncol,2007,84(3):233-244.
    16. Dehdashti AR, Hegi ME, Regli L, et al. New trends in the medical management of glioblastoma multiforme:the role of temozolomide chemotherapy. Neurosurg Focus, 2006,20(4):E6.
    17. Kato H, Kato S, Kumabe T, et al. Functional evaluation of p53 and PTEN gene mutations in gliomas. Clin Cancer Res,2000,6(10):3937-3943.
    18. Yue WY, Yu SH, Zhao SG, et al. Molecular markers relating to malignant progression in Grade Ⅱ astrocytoma. J Neurosurg,2009,110(4):709-714.
    19. Lu X, Nannenga B, Donehower LA. PPMID dephosphorylates Chkl and p53 and abrogates cell cycle checkpoints. Genes Dev,2005,19(10):1162-1174.
    20. Takekawa M, Adachi M, Nakahata A, et al. p53-inducible wipl phosphatase mediates a negative feedback regulation of p38 MAPK-p53 signaling in response to UV radiation. EMBO J,2000,19(23):6517-6526.
    21.Bulavin DV, Phillips C, Nannenga B, et al. Inactivation of the Wipl phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet,2004,36(4):343-350.
    22. Lu X, Nguyen TA, Zhang X, et al. The Wip1 phosphatase and Mdm2:cracking the "Wip" on p53 stability. Cell Cycle,2008,7(2):164-168.
    23. Bulavin DV, Saito S, Hollander MC, et al. Phosphorylation of human p53 by p38 kinase coordinates N-terminal phosphorylation and apoptosis in response to UV radiation. EMBO J,1999,18(23):6845-6854.
    24. Carpten JD, Faber AL, Horn C, et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature,2007,448(7152):439-444.
    25. Fu Y, Zhang Q, Kang C, et al. Inhibitory effects of adenovirus mediated COX-2, Aktl and PIK3R1 shRNA on the growth of malignant tumor cells in vitro and in vivo. Int J Oncol,2009,35(3):583-591.
    26. Le Guezennec X, Bulavin DV. WIP1 phosphatase at the crossroads of cancer and aging. Trends Biochem Sci,35(2):109-114.
    27. Lu X, Bocangel D, Nannenga B, et al. The p53-induced oncogenic phosphatase PPM1D interacts with uracil DNA glycosylase and suppresses base excision repair. Mol Cell, 2004,15(4):621-634.
    28. Diao FY, Xu M, Hu Y, et al. The molecular characteristics of polycystic ovary syndrome (PCOS) ovary defined by human ovary cDNA microarray. J Mol Endocrinol, 2004,33(1):59-72.
    29. Tan DS, Lambros MB, Rayter S, et al. PPM1D is a potential therapeutic target in ovarian clear cell carcinomas. Clin Cancer Res,2009,15(7):2269-2280.
    1. Pietsch EC, Sykes SM, McMahon SB, et al. The p53 family and programmed cell death. Oncogene,2008,27(50):6507-6521.
    2. Soussi T, Wiman KG. Shaping genetic alterations in human cancer:the p53 mutation paradigm. Cancer Cell,2007,12(4):303-312.
    3. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell,1997, 88(3):323-331.
    4. Lozano G, Elledge SJ. p53 sends nucleotides to repair DNA. Nature,2000, 404(6773):24-25.
    5. Ljungman M. Dial 9-1-1 for p53:mechanisms of p53 activation by cellular stress. Neoplasia,2000,2(3):208-225.
    6. Appella E, Anderson CW. Post-translational modifications and activation of p53 by genotoxic stresses. Eur J Biochem,2001,268(10):2764-2772.
    7. Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature,2004, 432(7015):307-315.
    8. Vousden KH, Lu X. Live or let die:the cell's response to p53. Nat Rev Cancer,2002, 2(8):594-604.
    9. Schuler M, Green DR. Mechanisms of p53-dependent apoptosis. Biochem Soc Trans, 2001,29(Pt 6):684-688.
    10. Bond GL, Hu W, Levine AJ. MDM2 is a central node in the p53 pathway:12 years and counting. Curr Cancer Drug Targets,2005,5(1):3-8.
    11. Toledo F, Wahl GM. Regulating the p53 pathway:in vitro hypotheses, in vivo veritas. Nat Rev Cancer,2006,6(12):909-923.
    12. de Rozieres S, Maya R, Oren M, et al. The loss of mdm2 induces p53-mediated apoptosis. Oncogene,2000,19(13):1691-1697.
    13. Lu X, Ma O, Nguyen TA, et al. The Wip1 Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell,2007,12(4):342-354.
    14. Fiscella M, Zhang H, Fan S, et al. Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA,1997,94(12):6048-6053.
    15. Yoda A, Xu XZ, Onishi N, et al. Intrinsic kinase activity and SQ/TQ domain of Chk2 kinase as well as N-terminal domain of Wipl phosphatase are required for regulation of Chk2 by Wip1. J Biol Chem,2006,281(34):24847-24862.
    16. Yamaguchi H, Minopoli G, Demidov ON, et al. Substrate specificity of the human protein phosphatase 2Cdelta, Wipl. Biochemistry,2005,44(14):5285-5294.
    17. Choi J, Appella E, Donehower LA. The structure and expression of the murine wildtype p53-induced phosphatase 1 (Wipl) gene. Genomics,2000,64(3):298-306.
    18. Bulavin DV, Demidov ON, Saito S, et al. Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet,2002,31(2):210-215.
    19. Moorhead GB, Trinkle-Mulcahy L, Ulke-Lemee A. Emerging roles of nuclear protein phosphatases. Nat Rev Mol Cell Biol,2007,8(3):234-244.
    20. Barford D, Das AK, Egloff MP. The structure and mechanism of protein phosphatases: insights into catalysis and regulation. Annu Rev Biophys Biomol Struct,1998, 27133-164.
    21. Jackson MD, Denu JM. Molecular reactions of protein phosphatases--insights from structure and chemistry. Chem Rev,2001,101 (8):2313-2340.
    22. Song JY, Han HS, Sabapathy K, et al. Expression of a homeostatic regulator, Wipl (wild-type p53-induced phosphatase), is temporally induced by c-Jun and p53 in response to UV irradiation. J Biol Chem,285(12):9067-9076.
    23. Takekawa M, Maeda T, Saito H. Protein phosphatase 2Calpha inhibits the human stress-responsive p38 and JNK MAPK pathways. EMBO J,1998,17(16):4744-4752.
    24. Hanada M, Kobayashi T, Ohnishi M, et al. Selective suppression of stress-activated protein kinase pathway by protein phosphatase 2C in mammalian cells. FEBS Lett, 1998,437(3):172-176.
    25. Takekawa M, Adachi M, Nakahata A, et al. p53-inducible wipl phosphatase mediates a negative feedback regulation of p38 MAPK-p53 signaling in response to UV radiation. EMBO J,2000,19(23):6517-6526.
    26. Bulavin DV, Saito S, Hollander MC, et al. Phosphorylation of human p53 by p38 kinase coordinates N-terminal phosphorylation and apoptosis in response to UV radiation. EMBO J,1999,18(23):6845-6854.
    27. Li J, Yang Y, Peng Y, et al. Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet,2002,31(2):133-134.
    28. Soussi T, Peyrat JP, Lubin R, et al. [P53 antibodies:a new method for the analysis of alterations of the p53 gene:application to breast cancer]. Pathol Biol (Paris),1996, 44(4):232-234.
    29. Hirasawa A, Saito-Ohara F, Inoue J, et al. Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification targets. Clin Cancer Res,2003,9(6):1995-2004.
    30. Saito-Ohara F, Imoto I, Inoue J, et al. PPM ID is a potential target for 17q gain in neuroblastoma. Cancer Res,2003,63(8):1876-1883.
    31. Loukopoulos P, Shibata T, Katoh H, et al. Genome-wide array-based comparative genomic hybridization analysis of pancreatic adenocarcinoma:identification of genetic indicators that predict patient outcome. Cancer Sci,2007,98(3):392-400.
    32. Ehrbrecht A, Muller U, Wolter M, et al. Comprehensive genomic analysis of desmoplastic medulloblastomas:identification of novel amplified genes and separate evaluation of the different histological components. J Pathol,2006,208(4):554-563.
    33. Mendrzyk F, Radlwimmer B, Joos S, et al. Genomic and protein expression profiling identifies CDK6 as novel independent prognostic marker in medulloblastoma. J Clin Oncol,2005,23(34):8853-8862.
    34. Castellino RC, De Bortoli M, Lu X, et al. Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D. J Neurooncol,2008,86(3):245-256.
    35. Bulavin DV, Phillips C, Nannenga B, et al. Inactivation of the Wipl phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet,2004,36(4):343-350.
    36. Nannenga B, Lu X, Dumble M, et al. Augmented cancer resistance and DNA damage response phenotypes in PPM1D null mice. Mol Carcinog,2006,45(8):594-604.
    37. Lu X, Nannenga B, Donehower LA. PPM ID dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints. Genes Dev,2005,19(10):1162-1174.
    38. Fujimoto H, Onishi N, Kato N, et al. Regulation of the antioncogenic Chk2 kinase by the oncogenic Wipl phosphatase. Cell Death Differ,2006,13(7):1170-1180.
    39. Oliva-Trastoy M, Berthonaud V, Chevalier A, et al. The Wip1 phosphatase (PPM1D) antagonizes activation of the Chk2 tumour suppressor kinase. Oncogene,2007, 26(10):1449-1458.
    40. Joe Y, Do MH, Seo E, et al. Fenofibrate antagonizes Chk2 activation by inducing Wip1 expression:implications for cell proliferation and tumorigenesis. Life Sci, 86(19-20):716-721.
    41. Choi J, Nannenga B, Demidov ON, et al. Mice deficient for the wild-type p53-induced phosphatase gene (Wipl) exhibit defects in reproductive organs, immune function, and cell cycle control. Mol Cell Biol,2002,22(4):1094-1105.
    42. Momand J, Jung D, Wilczynski S, et al. The MDM2 gene amplification database. Nucleic Acids Res,1998,26(15):3453-3459.
    43. Shreeram S, Demidov ON, Hee WK, et al. Wipl phosphatase modulates ATM-dependent signaling pathways. Mol Cell,2006,23(5):757-764.
    44. Lu X, Bocangel D, Nannenga B, et al. The p53-induced oncogenic phosphatase PPM1D interacts with uracil DNA glycosylase and suppresses base excision repair. Mol Cell,2004,15(4):621-634.
    45. Moon SH, Lin L, Zhang X, et al. Wild-type p53-induced phosphatase 1 dephosphorylates histone variant gamma-H2AX and suppresses DNA double strand break repair. J Biol Chem,285(17):12935-12947.
    46. Macurek L, Lindqvist A, Voets O, et al. Wip1 phosphatase is associated with chromatin and dephosphorylates gammaH2AX to promote checkpoint inhibition. Oncogene,29(15):2281-2291.
    47. Zhang X, Lin L, Guo H, et al. Phosphorylation and degradation of MdmX is inhibited by Wipl phosphatase in the DNA damage response. Cancer Res,2009, 69(20):7960-7968.
    48. Lowe JM, Cha H, Yang Q, et al. Nuclear factor-kappaB (NF-kappaB) is a novel positive transcriptional regulator of the oncogenic Wipl phosphatase. J Biol Chem, 285(8):5249-5257.
    49. Chew J, Biswas S, Shreeram S, et al. WIP1 phosphatase is a negative regulator of NF-kappaB signalling. Nat Cell Biol,2009, 11(5):659-666.
    50. Kong W, Jiang X, Mercer WE. Downregulation of Wip-1 phosphatase expression in MCF-7 breast cancer cells enhances doxorubicin-induced apoptosis through p53-mediated trariscriptional activation of Bax. Cancer Biol Ther,2009,8(6).
    51. Parssinen J, Alarmo EL, Karhu R, et al. PPM1D silencing by RNA interference inhibits proliferation and induces apoptosis in breast cancer cell lines with wild-type p53. Cancer Genet Cytogenet,2008,182(1):33-39.
    52. Tan DS, Lambros MB, Rayter S, et al. PPM1D is a potential therapeutic target in ovarian clear cell carcinomas. Clin Cancer Res,2009,15(7):2269-2280.

© 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