用户名: 密码: 验证码:
GA6与SHP2蛋白在肝细胞癌中的临床意义及其作用机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
1、实验目的:研究GA6(Hematopoietic PBX-interacting protein)蛋白在肝细胞癌中表达的临床意义及其在肝细胞癌中的致癌作用的分子生物学机制。
     2、研究方法:纳入2000年至2007年在中国人民解放军总医院行手术治疗的肝细胞癌(HCC)标本328例(均包括肿瘤组织T及相应的癌旁非肿瘤组织NT)。构建组织芯片,通过免疫组化染色分析GA6的表达与临床特征的相关性及其与预后的关系;纳入新鲜HCC标本49例(均含肿瘤组织及对应癌旁非肿瘤组织)并通过Western blot检测GA6蛋白的表达;在肝癌细胞系中,通过GA6的过表达及干扰技术,进行生长曲线、平板克隆形成、软琼脂克隆形成、细胞周期测定及裸鼠移植瘤等实验,研究GA6蛋白对肝癌细胞生长、增殖能力的影响。
     3、研究结果:(1)免疫组化显示HCC肿瘤组织GA6蛋白的表达明显高于癌旁组织(T:1.39±0.63,NT:0.92±0.57,P<0.001):肿瘤组织阳性率94.5%(310/328),癌旁组织阳性率85.4%(280/328,P<0.001)。肿瘤组织中GA6的表达与HCC各临床指标之间均无相关性,生存分析示GA6的表达与HCC患者预后无关(P=0.687)。(2)在肝癌细胞系中通过GA6的过表达及干扰技术,生长曲线实验、克隆形成实验、裸鼠移植瘤实验均显示,GA6可促进肝癌细胞生长和增殖;细胞周期及相关蛋白的表达测定显示,GA6可影响细胞周期调节蛋白的表达,使大量细胞通过G2/M期检测点,进入M期分裂,从而促进肿瘤细胞增殖。
     4、结论:体内与体外研究显示,GA6可促进肝癌细胞的生长与增殖;免疫组化显示,HCC肿瘤组织中GA6蛋白的表达明显高于癌旁组织,但与患者预后无关。GA6在肝细胞癌中的作用仍有待进一步研究。
     1、研究背景:研究发现蛋白酪氨酸磷酸酶SHP2(Src homology2domaincontaining phosphotyrosine phosphatase2)在多种恶性肿瘤中发挥了癌基因的作用,但近年来对其抑癌作用也有报道,本研究室在前期研究中通过质谱分析发现SHP2与在肝细胞癌(HCC)的发病中起重要促癌作用的YAP2蛋白存在相互作用。本研究的目的是在大样本临床标本中检测SHP2表达的临床意义,并通过细胞培养及分子生物学技术研究SHP2与YAP2的相互作用。
     2、研究方法:纳入2000年至2007年在我院行手术治疗的肝细胞癌临床标本(每例均有肿瘤组织T及相应的癌旁非肿瘤组织NT)共333例,构建组织芯片,行免疫组化并进行生存分析与Cox多因素分析研究。收集31例新鲜临床HCC标本,行Western blot与qPCR以检测SHP2的表达。依据蛋白酪氨酸磷酸酶蛋白家族的作用机制,构建2种SHP2的底物捕获突变体,运用过表达、底物捕获Co-IP、双荧光素酶报告基因系统及酪氨酸磷酸化等多种技术研究在工具细胞系中SHP2对YAP2的调控作用。
     3、研究结果:在肝细胞癌中,相对于癌旁非肿瘤组织,肿瘤组织中SHP2蛋白及其mRNA的表达均明显降低,肿瘤组织SHP2的蛋白表达阳性率为66.1%,而癌旁非肿瘤组织为96.7%。生存分析示SHP2的低表达与HCC患者较短的生存期显著相关(P<0.001),SHP2的表达降低(ΔSHP2)是HCC患者预后差的一个独立危险因素(P=0.023,HR:0.628,95%CI:0.420~0.939)。通过细胞培养及分子生物学技术,我们发现SHP2蛋白与YAP2蛋白确实存在相互作用,如SHP2轻度上调内源性YAP2的表达、突变型SHP2-DM与YAP2的Co-IP及SHP2-DM对YAP2的酪氨酸磷酸化作用等,但研究发现,SHP2对YAP2的表达、活性及酸磷化水平的影响不显著。
     4、结论:尽管PTPN11一直被认为是一个原癌基因,但SHP2蛋白在肝细胞癌中发挥抑癌作用并可作为一个新的预后指标。SHP2蛋白与YAP蛋白存在较弱的相互作用,但这可能并不是其抑癌作用的主要机制。由于SHP2的对肿瘤的作用具有组织特异性,基于人类PTPN11基因的靶向治疗及SHP2在肝细胞癌中的作用仍有待进一步的研究。
Objective:This research was performed to study the clinical significance of theGA6protein expression in human hepatocellular carcinoma (HCC), and to explorethe mechanism of the tumorigenesis function of GA6in HCC.
     Methods:A retrospective cohort of328consecutive HCC specimens whounderwent surgical resection in the PLA general hospital from2000to2007wereincluded, both the tumor tissues (T) and self-matched adjacent non-tumor tissues(NT). By the construction of tissue microarray, immunohistochemical staining ofGA6protein was performed and the clinical and prognostic significance was explored.The GA6expression was also confirmed by the Western blot analysis of anothercohort of49fresh HCC specimens (both the T and self-matched adjacent NT tissues).By the GA6overexpression and GA6knockdown technique in HCC cell lines, weexplored the function of GA6protein in the growth and proliferation of HCC cells inthe experiments of growth curves assay, flat plate clone forming assay, soft agar cloneforming assay, cell cycle analysis and nude mice xenografts assay.
     Results:1. Immunohistochemical staining found that the GA6protein expressionwas significantly higher in HCC tumor tissues than the adjacent NT tissues (H-scoreT:1.39±0.63, NT:0.92±0.57,P<0.001;positive expression rate T:94.5%, NT:85.4%,P<0.001). The high expression of GA6in HCC tumor tissues was not related to theclinical features of HCC and was also not related to short overall survival byKaplan-Meier analysis (P=0.687).2. Growth curves assay, clone forming assay andnude mice xenografts assay demonstrated that GA6overexpression could promote thegrowth of HCC cells while GA6knockdown could inhibit the growth of HCC cells.Cell cycle and the correlated protein analysis revealed that GA6could promote thegrowth and proliferation of HCC cells by adjusting the expression of G2/M phasecorrelated protein.
     Conclusions:GA6protein can promote the growth and proliferation of HCCcells both in vitro and in vivo. By immunohistochemistry we found that theexpression of GA6in tumor tissues was significantly higher than that in NT tissues,however, no significant clinical significance was found for the higher GA6expression.Further researches are still needed to evaluate the function of GA6protein in HCC.
     Objective:Nonreceptor protein-tyrosine phosphatase of SHP2(Src homologyphosphotyrosine phosphatase2) has been previously well identified as aproto-oncogene in a variety of malignancies. However, recently the tumor suppressorfunction of SHP2has also been reported. In our previous research we found by themass chromatographic analysis that YAP2protein which had an importantproto-oncogene role in the pathogenesis of hepatocellular carcinoma (HCC) had aninteraction with the SHP2protein. For this reason, the present study was conducted toinvestigate the expression of SHP2and its associated clinical significance in a largecohort of HCC specimens and to explore the interaction of SHP2protein with YAP2protein by the technique of cell culture and molecular biology.
     Methods:HCC patients were retrieved from2000to2007in our hospital. Underthe inclusion/exclusion criteria, we included333HCC specimens and a tissuemicroarray of HCC tumor tissue and the self-matched adjacent non-tumor tissues wasconstructed. We also included31pairs of HCC fresh specimens. SHP2expressionwas determined by immunohistochemistry, Western blotting and quantitativepolymerase chain reaction. The association of SHP2expression with clinical featureswas analyzed, overall survival analysis and multivariate analysis were performed.Two plasmids with SHP2substrate trapping mutation were constructed, and theinteraction of SHP2with YAP2was investigated by the techniques of overexpression,substrate trapping co-immunoprecipitation, dual luciferase reporter gene assay andtyrosine phosphorylation.
     Results:Significantly decreased SHP2expression in tumor tissues compared withadjacent non-tumor tissues could be detected and the positive rate was66.1%and96.7%, respectively. Survival analysis showed low SHP2expression was significantlyassociated with short overall survival (P<0.001). Multivariate analysis showed thedecrease of SHP2expression (ΔSHP2) was an independent prognostic marker (P=0.023, HR:0.628,95%CI:0.420-0.939). By cell culture and the molecularbiology experiments, we found a weak interaction of SHP2with the YAP2protein,for example, SHP2slightly up-regulates the endogenous YAP2expression, SHP2-DM(double substrate trapping mutations) co-immunoprecipitates with YAP2andSHP2-DM up-regulates the tyrosine phosphorylation of YAP2. However, SHP2didnot have a predominant effect in the aspects of YAP2expression, YAP2downstreamactivity and YAP2tyrosine phosphorylation.
     Conclusion:Although human gene PTPN11was previously well interpreted as aproto-oncogene, SHP2protein is a tumor suppressor and a new prognostic marker inhepatocellular carcinoma. The weak interaction of SHP2protein with the YAP2protein may be not the major mechanism of its tumor suppressor function inhepatocellular carcinoma. For the oncogenic role of SHP2was tissue-specific, thetherapeutic target of PTPN11and its associated mechanism should be furtherresearched.
引文
[1] El-Serag H B, Rudolph K L.Hepatocellular carcinoma: epidemiology and molecularcarcinogenesis. Gastroenterology,2007,132(7):2557~76
    [2] Bruix J,Sherman M,Llovet J M,et al.Clinical management of hepatocellular carcinoma.Conclusions of the Barcelona-2000EASL conference. European Association for the Studyof the Liver. J Hepatol,2001,35(3):421~30
    [3] Parkin D M,Bray F,Ferlay J,et al.Global cancer statistics,2002. CA Cancer J Clin,2005,55(2):74~108
    [4] Schutte K,Bornschein J,Malfertheiner P.Hepatocellular carcinoma-epidemiological trendsand risk factors. Dig Dis,2009,27(2):80~92
    [5] McGlynn K A,Tsao L,Hsing A W,et al.International trends and patterns of primary livercancer. Int J Cancer,2001,94(2):290~6
    [6] Fields A C,Cotsonis G,Sexton D,et al.Survivin expression in hepatocellular carcinoma:correlation with proliferation, prognostic parameters, and outcome. Mod Pathol,2004,17(11):1378~85
    [7] Yagihashi A,Asanuma K,Kobayashi D,et al.Autoantibodies to survivin in patients withchronic hepatitis and hepatocellular carcinoma. Autoimmunity,2005,38(6):445~8
    [8] Ikai I,Arii S,Ichida T,et al.Report of the16th follow-up survey of primary liver cancer.Hepatol Res,2005,32(3):163~72
    [9] El-Serag H B,Mason A C.Rising incidence of hepatocellular carcinoma in the United States.N Engl J Med,1999,340(10):745~50
    [10] Chemin I,Zoulim F.Hepatitis B virus induced hepatocellular carcinoma. Cancer Lett,2009,286(1):52~9
    [11] Bouchard M J,Schneider R J.The enigmatic X gene of hepatitis B virus. J Virol,2004,78(23):12725~34
    [12] Fattovich G,Giustina G,Schalm S W,et al.Occurrence of hepatocellular carcinoma anddecompensation in western European patients with cirrhosis type B. The EUROHEP StudyGroup on Hepatitis B Virus and Cirrhosis. Hepatology,1995,21(1):77~82
    [13] Llovet J M,Burroughs A,Bruix J.Hepatocellular carcinoma. Lancet,2003,362(9399):1907~17
    [14] Himoto T, Kuriyama S, Zhang J Y, et al.Analyses of autoantibodies againsttumor-associated antigens in patients with hepatocellular carcinoma. Int J Oncol,2005,27(4):1079~85
    [15] Ye C P,Qiu C Z,Huang Z X,et al.Relationship between survivin expression and recurrence,and prognosis in hepatocellular carcinoma. World J Gastroenterol,2007,13(46):6264~8
    [16]宋扬冯,张惠中等.原发性肝细胞癌中雌激素受体α与β的表达及临床意义.陕西医学杂志,2005,34(2):153~155
    [17]余艳军覃.肿瘤.性激素与肝细胞癌关系的研究进展,2005,2(3):291~293
    [18]周蕾.酵母双杂文技术筛选与ER相互作用蛋白质及其功能研究:[硕士学位论文].北京:军事医学科学院附属医院,2005
    [19] Nilsson S,Gustafsson J A.Estrogen receptor transcription and transactivation:Basic aspectsof estrogen action. Breast Cancer Res,2000,2(5):360~6
    [20] Klinge C M.Estrogen receptor interaction with estrogen response elements. Nucleic AcidsRes,2001,29(14):2905~19
    [21] Tsai M J, O'Malley B W.Molecular mechanisms of action of steroid/thyroid receptorsuperfamily members. Annu Rev Biochem,1994,63:451~86
    [22] Mangelsdorf D J,Thummel C,Beato M,et al.The nuclear receptor superfamily:the seconddecade. Cell,1995,83(6):835~9
    [23] Katzenellenbogen B S,Sun J,Harrington W R,et al.Structure-function relationships inestrogen receptors and the characterization of novel selective estrogen receptor modulatorswith unique pharmacological profiles. Ann N Y Acad Sci,2001,949:6~15
    [24] Aranda A,Pascual A.Nuclear hormone receptors and gene expression. Physiol Rev,2001,81(3):1269~304
    [25] Zhou D, Chen S.PNRC2is a16kDa coactivator that interacts with nuclear receptorsthrough an SH3-binding motif. Nucleic Acids Res,2001,29(19):3939~48
    [26] Abramovich C,Shen W F,Pineault N,et al.Functional cloning and characterization of anovel nonhomeodomain protein that inhibits the binding of PBX1-HOX complexes to DNA.J Biol Chem,2000,275(34):26172~7
    [27] Abramovich C,Chavez E A,Lansdorp P M,et al.Functional characterization of multipledomains involved in the subcellular localization of the hematopoietic Pbx interactingprotein (HPIP). Oncogene,2002,21(44):6766~71
    [28] DiMartino J F,Selleri L,Traver D,et al.The Hox cofactor and proto-oncogene Pbx1isrequired for maintenance of definitive hematopoiesis in the fetal liver. Blood,2001,98(3):618~26
    [29] Mahajan S S,Little M M,Vazquez R,et al.Interaction of HCF-1with a cellular nuclearexport factor. J Biol Chem,2002,277(46):44292~9
    [30] Manavathi B,Acconcia F,Rayala S K,et al.An inherent role of microtubule network in theaction of nuclear receptor. Proc Natl Acad Sci U S A,2006,103(43):15981~6
    [31] Boonyaratanakornkit V.Scaffolding proteins mediating membrane-initiated extra-nuclearactions of estrogen receptor. Steroids,76(9):877~84
    [32] Han J,Ding L,Yuan B,et al.Hepatitis B virus X protein and the estrogen receptor variantlacking exon5inhibit estrogen receptor signaling in hepatoma cells. Nucleic Acids Res,2006,34(10):3095~106
    [33] Zhang H,Xie X,Zhu X,et al.Stimulatory cross-talk between NFAT3and estrogen receptorin breast cancer cells. J Biol Chem,2005,280(52):43188~97
    [34] Ding L,Niu C,Zheng Y,et al.FHL1interacts with oestrogen receptors and regulates breastcancer cell growth. J Cell Mol Med,15(1):72~85
    [35] Wang X,Yang Z,Zhang H,et al.The estrogen receptor-interacting protein HPIP increasesestrogen-responsive gene expression through activation of MAPK and AKT. BiochimBiophys Acta,2008,1783(6):1220~8
    [36] Walton S P,Wu M,Gredell J A,et al.Designing highly active siRNAs for therapeuticapplications. FEBS J,277(23):4806~13
    [37]徐小洁范,韩永健等.慢病毒介导RNA干扰HPIP蛋白抑制肿瘤细胞增殖.生物技术通讯,2011,3(22):321~324
    [38] Hamilton SR A L.WHO消化系统肿瘤病理学和遗传学.虞积耀主译.人民卫生出版社,2006,第一版:193
    [39] SS S.诊断外科病理学.回允中主译.北京大学医学出版社,2003,第3版:1567~1592
    [40]邰艳红.Gankyrin在慢性肝炎、肝硬化及肝细胞癌中的表达及临床病理学意义:[博士学位论文].北京:中国人民解放军总医院,2010
    [41] Kononen J,Bubendorf L,Kallioniemi A,et al.Tissue microarrays for high-throughputmolecular profiling of tumor specimens. Nat Med,1998,4(7):844~7
    [42] Torhorst J,Bucher C,Kononen J,et al.Tissue microarrays for rapid linking of molecularchanges to clinical endpoints. Am J Pathol,2001,159(6):2249~56
    [43] Hsu F D,Nielsen T O,Alkushi A,et al.Tissue microarrays are an effective quality assurancetool for diagnostic immunohistochemistry. Mod Pathol,2002,15(12):1374~80
    [44] McClelland R A, Finlay P, Walker K J, et al.Automated quantitation ofimmunocytochemically localized estrogen receptors in human breast cancer. Cancer Res,1990,50(12):3545~50
    [45] Finn R S, Press M F, Dering J, et al.Estrogen receptor, progesterone receptor, humanepidermal growth factor receptor2(HER2), and epidermal growth factor receptorexpression and benefit from lapatinib in a randomized trial of paclitaxel with lapatinib orplacebo as first-line treatment in HER2-negative or unknown metastatic breast cancer. JClin Oncol,2009,27(24):3908~15
    [46] El-Serag H B,Davila J A,Petersen N J,et al.The continuing increase in the incidence ofhepatocellular carcinoma in the United States:an update. Ann Intern Med,2003,139(10):817~23
    [47] Vizoso F J, Rodriguez M, Altadill A, et al.Liver expression of steroid hormones andApolipoprotein D receptors in hepatocellular carcinoma. World J Gastroenterol,2007,13(23):3221~7
    [48] Zhang P J, Zhao J, Li H Y, et al.CUE domain containing2regulates degradation ofprogesterone receptor by ubiquitin-proteasome. EMBO J,2007,26(7):1831~42
    [49] Pan X,Zhou T,Tai Y H,et al.Elevated expression of CUEDC2protein confers endocrineresistance in breast cancer. Nat Med,17(6):708~14
    [50] Singhal S,Vachani A,Antin-Ozerkis D,et al.Prognostic implications of cell cycle, apoptosis,and angiogenesis biomarkers in non-small cell lung cancer:a review. Clin Cancer Res,2005,11(11):3974~86
    [51] Volm M,Koomagi R,Mattern J,et al.Cyclin A is associated with an unfavourable outcomein patients with non-small-cell lung carcinomas. Br J Cancer,1997,75(12):1774~8
    [52]沈俊.细胞周期素依赖激酶抑制蛋白p27与泌尿系统肿瘤.重庆医科大学学报,2003,28:394~397
    [53] Bard-Chapeau E A, Li S, Ding J, et al.Ptpn11/SHP2acts as a tumor suppressor inhepatocellular carcinogenesis. Cancer Cell,2011,19(5):629~39
    [54] Xu M Z,Yao T J,Lee N P,et al.Yes-associated protein is an independent prognostic markerin hepatocellular carcinoma. Cancer,2009,115(19):4576~85
    [55] Kim J S,Shin O R,Kim H K,et al.Overexpression of protein phosphatase non-receptor type11(PTPN11)in gastric carcinomas. Dig Dis Sci,2010,55(6):1565~9
    [56]赵硕.癌症组织中SHP2在mRNA水平上的表达:硕士学位论文.吉林大学,2007
    [57]吴建富.底物捕获法检测SHP1在MAPK信号通路中的靶蛋白:硕士学位论文.扬州大学,2008
    [58]王磊.SHP2蛋白在乳腺癌中的表达及临床意义:硕士学位论文.兰州大学,2010
    [59] Chan R J,Feng G S.PTPN11is the first identified proto-oncogene that encodes a tyrosinephosphatase. Blood,2007,109(3):862~7
    [60] Keilhack H,David F S,McGregor M,et al.Diverse biochemical properties of SHP2mutants.Implications for disease phenotypes. J Biol Chem,2005,280(35):30984~93
    [61] Chan G,Kalaitzidis D,Neel B G.The tyrosine phosphatase SHP2(PTPN11)in cancer.Cancer Metastasis Rev,2008,27(2):179~92
    [62] Ostman A,Hellberg C,Bohmer F D.Protein-tyrosine phosphatases and cancer. Nat RevCancer,2006,6(4):307~20
    [63] Zhou X D,Agazie Y M.Inhibition of SHP2leads to mesenchymal to epithelial transition inbreast cancer cells. Cell Death Differ,2008,15(6):988~96
    [64] Matozaki T,Murata Y,Saito Y,et al.Protein tyrosine phosphatase SHP2:a proto-oncogeneproduct that promotes Ras activation. Cancer Sci,2009,100(10):1786~93
    [65] Grinnell K L,Casserly B,Harrington E O.Role of protein tyrosine phosphatase SHP2inbarrier function of pulmonary endothelium. Am J Physiol Lung Cell Mol Physiol,2010,298(3):L361~70
    [66] Yu S J,Yu J K,Ge W T,et al.SPARCL1, SHP2, MSH2, E-cadherin, p53, ADCY-2andMAPK are prognosis-related in colorectal cancer. World J Gastroenterol,2011,17(15):2028~36
    [67]程玉生,沈华浩.蛋白酪氨酸磷酸酶SHP2与呼吸系统疾病.生理科学进展,2011,42(5):379~382
    [68]王钊张,侯媛媛,徐长庆,田野,张力.SHP2对人胃癌细胞克隆增殖的影响.中国药理学通报,2012,28(1):106~109
    [69] Crovello C S,Lai C,Cantley L C,et al.Differential signaling by the epidermal growthfactor-like growth factors neuregulin-1and neuregulin-2. J Biol Chem,1998,273(41):26954~61
    [70]张梅.SHP2酪氨酸磷酸酶在DNA损伤性放、化疗引发的骨髓细胞毒中的调控作用及机制初:硕士学位论文.安徽医科大学,2008
    [71] Kontaridis M I,Eminaga S,Fornaro M,et al.SHP2positively regulates myogenesis bycoupling to the Rho GTPase signaling pathway. Mol Cell Biol,2004,24(12):5340~52
    [72] Mohi M G,Neel B G.The role of SHP2(PTPN11)in cancer. Curr Opin Genet Dev,2007,17(1):23~30
    [73] Sturla L M, Zinn P O, Ng K, et al.Src homology domain-containing phosphatase2suppresses cellular senescence in glioblastoma. Br J Cancer,105(8):1235~43
    [74] Flint A J, Tiganis T, Barford D, et al.Development of "substrate-trapping" mutants toidentify physiological substrates of protein tyrosine phosphatases. Proc Natl Acad Sci U SA,1997,94(5):1680~5
    [75] Bennett A M,Hausdorff S F,O'Reilly A M,et al.Multiple requirements for SHPTP2inepidermal growth factor-mediated cell cycle progression. Mol Cell Biol,1996,16(3):1189~202
    [76] Agazie Y M,Hayman M J.Development of an efficient "substrate-trapping" mutant of Srchomology phosphotyrosine phosphatase2and identification of the epidermal growth factorreceptor, Gab1, and three other proteins as target substrates. J Biol Chem,2003,278(16):13952~8
    [77] Sudol M.Yes-associated protein (YAP65)is a proline-rich phosphoprotein that binds to theSH3domain of the Yes proto-oncogene product. Oncogene,1994,9(8):2145~52
    [78] Sudol M, Bork P, Einbond A, et al.Characterization of the mammalian YAP(Yes-associated protein)gene and its role in defining a novel protein module, the WWdomain. J Biol Chem,1995,270(24):14733~41
    [79]达春丽.胃癌及其癌前病变组织中Yes-associated protein和Survivin的表达及意义:[硕士学位论文].沈阳:中国医科大学,2009
    [80] Huang J,Wu S,Barrera J,et al.The Hippo signaling pathway coordinately regulates cellproliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP. Cell,2005,122(3):421~34
    [81] Liu A M,Xu M Z,Chen J,et al.Targeting YAP and Hippo signaling pathway in liver cancer.Expert Opin Ther Targets,14(8):855~68
    [82] Harvey K F,Pfleger C M,Hariharan I K.The Drosophila Mst ortholog, hippo, restrictsgrowth and cell proliferation and promotes apoptosis. Cell,2003,114(4):457~67
    [83] Pantalacci S, Tapon N, Leopold P.The Salvador partner Hippo promotes apoptosis andcell-cycle exit in Drosophila. Nat Cell Biol,2003,5(10):921~7
    [84] Udan R S,Kango-Singh M,Nolo R,et al.Hippo promotes proliferation arrest and apoptosisin the Salvador/Warts pathway. Nat Cell Biol,2003,5(10):914~20
    [85] Wu S,Huang J,Dong J,et al.hippo encodes a Ste-20family protein kinase that restricts cellproliferation and promotes apoptosis in conjunction with salvador and warts. Cell,2003,114(4):445~56
    [86] Lai Z C,Wei X,Shimizu T,et al.Control of cell proliferation and apoptosis by mob astumor suppressor, mats. Cell,2005,120(5):675~85
    [87] Dong J,Feldmann G,Huang J,et al.Elucidation of a universal size-control mechanism inDrosophila and mammals. Cell,2007,130(6):1120~33
    [88] Oh H,Irvine K D.In vivo analysis of Yorkie phosphorylation sites. Oncogene,2009,28(17):1916~27
    [89] Wu S, Liu Y, Zheng Y, et al.The TEAD/TEF family protein Scalloped mediatestranscriptional output of the Hippo growth-regulatory pathway. Dev Cell,2008,14(3):388~98
    [90] Peng H W, Slattery M, Mann R S.Transcription factor choice in the Hippo signalingpathway: homothorax and yorkie regulation of the microRNA bantam in the progenitordomain of the Drosophila eye imaginal disc. Genes Dev,2009,23(19):2307~19
    [91] Nolo R, Morrison C M, Tao C, et al.The bantam microRNA is a target of the hippotumor-suppressor pathway. Curr Biol,2006,16(19):1895~904
    [92] Hamaratoglu F, Willecke M, Kango-Singh M, et al.The tumour-suppressor genesNF2/Merlin and Expanded act through Hippo signalling to regulate cell proliferation andapoptosis. Nat Cell Biol,2006,8(1):27~36
    [93] Bennett F C, Harvey K F.Fat cadherin modulates organ size in Drosophila via theSalvador/Warts/Hippo signaling pathway. Curr Biol,2006,16(21):2101~10
    [94] Willecke M,Hamaratoglu F,Kango-Singh M,et al.The fat cadherin acts through the hippotumor-suppressor pathway to regulate tissue size. Curr Biol,2006,16(21):2090~100
    [95] Cho E, Irvine K D.Action of fat, four-jointed, dachsous and dachs in distal-to-proximalwing signaling. Development,2004,131(18):4489~500
    [96] Zhao B,Wei X,Li W,et al.Inactivation of YAP oncoprotein by the Hippo pathway isinvolved in cell contact inhibition and tissue growth control. Genes Dev,2007,21(21):2747~61
    [97] Callus B A,Verhagen A M,Vaux D L.Association of mammalian sterile twenty kinases,Mst1and Mst2, with hSalvador via C-terminal coiled-coil domains, leads to its stabilizationand phosphorylation. FEBS J,2006,273(18):4264~76
    [98] Chan E H,Nousiainen M,Chalamalasetty R B,et al.The Ste20-like kinase Mst2activatesthe human large tumor suppressor kinase Lats1. Oncogene,2005,24(12):2076~86
    [99] Praskova M,Xia F,Avruch J.MOBKL1A/MOBKL1B phosphorylation by MST1and MST2inhibits cell proliferation. Curr Biol,2008,18(5):311~21
    [100] Overholtzer M,Zhang J,Smolen G A,et al.Transforming properties of YAP, a candidateoncogene on the chromosome11q22amplicon. Proc Natl Acad Sci U S A,2006,103(33):12405~10
    [101] Lam-Himlin D M,Daniels J A,Gayyed M F,et al.The hippo pathway in human uppergastrointestinal dysplasia and carcinoma:a novel oncogenic pathway. Int J GastrointestCancer,2006,37(4):103~9
    [102] Steinhardt A A,Gayyed M F,Klein A P,et al.Expression of Yes-associated protein incommon solid tumors. Hum Pathol,2008,39(11):1582~9
    [103] Zender L,Spector M S,Xue W,et al.Identification and validation of oncogenes in livercancer using an integrative oncogenomic approach. Cell,2006,125(7):1253~67
    [104] Fleming I, Cooper J, Henson D.AJCC cancer staging manual.5th ed. Philadelphia:Lippincott-Raven,1997,98~126
    [105] Li H Y,Liu H,Wang C H,et al.Deactivation of the kinase IKK by CUEDC2throughrecruitment of the phosphatase PP1. Nat Immunol,2008,9(5):533~41
    [106] Miyamoto D, Miyamoto M, Takahashi A, et al.Isolation of a distinct class ofgain-of-function SHP2mutants with oncogenic RAS-like transforming activity from solidtumors. Oncogene,2008,27(25):3508~15
    [107] Fridberg M, Kjellstrom S, Anagnostaki L, et al.Immunohistochemical analyses ofphosphatases in childhood B-cell lymphoma:lower expression of PTEN and HePTP andhigher number of positive cells for nuclear SHP2in B-cell lymphoma cases compared tocontrols. Pediatr Hematol Oncol,2008,25(6):528~40

© 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