SIPA1通过下调Rap1-ERK通路抑制HCC的复发转移
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肝细胞癌(Hepatocellular carcinoma, HCC)是当前严重威胁我国人民生命健康的最主要恶性肿瘤之一。随着肝脏外科技术以及麻醉与围手术期处理水平的进步,肝癌的手术切除率显著提高,在一些大的肝脏外科中心肝癌手术死亡率已接近为零。尽管如此,以5年生存率为标志的肝癌整体治疗水平仍无显著改善,究其原因在于肝癌术后的复发转移率居高不下。据统计,肝癌切除术后5年的复发率高于60%,小肝癌也在40%以上。显然,肝癌的复发、转移已成为严重制约肝癌患者长期存活的关键因素。因此,深入研究肝癌复发转移机制,积极探索有效的抗复发治疗措施是当前肝癌诊治中的重点和难点,对于进一步改善肝癌患者长期存活率具有重要意义。
     已有的研究证实,SIPA1 (Signal-induced Proliferation-associated Gene 1,信号诱导的增殖相关基因1)在血液系统肿瘤的发生发展过程中发挥了非常重要的作用。新近的研究表明小鼠sipa1是决定小鼠乳腺癌转移倾向的重要基因。众所周知,肝脏是胚胎时期的重要造血器官,血细胞(如血小板)与肿瘤的转移密切相关。根据以上的研究结果我们推测SIPA1基因很可能在HCC复发转移中发挥重要作用。因此,本课题在研究SIPA1在临床肝癌病例及肝癌细胞系中表达情况的基础上,进一步通过质粒构建转染技术上调SIPA1的表达,并综合利用一系列体外、体内方法研究SIPA1调控HCC侵袭转移的分子机制。我们获得了如下研究结果:
     1.我们首先采用RT-PCR和Western blot等方法检测了SIPA1 mRNA和蛋白在30例新鲜HCC组织及相应邻近非癌肝组织(ANLT)中的表达水平,结果发现与ANLT比较,SIPA1 mRNA和蛋白水平在癌组织中表达均显著下调(P<0.05),且肝癌组织中SIPA1 mRNA和蛋白表达水平呈明显正相关(r=0.757,P<0.001)。进一步采用实时定量PCR检测显示ANLT中SIPA1 mRNA表达量为癌组织的4.17倍。进而,我们采用免疫组化技术检测了130例HCC组织中SIPA1的表达,结果显示SIPA1主要分布于细胞浆内,其在癌组织的阳性表达率为62.3%(81/130)。肝癌组织中SIPA1低表达与肿瘤多结节,分化差及静脉侵袭密切相关(P<0.05)。SIPA1阴性表达组HCC患者的总体生存时间显著短于SIPA1阳性表达组的患者(41.0±6.6个月vs.66.0±5.2个月,P=0.003),SIPA1阴性表达组的无瘤生存时间亦显著短于SIPA1阳性表达组(35.0±6.1个月vs.61.1±5.4个月,P=0.002)。多元Cox回归模型显示SIPA1阴性表达是HCC预后的独立危险因素。
     2. RT-PCR及Western blot方法检测SIPA1 mRNA和蛋白在HepG2、MHCC97-L和HCCLM3这3种侵袭转移潜能依次升高的肝癌细胞系中的表达水平,并以正常肝细胞系L02、常氏肝细胞系CCL13作为对照。结果显示SIPA1 mRNA和蛋白在HepG2、HCCLM3和MHCC97-L肝癌细胞系中的表达水平显著低于CCL13和L02正常肝细胞系(P<0.05),且其表达水平在HepG2. MHCC97-L、HCCLM3细胞中依次降低(P<0.05),提示SIPA1表达水平与肝癌细胞的侵袭转移潜能呈负相关。同时确定以SIPA1表达水平最低的HCCLM3为过表达质粒的靶细胞。
     3.我们构建了重组质粒pcDNA3.1-SIPA1,使用Lipofectamine 2000转染并行G418筛选,经RT-PCR和Western blot等方法验证SIPA1的表达,最终获得了稳定转染细胞系HCCLM3SIPA1+和空质粒对照组HCCLM3vector细胞。采用MTT法、粘附实验、划痕愈合实验和Transwell等实验方法分别检测两组细胞在细胞增殖与粘附能力、迁移运动能力和侵袭能力上的差别。MTT研究结果显示,HCCLM3SIPA1+的细胞增殖能力较HCCLM3vector显著下降(P<0.05);粘附实验结果显示与HCCLM3vector细胞相比,HCCLM3SIPA1+细胞与FN的粘附能力明显减弱(P<0.05);划痕愈合实验和Transwell侵袭实验显示HCCLM3SIPA1+与HCCLM3vector相比,细胞的迁移运动能力和侵袭能力均显著下降(P<0.05)。以上体外实验研究结果证实上调SIPA1的表达可显著抑制HCC细胞的增殖、粘附、迁移和侵袭能力。进一步采用免疫荧光技术检测HCCLM3SIPA1+细胞和HCCLM3vector细胞的骨架蛋白F-actin的形成情况,发现SIPA1能明显减少F-actin的形成。
     4.为进一步体内观察上调SIPA1对肝癌侵袭转移的影响,我们建立了裸鼠原位肝癌肺转移模型。结果显示,HCCLM3SIPA1+细胞在肝脏中成瘤体积明显较HCCLM3vector细胞小,分别为1.57±0.08cm3和6.28±0.13cm3,(P<0.05)。通过裸鼠肺组织连续切片观察肺转移灶的发生率,发现与HCCLM3vector组相比,HCCLM3SIPA1+组的肺转移率显著下降(16.7%vs.83.3%,P=0.021)。由此证明,上调SIPA1可显著抑制肝癌细胞的体内成瘤能力和侵袭转移能力。
     5.综合已有的研究,我们推测SIPA1可能通过Rapl-ERK信号传导通路调控肝癌细胞的侵袭转移。为证实上述假说,我们首先检测了HCCLM3SIPA1+细胞和HCCLM3Vector细胞中Rap1的活性,研究发现与后者相比,过表达SIPA1的HCCLM3SIPA1+细胞中,Rap1的活性显著降低(P<0.05);免疫共沉淀技术亦证实HCCLM3SIPA1+细胞中SIPA1与Rap1存在直接的相互作用。进而,我们采用Rap1激活因子8CPT-2Me-cAMP(终浓度100μM),恢复HCCLM3SIPA1+细胞中Rap1的活性,反向验证Rap1在SIPA1调控肝癌侵袭转移中的作用。结果发现,恢复Rap1的活性后,细胞的迁移运动能力和和侵袭能力得到显著恢复(P<0.05)且细胞骨架蛋白F-actin表达亦得到明显恢复。Western blot证实HCCLM3SIPA1+细胞中Rap1下游基因ERK的磷酸化水平显著下降(P<0.05),恢复Rap1活性后p-ERK的表达得到恢复。上述研究结果证实,SIPA1可通过下调Rap1-ERK信号传导通路抑制肝癌细胞的侵袭转移。
     综上所述,我们首次发现SIPA1在肝细胞癌组织中的表达显著下调,其表达水平与肝癌分化差、多结节、静脉侵犯等临床病理特征密切相关,同时与肝细胞癌预后差和术后早期发生复发转移等密切相关。SIPA1是肝细胞癌预后的独立危险因素,可作为预测肝细胞癌术后生存和复发转移的分子标志物。体内、体外研究证明,SIPA1通过下调Rap1-ERK信号通路抑制肝细胞癌的增殖和侵袭转移能力。
Hepatocellular carcinoma (HCC) is a main malignancy that threaten our people now. With the improvement of liver surgical technique and management level of perioperation, the resection rate of HCC is increasing and the operation mortality rate is approaching zero at large liver surgery centers. But the overall mortality of HCC patients remains high and the 5-year survial rate of patients is still low. The postoperation recurrence and metastasis of HCC is the main cause of high overall mortality. The 5-year recurrence rate of HCC after operation is more than 60%, even the small HCC the 5-year recurrence rate is above 40%. Obviously the recurrence and metastasis after HCC resection is the bottleneck problem that severely restricts the improvement of long survival rate of HCC patients. So deep study of the mechanism of recurrence and metastasis of HCC and explore the methods of anti-metastasis are focus of HCC research, which are very meaningful for improvement of long survival rate of HCC patients.
     Signal-induced Proliferation-associated Gene 1 (SIPA1) was found play an important role in the blood system tumors in the past decade. Recent research found that mouse sipal gene was an important gene that control the metastasis of mouse breast cancer. It is well-known that the liver is a hematogenic organ in embryonic period and blood cells (for example platelet) had close relationship with tumor metastasis. These facts urge us to explore the role of SIPA1 gene in the HCC which had high potential of metastasis. Up to now there is no report about this both at home and abroad. So in this study we first examine the expression of SIPA1 gene in HCC specimens and HCC cell lines, then overexpress the gene by cotransfection the recombinant plasmid (pcDNA3.1-SIPA1) with HCC cell line. Molecullar mechenism of the regulation of SIPA1 to HCC metastasis was studied by in vitro and in vivo. The results as follows was observed:
     1. The SIPA1 mRNA and protein in 30 fresh HCC specimens, which consisted of cancer tissue and corresponding ANLT(adjacent non-tumor liver tissue), was detected by Reverse Transcription PCR(Polymerase chain reaction) and Western blot.The expression of mRNA and protein of SIPA1 in fresh HCC tissues was lower than in ANLTs significantly(P<0.05). The expression of SIPA1 mRNA was positively correlated with SIPA1 protein(r=0.757, P<0.001). The real time PCR was used to detect the relative accurate exprssion of SIPA1 mRNA, and the expression of SIPA1 mRNA in ANLT was found to be 4.17 times of that in cancer tissue. The expression of SIPA1 in 130 paraffin HCC specimens was dectected by immunohistochemistry, the positive expression rate of SIPA1 protein in cancer tissue was 62.3% (81/130). Low expression of SIPA1 in cancer tissue of HCC had close relationship with multiple tumor nodules, poor differentiation and vein invasion (P<0.05). Patients with negative expression of SIPA1 in cancer tissue had a shorter overall survial (OS) than the positive group (41.0±6.6 months vs. 66.0±5.2 months, P=0.003), the disease-free survial (DFS) of negative group of SIPA1 was shorter than the positive group(35.0±6.1 months vs.61.1±5.4 months, P=0.002). Multiple cox regression showed negative expression of SIPA1 in cancer tissue was a independent risk factor for HCC patients. Thus results suggested that negative expression of SIPA1 gene had relationship with tumor invasion and metastasis.
     2. Through reverse transcriptase-PCR and Western blot the expression of SIPA1 mRNA and protein was detected in L02 cell line, CCL13 cell line and three HCC cell lines (HepG2, MHCC97-L, HCCLM3), their invasion ability increases in order. The expression of SIPA1 mRNA and protein in HCCLM3 was found to be lowest among the five cell lines (P<0.05).Thus results indicated that low expression of SIPA1 gene had relationship with invasion of HCC cells. In addition, HCCLM3 cell line was chosen as the target cell for overexpression of SIPA1.
     3. To analyze the clinical findings, overexpression SIPA1 plasmid was constructed with gene cloning and recombinant DNA technique. The HCCLM3SIPA1+cell line overexpressing SIPA1 stably was established successfully through Lipofectamine 2000 transfection and G418 screening. Through reverse transcriptase PCR and Western blot, the SIPA1 mRNA and protein was proved expressing higher in HCCLM3SIPA1+cell line than in HCCLM3vector(P< 0.05). MTT [3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay was used to detect the proliferation and adhesion ability of HCCLM3SIPA1+cells, and overexpression of SIPA1 gene was found to inhibit the proliferation ability and adhesion ability to fibronectin of HCC cells (P<0.05). The abilities of Wound-healing, Transwell invasion, formation of cytoskeleton of HCCLM3SIPA1+cell line were all weaker than that of HCCLM3vector cell line (P<0.05). Thus results all showed that overexpression of SIPA1 gene could inhibit the proliferaton, adhesion, motility and invasion ability of HCC cells in vitro.
     4. We further examined the in vivo relevance of the potential role for SIPA1 in HCC tumorigenesis and metastasis by using a orthotopic nude mice implantation liver cancer model. The tumors of mouse livers were measured, we found that the average volume of primary tumors in HCCLM3SIPA1+group was dramatically smaller than that of HCCLM3vector group (1.57±0.08cm3 vs.6.28±0.13cm3), which suggested that up-regulation of SIPA1 in vivo could inhibit growth of HCC. The expression of SIPA1 in liver tumors was determined by immunohistochemistry to ensure the difference of SIPA1 expression in vivo. The pulmonary metastasis was observed in the lung tissue sections, and the ratio of pulmonary metastasis in HCCLM3SIPA1+cell group was found significantly less than in HCCLM3vector cell group (16.7% vs.83.3%, P=0.021). Together, these data supported that SIPA1 played an important role in inhibiting HCC tumorigenesis and metastasis in vivo.
     5. Many researches have proved that SIPA1 gene play an import role in bood system tumors through regulating the Rap1 small G protein in hematopoietic progenitor cells. So we speculate that SIPA1 may regulate the Rap1 protein in HCC cells. Pull-down assay showed the level of Rap1-GTP was lower in HCCLM3SIPA1+cells than in HCCLM3vector cells (P< 0.05). Next, co-immunoprecipitations confirmed the direct interaction of SIPA1 and Rap1 proteins in HCC cells. When HCCLM3SIPA1+cells were exposed in chemical factor 8CPT-2Me-cAMP (final concentration 100μM), the Rap1 activity recovered, and the motility, invasion ability and the formation of F-actin cytoskeletal protein of HCCLM3SIPA1+cells also restored. Western blot showed the expression of p-ERK decreased when the SIPA1 was up-regulated (P<0.05), and the expression of p-ERK restored when Rap1 activity was recoverd by adding chemical factor 8CPT-2Me-cAMP(final concentration 100μM) to the HCCLM3SIPA1+ cells.Thus results indicated that SIPA1 inhibited the invasion and metastasis of HCC cells probably through down-regulating the activity of Rap 1-ERK pathway.
     Conclusions:SIPA1 is firstly found significantly down-regulated in HCC tissue. Low expression of SIPA1 in HCC is related with multiple tumor nodules, poor differentiation, vein invasion and early recurrence and metastasis. SIPA1 is an independent risk factor for the prognosis of HCC and may became a molecular marker of survival and recurrence after operaton. In vitro and in vivo exprements prove that SIPA1 plays an important role in inhibiting the proliferation ability, invasion and metastasis of HCC cells through down-regulating Rap1-ERK pathway.
引文
[1]El-Serag HB, Rudolph KL. Hepatocellular carcinoma:epidemiology and molecular carcinogenesis.Gastroenterology,2007,132(7):2557-2576
    [2]Parkin DM, Bray F, Ferlay J, et al.Global cancer statistics,2002. CA Cancer J Clin,2005,55(2):74-108
    [3]张思维,陈万青,孔灵芝,等.中国部分市县2003年恶性肿瘤发病年度报告.中国肿瘤,2007,16(7):494-507
    [4]吴孟超.肝癌外科综合治疗的现状和前景.中华肝胆外科杂志,2006,12(1):1-4
    [5]杨连粤,黄耿文,黄建华,等.不阻断肝门的大肝癌切除术.中华肝胆外科杂,2003,9(6):331-333
    [6]Johnson P J. Hepatocellular carcinoma:is current therapy really altering outcome? Gut,2002,51(4):459-462
    [7]欧迪鹏,杨连粤,黄耿文,等.肝细胞癌术后复发转移相关危险因素分析,中南大学学报(医学版),2005;30(5):540-543
    [8]Tang ZY. Hepatocellular carcinoma-Cause, treatment and metastasis. World J Gastroenterol,2001,7(4):445-454
    [9]周信达.肝癌转移复发的研究进展与展望。中华肝脏病杂志,2004,12(11):690
    [10]王杰军,高勇,许青.肿瘤转移机制及诊疗进展.上海:第二军医大学出版社,2002.76-77
    [11]Wang W, Yang LY, Huang GW, et al.Genomic analysis reveals RhoC as a potential marker in hepatocellular carcinoma with poor prognosis. Br J Cancer. 2004,90(12):2349-2355
    [12]刘合利,杨连粤,黄耿文,等.整合素av亚基对肝细胞性肝癌血管生成、侵袭转移的影响.中华普通外科杂志,2002,17(9):542-543
    [13]薛开先.肝癌发生的分子遗传学和表遗传学研究.癌症,2005,24(6):757-768
    [14]Dong JT, Lamb PW, Rinker-Schaeffer CW, et al.KAI1, a metastasis suppressor gene for prostate cancer on human chromosome 11p11.2. Science,1995,268 (5212):884-886
    [15]Steeg PS, Bevilacqua G, Sobel ME, Liotta LA. Identification and characterization of differentially expressed genes in tumor metastasis:the nm23 gene. Basic Life Sci,1991,57:355-360
    [16]Tuck AB, Wilson SM, Chambers AF. ras transfection and expression does not induce progression from tumorigenicity to metastatic ability in mouse LTA cells. Clin Exp Metastasis,1990,8(5):417-431
    [17]Guy CT, Cardiff RD, Muller WJ.Induction of mammary tumors by expression of polyomavirus middle T oncogene:a transgenic mouse model for metastatic disease. Mol Cell Biol,1992,12(3):954-961
    [18]Park YG, Zhao X, Lesueur F, et al. Sipal is a candidate for underlying the metastasis efficiency modifier locus Mtesl.Nat Genet,2005,37(10):1026-1027
    [19]Crawford NP, Ziogas A, Peel DJ, et al. Germline polymorphisms in SIPA1 are associated with metastasis and other indicators of poor prognosis in breast cancer. Breast Cancer Res,2006,8(2):R16
    [20]Edmondson HA, Steiner PE. Primary carcinoma of the liver:a study of 100 cases among 48,900 necropsies. Cancer,1954,7:462-504
    [21]Livak KJ and Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods,2001, 25 (4):402-408
    [22]Shimizu M, Saitoh Y, Itoh H. Immunohistochemical staining of Ha-ras oncogene product in normal, benign, and malignant human pancreatic tissues.Hum Pathol, 1990,21(6):607-12
    [23]郝新保,张利朝,殷缨,等.MTT比色法测定细胞生长曲线.第四军医大学学报,1997,18(4):390-391
    [24]Ou DP, Tao YM, Tang FQ, et al. The hepatitis B virus X protein promotes hepatocellular carcinoma metastasis by upregulation of matrix metalloproteinases. Int J Cancer. 2007,120:1208-1214
    [25]Ou DP, Tao YM, Chang ZG, et al. Hepatocellular carcinoma cells containing hepatitis B virus X protein have enhanced invasive potential conditionally. Dig Liver Dis.2006,38(4):262-267
    [26]Pilkington GJ, Bjerkvig R, De Ridder L,et al. In vitro and in vivo models for the study of brain tumour invasion. Anticancer Res.1997,17(6B):4107-4109
    [27]陈宏颉.PTEN基因转染对胶质瘤细胞粘附力的影响.福州总医院学报,2004,11(3):181-182
    [28]Zhao J, Dong L, Lu B,et al. Down-regulation of osteopontin suppresses growth and metastasis of hepatocellular carcinoma via induction of apoptosis. Gastroenterology.2008,135(3):956-968
    [29]Zhang ZL, Liu ZS and Sun Q. Anti-tumor effect of thalidomide and paclitaxel on hepatocellular carcinoma in nude mice. Chin Med J,2005,118(20):1688-1694
    [30]Li WC, Ye SL, Sun RX, et al. Inhibition of growth and metastasis of human hepatocellular carcinoma by antisense oligonucleotide targeting signal transducer and activator of transcription 3. Clin Cancer Res,2006,12(23):7140-7148
    [31]Aravalli RN, Steer CJ, Cressman EN. Molecular mechanisms of hepatocellular carcinoma. Hepatology,2008,48(6):2047-2063
    [32]Takai Y, Sasaki T, Matozaki T. Small GTP-binding proteins. Physiol Rev, 2001,81(1):153-208
    [33]Giannelli G, Antonaci S.Novel concepts in hepatocellular carcinoma:from molecular research to clinical practice. J Clin Gastroenterol,2006,40(9):842-846
    [34]Ou DP, Yang LY, Huang GW, et al. Clinical analysis of the risk factors for recurrenceof HCC and its relationship with HBV. World J Gastroenterol,2005,11 (14):2061-2066
    [35]Yang LY, Fang F, Ou DP, et al. Solitary large hepatocellular carcinoma:a specific subtype of hepatocellular carcinoma with good outcome after hepatic resection. Ann Surg,2009,249(1):118-123
    [36]Yang LY, Tao YM, Ou DP, et al. Increased expression of Wiskott-Aldrich syndrome protein family verprolin-homologous protein 2 correlated with poor prognosis of hepatocellular carcinoma. Clin Cancer Res,2006,12(19):5673-5679
    [37]Wu F, Yang LY, Li YF, et al. Novel role for epidermal growth factor-like domain 7 in metastasis of human hepatocellular carcinoma. Hepatology,2009,50(6):
    1839-1850
    [38]Ishida D, Kometani K, Yang H, et al.Myeloproliferative stem cell disorders by deregulated Rapl activation in SPA-1-deficient mice. Cancer Cell,2003,4(1): 55-65
    [39]Martin MA, Bhatia M. Analysis of the human fetal liver hematopoietic microenvironment. Stem Cells Dev,2005;14(5):493-504
    [40]Honn KV, Tang DG, Crissman JD. Platelets and cancer metastasis:a causal relationship?Cancer Metastasis Rev,1992;11(3-4):325-351
    [41]Kurachi H, Wada Y, Tsukamoto N, et al. Human SPA-1 gene product selectively expressed in lymphoid tissues is a specific GTPase-activating protein for Rapl and Rap2:segregate expression profiles from a rap1 GAP gene product. J Biol Chem,1997,272(44):28081-28088
    [42]高进,章静波.癌的侵袭与转移基础与临床.北京:科学出版社,2003.123-150
    [43]Yang Y, Nagano H, Ota H, et al. Patterns and clinicopathologic features of extrahepatic recurrence of hepatocellular carcinoma after curative resection. Surgery,2007,141 (2):196-202
    [44]Poon RT, Fan ST, Wong J, et al. Risk factors, prevention, and management of postoperative recurrence after resection of hepatocellular carcinoma. Ann Surg, 2000,232(1):10-24
    [45]Piao Z, Kim H, Jeon BK, et al. Relationship between loss of heterozygosity of tumor suppressor genes and histologic differentiation in hepatocellular carcinoma-Cancer,1997,80(5):865-872
    [46]杨连粤,刘合利,黄耿文,等.孤立性大肝癌分子病理特征的初步研究.中华实验外科杂志.2004,21(1):94-96
    [47]Yang LY, Wang W, Peng JX, et al. Differentially expressed genes between solitary large hepatocellular carcinoma and nodular hepatocellular carcinoma. World J Gastroenterol,2004; 10(24):3569-3573
    [48]杨连粤,常志刚.孤立性大肝癌的特征及治疗策略.中国医刊,2004,39(2):14-17
    [49]Felgner PL, Gadek TR, Holm M, et al. Lipofection:a highly efficient, lipid-mediated DNA-transfection procedure. Proc Natl Acad Sci USA,1987,84 (21):7413-7417
    [50]Gupta GP, Massague J.Cancer metastasis:building a framework. Cell, 2006,127(4):679-695
    [51]Wang H, Hou Z, Wu Y, et al. p150 ADAR1 isoform involved in maintenance of HeLa cell proliferation.BMC Cancer,2006,6:282
    [52]Benoliel AM, Pirro N, Marin V, et al. Correlation between invasiveness of colorectal tumor cells and adhesive potential under flow. Anticancer Res,2003, 23(6c):4891-4896
    [53]Nystrom ML, Thomas GJ, Stone M, et al. Development of a quantitative method to analyse tumour cell invasion in organotypic culture. J Pathol,2005,205 (4): 468-475
    [54]Wang W, Yang LY, Yang ZL, et al. Elevated expression of autocrine motility factor receptor correlates with overexpression of RhoC and indicates poor prognosis in hepatocellular carcinoma. Dig Dis Sci,2007,52(3):770-775
    [55]Carlier MF, Pantaloni D. Control of actin assembly dynamics in cell motility.J Biol Chem,2007,282(32):23005-23009
    [56]彭方兴,严律南.肝癌动物模型的研究进展.中华实验外科杂志,2000,17(5):475-476
    [57]Li Y, Tian B, Yang J, et al. Stepwise metastatic human hepatocellular carcinoma cell model system with multiple metastatic potentials established through consecutive in vivo selection and studies on metastatic characteristics. J Cancer Res Clin Oncol,2004,130(8):460-468
    [58]Li Y, Tang Y, Ye L, et al. Establishment of a hepatocellular carcinoma cell line with unique metastatic characteristics through in vivo selection and screening for metastasis-related genes through cDNA microarray. J Cancer Res Clin Oncol, 2003,129(1):43-51
    [59]M'Rabet L, Coffer P, Zwartkruis F, et al.Activation of the small GTPase rapl in human neutrophils.Blood,1998,92(6):2133-2140
    [60]Lorenowicz MJ, van Gils J, de Boer M,et al, Epacl-Rapl signaling regulates monocyte adhesion and chemotaxis. J Leukoc Biol,2006;80(6):1542-1552
    [61]Hattori M, Minato N.Rapl GTPase:functions, regulation, and malignancy. J Biochem,2003,134(4):479-484
    [62]Bos JL, de Bruyn K, Enserink J, et al. The role of Rap 1 in integrin-mediated cell adhesion.Biochem Soc Trans,2003,31(Pt 1):83-86
    [63]Giancotti FG, Ruoslahti E. Integrin signaling. Science,1999,285 (5430):1028-1032
    [64]Kitayama H, Sugimoto Y, Matsuzaki T, et al. A ras-related gene with transformation suppressor activity. Cell,1989,56(1):77-84
    [65]Schmitt JM, Stork PJ.Cyclic AMP-mediated inhibition of cell growth requires the small G protein Rapl.Mol Cell Biol,2001,21(ll):3671-3683
    [66]Newell P, Toffanin S, Villanueva A, et al. Ras pathway activation in hepatocellular carcinoma and anti-tumoral effect of combined sorafenib and rapamycin in vivo. J Hepatol,2009,51(4):725-733
    [1]McKenna MT, Michaud CM, Murray CJ, et al. Assessing the burden of disease in the United States using disability-adjusted life years. Am J Prev Med, 2005,28(5):415-423
    [2]薛开先.肝癌发生的分子遗传学和表遗传学研究.癌症,2005,24(6):757-768
    [3]Sporn MB. The war on cancer. Lancet,1996,347(9012):1377-1381
    [4]周伟平,单云峰,王红阳,等.肝癌复发和转移的分子生物学研究.肝胆外科杂志,2004,12(1):6-8
    [5]Park YG, Zhao X, Lesueur F, et al. Sipal is a candidate for underlying the metastasis efficiency modifier locus Mtesl. Nat Genet,2005,37(10):1055-1062
    [6]Hsieh SM, Look MP, Sieuwerts AM, er al. Distinct inherited metastasis susceptibility exists for different breast cancer subtypes:a prognosis study. Breast Cancer Res,2009,11(5):R75.
    [7]Sprent J. Lifespans of naive, memory and effector lymphocytes. Curr Opin Immunol,1993,5(3):433-438
    [8]Hattori M, Tsukamoto N, Nur-e-Kamal MS, et al. Molecular cloning of a novel mitogen-inducible nuclear protein with a Ran GTPase-activating domain that affects cell cycle progression. Mol Cell Biol,1995,15(1):552-560
    [9]Kurachi H, Wada Y, Tsukamoto N, et al. Human SPA-1 gene product selectively expressed in lymphoid tissues is a specific GTPase-activating protein for Rapl and Rap2:segregate expression profiles from a rapl GAP gene product. J Biol Chem, 1997,272(44):28081-28088.
    [10]Rubinfeld B, Crosier WJ, Albert I, Conroy L, Clark R, McCormick F, Polakis P. Localization of the rapl GAP catalytic domain and sites of phosphorylation by mutational analysis. Mol Cell Biol,1992,12(10):4634-4642
    [11]Kitayama H, Sugimoto Y, Matsuzaki T, Ikawa Y, Noda M. A ras-related gene with transformation suppressor activity. Cell,1989,56(1):77-84
    [12]Zhu JJ, Qin Y, Zhao M, et al. Ras and Rap control AMPA receptor trafficking during synaptic plasticity.Cell,2002,110(4):443-455
    [13]Bos JL, de Bruyn K, Enserink J, et al. The role of Rap 1 in integrin-mediated cell adhesion.Biochem Soc Trans,2003,31(Pt 1):83-86
    [14]Stork PJ, Dillon TJ.Multiple roles of Rapl in hematopoietic cells: complementary versus antagonistic functions.Blood,2005,106(9):2952-2961
    [15]Hattori M, Minato N. Rapl GTPase:functions, regulation, and malignancy. J Biochem,2003,134(4):479-484
    [16]Ishida D, Kometani K, Yang H, et al.Myeloproliferative stem cell disorders by deregulated Rapl activation in SPA-1-deficient mice.Cancer Cell,2003,4(1):55-65
    [17]Kometani K, Ishida D, Hattori M, et al.Rapl and SPA-1 in hematologic malignancy.Trends Mol Med,2004,10(8):401-408
    [18]Birnbaum RA, O'Marcaigh A, Wardak Z, et al. Nfl and Gmcsf interact in myeloid leukemogenesis.Mol Cell,2000,5(1):189-195
    [19]王杰军,高勇,许青.肿瘤转移机制及诊疗进展.上海:第二军医大学出版社,2002.76-77
    [20]Farshid M, Tabor E. Expression of oncogenes and tumor suppressor genes in human hepatocellular carcinoma and hepatoblastoma cell lines.J Med Virol, 1992,38(4):235-239
    [21]Uchida T, Gao JP, Wang C, et al.Clinical significance of p53, mdm2, and bcl-2 proteins in renal cell carcinoma.Urology,2002,59(4):615-620
    [22]Guo HB, Liu F, Zhao JH, et al. Down-regulation of N-acetylglucosaminy-ltransferase V by tumorigenesis-or metastasis-suppressor gene and its relation to metastatic potential of human hepatocarcinoma cells. J Cell Biochem, 2000,79(3):370-385
    [23]Giatromanolaki A, Stathopoulos GP, Tsiobanou E, et al.Combined role of tumor angiogenesis, bcl-2, and p53 expression in the prognosis of patients with colorectal carcinoma.Cancer,1999,86(8):1421-1430
    [24]Endo K, Terada T. Protein expression of CD44 (standard and variant isoforms) in hepatocellular carcinoma:relationships with tumor grade, clinicopathologic parameters, p53 expression, and patient survival.J Hepatol,2000,32(1):78-84
    [25]刘合利,杨连粤,黄耿文,等.整合素av亚基对肝细胞性肝癌血管生成、侵袭转移的影响.中华普通外科杂志,2002,17(9):542-543
    [26]McKenna GJ, Chen Y, Smith RM, et al. A role for matrix metalloproteinases and tumor host interaction in hepatocellular carcinomas.Am J Surg,2002,183(5): 588-594
    [27]Rabbani SA, Gladu J.Urokinase receptor antibody can reduce tumor volume and detect the presence of occult tumor metastases in vivo.Cancer Res,2002,62(8): 2390-2397
    [28]Huang GW, Yang LY, LuWQ. Expression of hypoxia-inducible factor 1 alpha and vascular endothelial growth factor in hepatocellular carcinoma:Impact on neovascularization and survival.World J Gastroenterol,2005,11 (11):1705-1708
    [29]Lifsted T, Le Voyer T, Williams M, et al. Identification of inbred mouse strains harboring genetic modifiers of mammary tumor age of onset and metastatic progression.Int J Cancer,1998,77(4):640-644
    [30]Crawford NP, Qian X, Ziogas A, et al. Rrplb, a new candidate susceptibility gene for breast cancer progression and metastasis. PLoS Genet,2007;3(11):e214
    [31]Collins FS, Green ED, Guttmacher AE, et al. A vision for the future of genomics research. Nature,2003,422(6934):835-847
    [32]杨永强,王巍杰,徐长波.单核苷酸多态性研究进展.化学与生物工程,2009,26(8):19-21
    [33]纪龙,余红平.单核苷酸多态性、环境因素与肝细胞肝癌遗传易感性的关系.卫生研究,2009,38(2):233-237
    [34]Crawford NP, Ziogas A, Peel DJ, et al. Germline polymorphisms in SIPA1 are associated with metastasis and other indicators of poor prognosis in breast cancer. Breast Cancer Res,2006;8(2):R16
    [35]Hsieh SM, Look MP, Sieuwerts AM, et al. Distinct inherited metastasis susceptibility exists for different breast cancer subtypes:a prognosis study. Breast Cancer Res,2009,11(5):R75.
    [36]Brooks R, Kizer N, Nguyen L, et al. Polymorphisms in MMP9 and SIPA1 are associated with increased risk of nodal metastases in early-stage cervical cancer.Gynecol Oncol,2010,116(3):539-543
    [37]Threadgill DW. Metastatic potential as a heritable trait. Nat Genet,2005,37(10): 1026-1027
    [38]Nowell PC. The clonal evolution of tumor cell populations. Science. 1976,194(4260):23-28
    [39]Fidler IJ, Kripke ML.Metastasis results from preexisting variant cells within a malignant tumor.Science,1977,197(4306):893-895
    [40]Ramaswamy S, Ross KN, Lander ES, et al. A molecular signature of metastasis in primary solid tumors. Nat Genet.2003,33(1):49-54
    [41]Wang W, Yang LY, Huang GW, et al.Genomic analysis reveals RhoC as a potential marker in hepatocellular carcinoma with poor prognosis.Br J Cancer, 2004,90(12):2349-2355
    [42]杨连粤,常志刚.孤立性大肝癌的特征及治疗策略.中国医刊,2004,39(2):14-17
    [43]Yang LY, Fang F, Ou DP, et al. Solitary large hepatocellular carcinoma:a specific subtype of hepatocellular carcinoma with good outcome after hepatic resection. Ann Surg,2009;249(1):118-123
    [44]Hunter KW. Allelic diversity in the host genetic background may be an important determinant in tumor metastatic dissemination. Cancer Lett,2003,200(2):97-105
    [45]Budhu AS, Zipser B, Forgues M, et al. The molecular signature of metastases of human hepatocellular carcinoma. Oncology,2005,69 (Suppl 1):23-27
    [46]Ye QH, Qin LX, Forgues M, et al.Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning. Nat Med,2003,9(4):416-423
    [47]周信达.肝癌转移复发的研究进展与展望.中华肝脏病杂志,2004,12(11):690

© 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