Cdc42-shRNA表达载体的构建及其对肝癌细胞生物学行为的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:构建并转染针对肝癌细胞系沉默Cdc42的高效率RNA干扰载体。在体内外实验探讨Cdc42-shRNA重组质粒对肝癌细胞SMMC-7721增殖、迁移、侵袭等恶性生物学行为的影响。
     方法:根据Genebank报道的Cdc42的mRNA序列,利用Promega公司siRNA在线设计软件设计特征的靶序列,分别合成3对寡核苷酸链,克隆到含U6启动子的pBS/U6载体,进行酶切鉴定和测序。采用脂质体法将构建的载体转染至肝癌细胞SMMC-7721中,用Westernblot及免疫荧光染色检测对Cdc42蛋白表达的抑制情况。采用计数法绘制细胞生长曲线;MTT法检测细胞增殖;划痕愈合实验方法测定细胞迁移能力;穿孔迁移实验方法观察细胞的侵袭能力。Westernblot方法检测PCNA和AFP蛋白表达。建立裸鼠皮下人肝癌移植瘤模型,观察裸鼠成瘤情况,免疫组织化学方法检测裸鼠瘤块中Cdc42的表达情况。
     结果:酶切鉴定和测序结果证实Cdc42-shRNA重组质粒构建完全正确;转染肝癌细胞SMMC-7721后,Westernblot检测肝癌细胞内源性Cdc42表达明显抑制。免疫荧光方法结果显示肝癌细胞荧光强度显著下降。细胞计数法显示转染Cdc42-shRNA2细胞生长速度明显减慢。MTT实验显示转染重组质粒后吸光度显著降低,并且呈剂量依赖性。划痕实验显示转染Cdc42-shRNA2的SMMC-7721细胞划痕愈合能力显著减弱,相对迁移距离比较差异显著。穿孔迁移实验方法显示肝癌细胞侵袭能力明显减弱。显著地抑制肝癌细胞的肿瘤标志蛋白AFP的表达水平及细胞增殖抗原PCNA表达水平。裸鼠成瘤实验转染组肿瘤生长速度减慢,瘤块重量明显减小。瘤块免疫组化结果Cdc42表达明显减弱。
     结论:成功构建了Cdc42-shRNA表达载体;并成功转染肝癌SMMC-7721细胞,高效地抑制了肝癌细胞内源性Cdc42的表达。Cdc42干扰质粒成功地转染肝癌SMMC-7721细胞后,明显改变了人肝癌细胞的恶性生物学行为,细胞生长速度减慢,增殖能力降低,细胞的迁移和穿过Matrigel胶的侵袭能力均有不同程度的降低;发现RNAi沉默Cdc42表达导致了AFP蛋白和PCNA蛋白表达含量的明显改变,降低肝癌细胞的恶性程度;体内成瘤实验也证实RNA干扰技术可沉默肝细胞癌Cdc42蛋白的表达,并且明显抑制肿瘤生长。
BACKGROUNDBACKGROUNDS:Primary hepatocellular carcinoma(HCC)is a commonmalignant tumor of digestive system with poor early diagnosis;the most patiens ofdiagnosis have reached the advanced or distant metastasis; In recent years, livercancer has been greatly improved with multidisciplinary treatment modalities,but theoverall curative effect is bad. Therefore, we must better understand the incidence anddevelopment mechanism of hepatocellular carcinoma. To find a new treatmentstrategies to improve the prognosis of liver cancer is the focus of the study.
     The occurrence and development of HCC is a multi-gene and multi-stepprocess,with the development of gene technology,the research of gene treatment toHCC become a hotspot worldwide.RNA interference(RNAi)means degradation ofhomologous mRNA induced by endogenous or exogenous short double-strandedRNA(dsRNA)molecule,its effector molecule is 21-23 nucleotide(nt)dsRNA calledsmall interfering RNA(siRNA).The gene knockout effect caused by RNAi technologycan inhibit the expression of oncogene,tumor suppression gene and gene mutatin etal.The short hairpin RNA(shRNA) expressed by vector transfected into target cellscan be recognized and splitted into siRNA.Use the plasmid vector system which cansteadily express shRNA can inhibit gene expression permanently.
     The current study found that, the most important function of Rho GTPases is toregulate the actin-related protein and cytoskeleton-mediated construction, and relatedclosely with the malignant transformation, growth, invasion and metastasis, cell cycleregulation and apoptosis. It is clear that Rho proteins was involved almost in everystage of tumor formation and played an important role of the pathophysiologicalprocess in the tumor,and Rho proteins is promising target for cancer treatment that hasbecome one of the research frontier.Cdc42(Cell division cycle42),a member of RhoGTPases,Involved in actin reorganization, formation of cell polarity, pseudopodgeneration, membrane transport and transcriptional regulation of expression and otherphysiological processes,and acts as a GTP-binding Protein switch to regulate multiple signal transduction pathways that control key cellular proeesses such as cellproliferation,survival,cytoarehitecture,adhesion,migration,and transcriptionalregulation. Several studies found that Cdc42 is over-expression in breast cancer, lungcancer, esophagus cancer, colon cancer, bladder cancer,and other organizations andtumor cells. In hepatocellular carcinoma tissue Cdc42 molecules expression wassignificantly increased than non-tumor liver tissue,and in highly metastatichepatocellular carcinoma cell lines expressed more than in low metastatichepatocellular carcinoma cell lines. Therefore, speculated that the protein may be inhepatocellular carcinoma cell proliferation and transformation play a role.
     At present, the application RNA interference technology research Cdc42 affectthe biological behavior of hepatocellular carcinoma cells not been reported at homeand abroad. We studied hepatocellular carcinoma cell lines to explore the Cdc42protein in liver cancer cell proliferation, migration, invasion and so on role and theapplication of RNA interference inhibition of the Cdc42 protein and cell biologicalbehavior in order to study the role of Cdc42 in the occurrence and development ofhepatocellular carcinoma, and exploration of the feasibility of targeted Cdc42treatment of hepatocellular carcinoma.
     This study is to establish the Cdc42-shRNA vector and transfect the hepatocellularcarcinoma cells SMMC-7721,detect the inhibition effect of Cdc42 expression,andobserve the growth,proliferation,mobility of SMMC-7721 cells and the expression ofAFP ansd PCNA;establish the nude mice model transplanted with SMMC-7721 cellssubcutaneously to observe the growth of transplanted tumor.Thus to provide a neweffective target for the treatment of HCC. This study is divided into three parts:
     1 Construction and identification of Cdc42-shRNA and its inhibition effect onCdc42 expression of hepatoma cells.
     Objective: To construct high efficiency of RNA silencing Cdc42 interferingcarrier. for liver cancer cell lines
     Methods According to the Cdc42 mRNA sequence in Genebank,use the siRNAdesign software of Promega to find the right target sequence,BLAST to confirm nohomologous with known human gene.Three pair of oligonucleotide strand wassynthesized.The sequence is 19nt sense sequence,6nt loop and 19nt anti-sensesequence and terminal signal,cloned the sequence to pBS/U6 vector.Rename the recombinant plasmid Cdc42-shRNA1、Cdc42-shRNA2、Cdc42-shRNA3,andrestriction enzyme digestion and sequencing to confirm the correctness.Transfect theplasmid into hepatoma cells SMMC-7721 to detect the expression of Cdc42 byWesternblot and immunofluorescence.
     Results: The enzyme electrophoresis confirmed that Cdc42-shRNA plasmid wassuccessfully constructed, inserted fragment sequencing results with the expected resultsare consistent with the synthesis of siRNA confirmed the synthesis of recombinantplasmid completely correct; Westernblot detect Cdc42-shRNA inhibited SMMC-7721hepatocellular carcinoma cells expressed endogenous Cdc42. Cdc42-shRNAsrecombinant plasmid is completely correct comfirmed by restriction enzyme digestionand sequencing.All the plasmids have gene silencing effect and the Cdc42-shRNA2 ismost significant.It can inhibit the endogenous Cdc42 expression about 80%.Thefluorescence intensity of hepatoma cells transfected by Cdc42- shRNA2 is much lower.
     Conclusion: The successful construction of the high efficiency of RNAi silencingCdc42 expression vectors, the restriction enzyme digestion and sequencing confirmedthe plasmid completely correct; successfully transfected with recombinant plasmidliver cancer SMMC-7721 cells, liver cells efficiently inhibited endogenous Cdc42expression. Tips, RNAi technology can be used as Cdc42 silence the expression ofliver cancer in an effective way for further experiments laid the experimental basis.
     2.Influence of Cdc42-shRNA to proliferation and mobility and expression ofAFP and PCNA of hepatoma cells.
     Objective: To study the influence of Cdc42-shRNA plasmid to proliferation,migration, invasion and other malignant biological behavior in hepatoma SMMC-7721cells.
     Methods: PBS/U6 Cdc42-shRNA interfering vector transfected to SMMC-7721cells with liposome method,The growth curve of ransfected cells SMMC-7721、SMMC-7721/pBS/U6-control、SMMC-7721/Cdc42-shRNA2 was detected bycountnumber method;the cells proliferation was detected by MTT,and calculate theinhibition rate.The cells mobility was detected by wound healing experiment.Transwell chamber experiments to observe the cell migration and invasion.DetectAFP and PCNA expression level by Westernblot.
     Results:The growth of hepatoma cells transfected by Cdc42-shRNA2 isslower.The absorbance of SMMC-7721 cells transfected by10ng and 30ng Cdc42-shRNA2 is much lower in MTT experiment,and there is a dose-dependentrelationship,the maximum inhibition rate is 40%. The wound healing capatility ofSMMC-7721 cells transfected by Cdc42-shRNA2 is weaker than pBS/U6-controlgroup with comparison of relative mobility distance significantly different.Transwellchamber experimental methods showed Cdc42 protein expression after the silence,SMMC-7721 decreased significantly invasion ability.The AFP and PCNA expressionof hepatoma cells is siginificantly inhibited after the Cdc42-shRNA2 was transfectedcompared with pBS/U6-control group,the inhibition rate is about 75%-80%.
     Conclusion: Cdc42 interfere with plasmid successfully transfected hepatomaSMMC-7721 cells, significant changes in human malignant biological behavior ofhepatocellular carcinoma cells, cell growth slowed down to reduce proliferation, cellcycle arrest, cell migration and the invasive ability of plastic have different degrees ofreduced through the Matrigel.RNAi silencing Cdc42 led expression level of AFPprotein expression to significant changes in content, and reduce liver cancer cellinvasion and metastasis of capacity;
     3.Effect of Cdc42-shRNA vector on nude mice model transplanted with hepatomacells subcutaneously.
     Objective: To evaluate the in vivo gene silencing technology, the impact ofinterference with Cdc42 on the hepatocellular carcinoma.
     Methods:15 BALB/c-nu/nu mice was randomly divided into 3 groups,5 eachgroup;group A was inoculated with SMMC-7721,and B group with SMMC- 7721/pBS/U6-control and C group SMMC-7721/Cdc42-shRNA2.The nude mice modelwasestablished with 6×106/0.2ml cells inoculated subcutaneously in the armpit.Afterthe tumor grown,measure the length and width and calculate the volume of tumorevery 7 days.Execute the nude mice on the 28th day and cut the tumor and weigh,calculate the inhibition rate.Detect the expression of Cdc42 of the tumor by immunohistochemistry.Results Nude mice suffered from tumor blocks after inoculation of tumor cells in.
     In which A group and B group can be the 6th day after inoculation into the tumor,while the C composition of tumors was delayed until 8-11 days after inoculation. Withthe extension of feeding time, the tumor block increased gradually, without thephenomenon of spontaneous remission. Tumor volume growth curves show that thetumor volume of group A and group B is about same otherwise group C is siginificantly smaller.All the nude mice were executed on 28th day and the tumor wereweight,the tumor weight of group C is much lighter than group A and B,the inhibitionrate is 63.7%.The Cdc42 expression is strong positive in group A and B,but weakpositive in group C.
     Conclusion:In vivo tumor experiments also confirmed that RNA interferencetechnology can be silent Cdc42 protein expression in hepatocellular carcinoma andsilencing Cdc42 expression could significantly inhibit tumor growth.
     In conclusion, a high efficiency of RNAi silencing Cdc42 expression vectors wassuccessfully constructed, transfected hepatoma SMMC-7721 cells and inhibitedefficiently the expression of endogenous Cdc42 protein. RNAi technology can be usedas silence the Cdc42 expression of liver cancer in an effective way for furtherexperiments to lay an experimental basis; Cdc42 interfere with plasmid successfullytransfected hepatoma SMMC-7721 cells, significant changes in human malignantbiological behavior of hepatocellular carcinoma cells, cell growth slowed down toreduce proliferation, cell cycle arrest, cell migration and the invasive ability of plastichave different degrees of reduced through the Matrigel.RNAi silencing Cdc42 ledexpression level of AFP protein expression to significant changes in content, and reduceliver cancer cell invasion and metastasis of capacity; In vivo tumor experiments alsoconfirmed that RNA interference technology can be silent Cdc42 protein expression inhepatocellular carcinoma and silencing Cdc42 expression could significantly inhibittumor growth.
     This test verified Cdc42 is a important elements of development and progressionof liver cancer. Cdc42 liver cancer is most likely a potential therapeutic target, with abroad clinical application.
引文
[1] Caldwell S,Park SH.The epidemiology of hepatocellular cancer:from theperspectives of public health problem to tumor biology[J].J Gastroenterol,2009,44(Suppl19):96-101.
    [2] Napoli C,Lemieux C,Jorgensen R.Introduction of a Chimeric Chalcone SynthaseGene into Petunia Results in Reversible Co-Suppression of Homologous Genes intrans[J]. Plant Cell,1990,2(4):279-289.
    [3] Van der Krol AR,Mur LA, de Lange P,Mol JN,et al.Inhibition of flowerpigmentation by antisense CHS genes:promoter and minimal sequencerequirements for the antisense effect[J].Plant Mol Biol, 1990,14(4):457-66.
    [4] Guo S,KemPhues KJ.Par-l a gene required for establishing Polarity in C.elegansembryos,eneodes a Putative Ser/Thr kinase that is asymmetrieally distributed[J].Cell,1995,81(4):611-620.
    [5] Fire A,Xu S,Montgomery MK,et al.Potent and specific genetic interference bydouble-stranded RNA in Caenorhabditis elegans[J].Nature,1998,391(6669): 806-811.
    [6] Tuschl T,Zamore PD,Lehmann R,et al.Targeted mRNA degradation bydouble-stranded RNA in vitro[J].Genes Dev,1999,13(24):3191-3197.
    [7] Elbashir SM,Harborth J,Lendeekel W,et al.DuPlexes of nucleotide RNAsMediate RNA interference in cultured mammalian cells[J].Nature,2001,411(6836):494-498.
    [8] Bernstein E,Caudy AA,Hammond SM,et al.Role for a bidentate ribonuclease inthe initiation step of RNA interference[J].Nature,2001,409(68):363-366.
    [9] Brummelkamp TR,Bernards R,Agami R.A system for stable expression of shortinterfering RNAs in mammalian cells[J].Science,2002,296(5567):550-553.
    [10] Sui GS,Soohoo C,Affar EB.A DNA vector based RNAi technology to suppressiongene expression in mammalian cells[J].PANS,2002,99(8):5515-5521.
    [11] Bosher JM,Labouesse M.Rna Interference:Genetic Wand and Genetic Watchdog[J].Nat Cell Biol,2000,2(2):E31-36.
    [12] Plasterk RH.Rna Silencing:The Genome's Immune System[J].Science, 2002,296(5571):1263-1265.
    [13] McManus MT.,Sharp PA.Gene Silencing in Mammals by Small InterferingRnas[J].Nat Rev Genet,2002,3(10):737-747.
    [14] Howard-Till RA, Yao MC. Induction of gene silencing by hairpin RNAexpression in Tetrahymena thermophila reveals a second small RNA pathway[J].Mol Cell Biol, 2006,26(23): 8731-8742.
    [15] Nykanen A,Haley B,Zamore PD.ATP requirements and small interfering RNAstructure in the RNA interference pathway[J].Cell,200l,107(3):309-321.
    [16] Calderon AJ, Lavergne JA. RNA interference: a novel and physiologicmechanism of gene silencing with great therapeutic potential[J]. P R Health SciJ, 2005,24(1): 27-33.
    [17] Zhang DX,Pan B,Liu B,et al. Experimental strategies of the application of RNAitechnique in mammalian cells[J]. Yi Chuan, 2005,27(5): 839-844.
    [18] Zhou D, He QS, Wang C, Zhang J, Wong-Staal F. RNA interference andpotential applications[J]. Curr Top Med Chem, 2006,6(9): 901-911.
    [19] Nykanen A,Haley B,Zamore PD.ATP requirements and small interfering RNAstructure in the RNA interference pathway[J].Cell,200l,107(3):309-321.
    [20] Haussecker D, Proudfoot NJ. Dicer-dependent turnover of intergenic transcriptsfrom the human beta-globin gene cluster[J]. Mol Cell Biol, 2005,25(21): 9724-9733.
    [21] Xie Z, Allen E, Wilken A, Carrington JC. DICER-LIKE 4 functions intrans-acting small interfering RNA biogenesis and vegetative phase change inArabidopsis thaliana[J]. Proc Natl Acad Sci U S A, 2005,102(36): 12984-12989.
    [22] Harris KS, Zhang Z, McManus MT, Harfe BD, Sun X. Dicer function isessential for lung epithelium morphogenesis[J]. Proc Natl Acad Sci U S A,2006,103(7): 2208-2213.
    [23] Kuehbacher A, Urbich C, Zeiher AM, Dimmeler S. Role of Dicer and Droshafor endothelial microRNA expression and angiogenesis[J]. Circ Res, 2007,101(1): 59-68.
    [24] Luo H, Hu DX, Chen P.Dicer efficiently converts large dsRNAs into siRNAssuitable for COX-2 gene[J]. Zhong Nan Da Xue Xue Bao Yi Xue Ban, 2007,32(3): 437-442.
    [25] Suarez Y, Fernandez-Hernando C, Pober JS, Sessa WC. Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells[J]. CircRes, 2007,100(8): 1164-1173.
    [26] Jaskiewicz L, Filipowicz W. Role of Dicer in posttranscriptional RNA silencing[J]. Curr Top Microbiol Immunol, 2008,320: 77-97
    [27] Tijsterman M, Plasterk RH. Dicers at RISC; the mechanism of RNAi[J]. Cell,2004,117(1): 1-3.
    [28] Hannon GJ.RNA interference[J].Nature,2002,418(6894):244-251.
    [29] Tuschl T,Zamore PD,Lehmann R,et al.Targeted mRNA degradation by doublestrandedRNA in vitro[J].Genes,1999,13(24):3191-3197.
    [30] Rand TA,Petersen S,Du F,et al.Argonaute 2 cleaves the anti-guide strand ofsiRNA during RISC activation[J].Cell,2005,123(4):621-629.
    [31] Sen GL, Blau HM. Argonaute 2/RISC resides in sites of mammalian mRNAdecay known as cytoplasmic bodies[J]. Nat Cell Biol, 2005,7(6): 633-636.
    [32] Vestin A, Weinstein J, Davidov E, Sidi Y, Yakobson EA. Gene silencing RNAitechnology: possible application to therapy[J]. Harefuah, 2006,145(2): 156-159,163.
    [33] Aigner A. Applications of RNA interference: current state and prospects forsiRNA-based strategies in vivo[J]. Appl Microbiol Biotechnol, 2007,76(1): 9-21.
    [34] Maliogka VI, Dovas CI, Katis NI. Demarcation of ilarviruses based on thephylogeny of RNA2-encoded RdRp and a generic ramped annealing RT-PCR[J].Arch Virol, 2007,152(9): 1687-1698.
    [35] Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl TDuplexes of21-nucleotide RNAs mediate RNA interference in cultured mammalian cells[J].Nature, 2001,24,411(6836):494-8.
    [36] Tuschl T,Zamore PD,Lehmann R,et al.Targeted mRNA degradation bydouble-stranded RNA in vitro[J].Genes Dev,1999,13(24):3191-3197.
    [37] Reynolds A,Leake D,Boe3e Q,et a1.Rational siRNA design for RNAinterference[J].Nat Biotechnol,2004,22(3):326-30.
    [38] Ui-Tei K, Saigo K.Molecular mechanism of RNA interference and the selectionof highly effective siRNA sequences[J].Tanpakushitsu Kakusan Koso,2004,49(17 Suppl):2662-70.
    [39] Luo KQ, Chang DC.The gene-silencing efficiency of siRNA is stronglydependent on the local structure of mRNA at the targeted region[J].BiochemBiophys Res Commun, 2004,21,318(1):303-10.
    [40] Schwarz DS, Hutvágner G, Du T, Xu Z, Aronin N, Zamore PD.Asymmetry inthe assembly of the RNAi enzyme complex[J].Cell,2003,17,115(2):199-208.
    [41] Zamore PD,Tuschl T,Sharp PA,et al.Double-Stranded Rna Directs theAtp-Dependent Cleavage of Mrna at 21 to 23 Nucleotide Intervals[J]. Cell,2000,101(1):25-33.
    [42] Davenport RJ.Gene Silencing.A Faster Way to Shut Down Genes[J].Science,2001,292(5521):1469-1471.
    [43] Hannon GJ.Rna Interference[J].Nature,2002,418(6894):244-251.
    [44] Keating CD,Kriek N,Daniels M,et al.Whole-genome analysis of 60 Gprotein-coupled receptors in Caenorhabditis elegans by gene knockout withRNAi[J].Curr Biol,2003,13(19):1715-1720.
    [45] Jackson AL,Bartz SR,Schelter J,et al.Expression profiling reveals off-target generegulation by RNAi[J].Nat Biotechnol,2003,21(6):635–637.
    [46] Jackson AL,Burchard J,Schelter J,et al.Widespread siRNA“off-target”transcriptsilencing mediated by seed region sequence complementarity[J]. RNA, 2006,12(7):1179–1187.
    [47] Kariko K,Bhuyan P,Capodici J,et al.Small interfering RNAs mediate sequenceindependent gene suppression and induce immune activation by signalingthrough toll-like receptor 3[J].J Immunol,2004,172(11):6545–6549.
    [48] Barik S. Silence of the transcripts: RNA interference in medicine[J]. J Mol Med,2005,83(10): 764-773.
    [49] Aagaard L, Rossi JJ. RNAi therapeutics: principles, prospects and challenges[J].Adv Drug Deliv Rev, 2007,59(23): 75-86.
    [50] Xu Y, Wang YH, Gao JD,et al.Suppression of c-myc expression by interferenceRNA in HepG2 hepatocellular carcinoma cells[J].Zhonghua Zhong Liu ZaZhi,2004,26(8):458-60.
    [51] Hx Sun, Hw He, Sh Zhang,et al.Suppression of N-Ras by shRNA-expressingplasmid increases sensitivity of HepG2 cells to vincristine-induced growthinhibitionSuppression of N-Ras by shRNA-expressing plasmid[J].Cancer GeneTherapy 2009,16, 693-702 .
    [52] Chan DW, Ng IO.Knock-down of hepatitis B virus X protein reduces thetumorigenicity of hepatocellular carcinoma cells[J]. J Pathol, 2006Feb,208(3):372-80.
    [53] Li H, Fu X, Chen Y,et al.Use of adenovirus-delivered siRNA to target oncoproteinp28GANK in hepatocellular carcinoma[J].Gastroenterology,2005, 128(7):2029-41.
    [54] Cho-Rok J, Yoo J, Jang YJ, Kim S, Chu IS, Yeom YI, Choi JY, ImDS.Adenovirus-mediated transfer of siRNA against PTTG1 inhibits liver cancercell growth in vitro and in vivo[J]. Hepatology, 2006,43(5):1042-52.
    [55] Beraza N, Trautwein C.Restoration of p53 function: a new therapeutic strategyto induce tumor regression[J]? Hepatology, 2007,45(6):1578-9.
    [56] Wang S,Lan F,Huang L,et al.Suppression of hLRH-1 mediated by a DNAvector-based RNA interference results in cell cycle arrest and induction ofapoptosis in hepatocellular carcinoma cell BEL-7402[J].Biophys Res Commun,2005,333(3):917.
    [57] Li K,Lin SY,Brunicardi FC,et al.Use of RNA interference to target cyclinE-overexpressing hepatocellular carcinoma [J].Cancer Res,2003,63(1):3593-3597.
    [58] Ito T,Shira ki K,Sugimoto K,et al.Survivin promotes cell proliferation inhuman hepatocellular carcinoma[J].Hepatology,2000,31(5):1080-1085.
    [59] Chang Q,Liu ZR,Wang DY,et al.Survivin expression induced by daxorubicin incholangiocarcinoma.[J].World J Gastroenterol,2004,10(3):415-8.
    [60] Chang Q,Liu ZR,Wang DY,et al.Survivin expression induced by doxorubicin incholangiocarcinoma[J].World J Gastroenterol,2004,10(3):415-8.
    [61] Sheng-Quan Cheng Wen-Liang Wang Wei Yan,et al.Knockdown of survivingene expression by RNAi induces apoptosis in human hepatocellular carcinomacell line SMMC-7721[J]. World J Gastroenterology,2005,11(5):756-759.
    [62] Li J,Shen F,Wu D,et al.Expression level of Bcl-XL critically affects sensitivityof hepatocellular carcinoma cells to LIGHT-enhanced and interferon-gammainducedapoptosis[J].Oncol Rep,2007,17(5): 1067-1075.
    [63] Nieth C, Lage H.Induction of the ABC-transporters Mdr1/P-gp (Abcb1), mrpl(Abcc1), and bcrp (Abcg2) during establishment of multidrug resistancefollowing exposure to mitoxantrone[J]. J Chemother,2005 ,17(2):215-23.
    [64]张鹏辉,涂植光,杨明清,等.RNAi干扰技术靶向hTERT基因治疗肝癌的实验研究[J].癌症,2004,23(6):619-625.
    [65] Salvi A, Arici B, De Petro G, Barlati S.Small interfering RNA urokinasesilencing inhibits invasion and migration of human hepatocellular carcinomacells[J]. Mol Cancer Ther, 2004,3(6):671-8.
    [66]朱晓群,叶青海,雷科峰等,特异性小干扰性RNA降低骨桥蛋白基因表达对肝癌细胞侵袭性的影响[J].中华肿瘤杂志,2006,28(6):404-407.
    [67]瘳维甲,梅铭惠,徐静等.小干扰RNA抑制肝癌细胞株BEL7402ICAM-1基因表达的研究[J].中国现代医学杂志,2006,16(20):3065-3068.
    [68]刘权焰,刘志苏,邬开朗等.siRNA抑制肝癌细胞甲硫氨酸腺苷转移酶2A基因表达[J].中华实验外科杂志,2005,22(5):541-543
    [69] Ridley AJ.Rho proteins and cancer[J].Breast Cancer Res Treat.2004,84 (1):13-19.
    [70] Adams AE,Johnson DI,Longnecker BF,et al.Cdc42 and Cdc43,two additionalgenes involved in budding and the establishment of cell polarity in the yeastSaccharomyces cerevisiae[J].J Cell Biol, 1990,111(1):131-142.
    [71] Evans T,Brown ML,Fraser ED,et al.Purification of the major GTP-bindingproteins from human placental membranes[J].J Biol Chem,1986, 261(15):7052-7059.
    [72] Waldo GL,Evans T,Fraser ED,et al.Identification and purification from bovinebrain of a guanine-nucleotide-binding protein distinct from Gs, Gi and Go[J].Biochem J,1987.246(2):431-439.
    [73] Hart M,Polakis R,EvansT,et al.Identification and characterization of a lowmolecular weight GTP binding protein which is a phosphosubstrate for theepidermal growth factor receptor/ tyrosine kinase[J].J Biol Chem,1990,265,5990-6001.
    [74] Shinjo K, Koland JG,Hart MJ,et al.Molecular cloning of the gene for the humanplacental GTP-binding protein Gp (G25K): Identification of this GTP-bindingprotein as the human homolog of the yeast cell-division-cycle protein Cdc42[J].Proc Natl Acad Sci,1990,87(24),9853-9857.
    [75] Johnson DI,Pringle JR.Molecular characterization of Cdc42,a Saccharomycescerevisiae gene involved in the development of cell polarity[J]. J CellBiol,1990,111(1):143-152.
    [76] Mirbod F, Nakashima S, KitajimaY,et al.Molecular cloning of a Rho family,Cdc42Ca gene from Candida albicans and its mRNA expression changes duringmorphogenesis[J]. J Med Vet Mycol,1997,35(3):173-179.
    [77] Miller P,Johnson DI.Cdc42p GTPase is involved in controlling polarized cellgrowth in Schizosaccharomyces pombe[J].Mol Cell Biol,1994,14(2):1075-1083.
    [78] Chen W, Lim HH, Lim L.The Cdc42 homologue from Caenorhabditis elegans[J].J Biol Chem,1993,268(18):13280-13285.
    [79] Run JQ, Steven R,Hung MS,et al.Suppressors of the unc-73 gene of Caenorhabditiselegans[J].Genetics,1996,143(1):225-236.
    [80] Luo L,Liao YJ, Jan LY,et al.Distinct morphogenetic functions of similar smallGTPases: Drosophila Drac1 is involved in axonal outgrowth and myoblastfusion[J]. Genes,1994, 8(15):1787-1802.
    [81] Gong TW, Shin JJ, Burmeister M,et al. Complete cDNAs for Cdc42 fromchicken cochlea and mouse liver[J].Biochim.Biophy.Acta Gene Struct.Express,1997, 1352(2):282-292.
    [82] Shinjo K, Koland JG, Hart MJ, et al.Molecular cloning of the gene for thehuman placental GTP-binding protein Gp (G25K): Identification of thisGTP-binding protein as the human homolog of the yeast cell-division-cycleprotein Cdc42[J]. Proc Natl Acad Sci USA,1990,87(24):9853-7.
    [83] Marks PW, Kwiatkowski DJ.Genomic organization and chromosomal locationof murine Cdc42[J]. Genomics,1996,38(1):13-18.
    [84] Munemitsu S,Innis MA,Clark R,et al.Molecular cloning and expression of aG25K cDNA, the human homolog of the yeast cell cycle gene Cdc42[J]. MolCell Biol,1990,10(11):5977-5982.
    [85] Feltham, JL,Dotsch V,Raza S,et al.Definition of the switch surface in thesolution structure of Cdc42Hs[J]. Biochemistry,1997,36(29):8755-8766.
    [86] Hart, M. J.,Shinjo K,Hall A,et al.Identification of the human platelet GTPaseactivating protein for the Cdc42Hs protein[J].J Biol Chem,1991,266(31):20840-20848.
    [87] Shinjo K,Koland JG,Hart MJ,et al.Molecular cloning of the gene for the humanplacental GTP-binding protein Gp(G25K):identification of this GTP- bindingProtein as the human homolog of the yeast cell-division-cycle protein Cdc42[J].Proc Natl Acad Sci USA,1990:87(24):9853-7.
    [88] Wennerberg K,Der CJ.Rho-fanmily GTPases:it’s not only Rac and Rho(and Ilike it ) [J].J Cell Sci,2004,117(8):1301-1312.
    [89] Karnoub AE.Symons M,Campbell SL,et al.Molecular basis for Rho GTPasesignaling specificity[J].Breast Cancer Res Treat,2004,84(1):61-71.
    [90] Rossman KL, Der CJ, Sondek J.GEF means go:turning on RHO GTPases withguanine nucleotide-exchange factors[J]. Nat Rev Mol Cell Biol, 2005 Feb,6(2):167-80.
    [91] Gérard A,vander Kammen RA,Janssen H, et al.The Rac activator Tiam1controls efficient T-cell trafficking and route of transendothelial migration[J].Blood, 2009,113(24):6138-47.
    [92] Meller N, Merlot S, Guda C.CZH proteins: a new family of Rho-GEFs[J]. J CellSci, 2005,118(Pt 21):4937-46.
    [93] Durkin ME,Yuan BZ,Zhou X,etal.DLC-1:a Rho GTPase-activating protein andtumour suppressor[J].J Cell mol med,2007:11(5),1185-1207.
    [94] DerMardirossian C,Bokoch GM,GDIs:central regulatory molecules in RhoGTPaseactivation[J].Trends Cell Biol, 2005,15(7),356–363.
    [95] Bokoch GM.Biology of the p21-activated kinases[J].Annu Rev Biochem,2003,72:743–781.
    [96] Mott HR,Owen D,Nietlispach D,et al.Structure of the small G protein Cdc42bound to the GTPase-binding domain of ACK[J].Nature,1999,399(6734): 384-8.
    [97] Spuriing CC,Godman CA,Noonan EJ,et al.HDAC3 over expression and coloncancer cell proliferation and differentiation[J].MolCarcinog,2008,47(2): 137-47.
    [98] Coso OA,Chiariello M,Yu JC,etal.The small GTP-binding proteins Rac1 andCdc42 regulate the activity of the JNK/SAPK signaling Pathway[J]. Cell, 1995:81(7):1137-46.
    [99] Rodal AA, Motola-Barnes RN, Littleton JT.Nervous wreck and Cdc42cooperate to regulate endocytic actin assembly during synaptic growth[J].JNeurosci,2008,28(33):8316-25.
    [100]Luna A,Matas OB, Martínez-Menárguez JA,et al.Regulation of protein transportfrom the Golgi complex to the endoplasmic reticulum by Cdc42 and N-WASP[J]. Mol Biol Cell,2002,13(3):866-79.
    [101]Atwood SX, Chabu C, Penkert RR,et al.Cdc42 acts downstream of Bazooka toregulate neuroblast polarity through Par-6 aPKC[J]. J Cell Sci,2007,120(Pt18):3200-6.
    [102]Fritz G,Brachetti C,BahlmannF,etal.Rho GTPases in human breast tumors:expression and mutation analyses and correlation with clinical parameters[J].BrJ Cancer,2002:87(6):635-44.
    [103]Cuiyan Z,Jie H,Fang Z,et al.Overexpression of RhoE in non-small Cell LungCaneer (NSCLC) is associated with smoking and correlates with DNA copynumber changes[J]. Caneer Biol Ther,2007,6(3):335-42.
    [104]刘赞李卉马文静,等.Cdc42,Rb基因在哈萨克族食管癌中的表达及其意义的研究[J].地方病通报,2008,23(4):1-3.
    [105]郑红,张学光.GTPase在肝癌细胞系中的表达和意义[J].中国临床医学,2004,11(5):775-780.
    [106]Kamai T,Yamanishi T,Shirataki H,et al.overexpression of RhoA,Rac1,andCdc42 GTPases is associated with progression in testicular cancel[J].ClinCaneerRes,2004,10(14):4799-805.
    [107]Frirz G,Just I,Kaina B.Rho GTPases are over-expressed in human tumors[J].IntJ Cancel,1999:81(5):682-7.
    [108]Kamai T,Tsujii T,Aiai K,et al.Significant association of Rho/ROCK Pathwaywith invasion and metastasis of bladder caneer[J].Clin Cancer Res, 2003,9(7):2632-41.
    [109]Lin R, Bagrodia S, Cerione R, Manor D.A novel Cdc42Hs mutant inducescellular transformation[J]. Curr Biol,1997,7(10):794-7.
    [110]Lin R, Cerione RA, Manor D.Specific contributions of the small GTPases Rho,Rac, and Cdc42 to Dbl transformation[J]. J Biol Chem,1999,274(33): 23633-41.
    [111]Tu SS,Wu WJ,Yang W,Nolbant P,Hahn K,Cerione RA.Antiapoptotic Cdc42mutants are potent activators of cellular transformation[J].Biochemistry,2002,41(41):12350-8.
    [112] Wu WJ,Tu S,Cerione RA.Activated Cdc42 sequesters c-Cbl and prevents EGFreceptor degradation[J]. Cell, 2003,114(6):715-25.
    [113] Levkowitz G, Waterman H, Zamir E, Kam Z, Oved S, Langdon WY, Beguinot L,Geiger B, Yarden Y.c-Cbl/Sli-1 regulates endocytic sorting and ubiquitination ofthe epidermal growth factor receptor[J]. Genes Dev, 1998,12(23):3663-74.
    [114]Levkowitz G, Waterman H, Ettenberg SA, Katz M, Tsygankov AY, Alroy I,Lavi S, Iwai K, Reiss Y, Ciechanover A, Lipkowitz S, Yarden Y.Ubiquitinligase activity and tyrosine phosphorylation underlie suppression of growthfactor signaling by c-Cbl/Sli-1[J]. Mol Cell,1999,4(6):1029-40.
    [115]Joazeiro CA, Wing SS, Huang H, Leverson JD, Hunter T, Liu YC.The tyrosinekinase negative regulator c-Cbl as a RING-type, E2-dependent ubiquitin-proteinligase[J]. Science,1999,286(5438):309-12.
    [116]Joazeiro CA, Weissman AM.RING finger proteins: mediators of ubiquitin ligaseactivity[J].Cell,2000,102(5):549-52.
    [117] Hirseh DS,ShenY,WUWJ.Growth and motility inhibition of breast cancer cellsby epidermal growth factor receptor degradation is correlated with inactivationof Cdc42[J].Cancer Res,2006:66(7):3523-30.
    [118] Callow MG,Clairvoyant F,Zhu S,et al.Requirement for PAK4 in the anchorageindependentgrowth of human cancer cell lines[J].J Biol Chem,2002,277(1):550–558.
    [119]Bagrodia S, Cerione RA.Pak to the future[J]. Trends Cell Biol,1999,9(9): 350-5.
    [120]Manser E, Leung T, Salihuddin H, Zhao ZS, Lim LA brain serine/threonineprotein kinase activated by Cdc42 and Rac1[J]. Nature,1994,367(6458):40-6.
    [121]Rudel T, Bokoch GM.Membrane and morphological changes in apoptotic cellsregulated by caspase-mediated activation of PAK2[J].Science,1997,276(5318):1571-4.
    [122]Tzu-Ling Cheng, Marc Symons, Tzuu-Shuh Jou Regulation of anoikis by Cdc42and Rac1[J].Experimental Cell Research,2004,295(2):497-511.
    [123]Qiu RG, Chen J, Kirn D, McCormick F, Symons M. An essential role for Rac inRas transformation[J].Nature,1995,374(6521):457-9.
    [124]Qiu RG, Chen J, McCormick F, Symons M.A role for Rho in Ras transformation[J].Proc Natl Acad Sci USA,1995,92(25):11781-5.
    [125]Qiu RG,Abo A,McCormick F,Symons M.Cdc42 regulatesanchorageindependent growth and is necessary for Ras transformation[J].MolCell Biol, 1997,17(6): 3449-58.
    [126]Olson MF, Ashworth A, Hall A. Science. An essential role for Rho, Rac, andCdc42 GTPases in cell cycle progression through G1.Science, 1995,269(5228):1270-2.
    [127]Spurling CC, Godman CA, Noonan EJ, Rasmussen TP, Rosenberg DW,Giardina C.HDAC3 overexpression and colon cancer cell proliferation anddifferentiation[J].Mol Carcinog,2008 ,47(2):137-47.
    [128]Chou MM, Masuda-Robens JM, Gupta ML). Cdc42 promotes G1 progressionthrough p70 S6 kinase-mediated induction of cyclin E expression[J]. (J BiolChem,2003,278(37):35241-7.
    [129]Tucci MG, Lucarini G, Brancorsini D, Zizzi A, Pugnaloni A, Giacchetti A,Ricotti G, Biagini G.Involvement of E-cadherin, beta-catenin, Cdc42 andCXCR4 in the progression and prognosis of cutaneous melanoma[J]. Br JDermatol,2007 ,157(6):1212-6.
    [130]郑红,张学光GTPase在肝癌细胞系中的表达和意义[J].中国临床医学2004,11(5):779-781.
    [131]Lai SY, Ziober AF, Lee MN, Cohen NA, Falls EM, Ziober BL.Activated Vav2modulates cellular invasion through Rac1 and Cdc42 in oral squamous cellcarcinoma[J].Oral Oncology, 2008, 44(7):683-688.
    [132]Hirsch DS,Shen Y,Wu WJ.Growth and motility inhibition of breast Cancercells by epidernal growth factor degradation is correlated with inactivation ofCdc42[[J].J Cancer Res,2006,66(7):3523-3530.
    [133]Lim KB, Bu W, Goh WI, Koh E, Ong SH, Pawson T, Sudhaharan T, AhmedSThe Cdc42 effector IRSp53 generates filopodia by coupling membraneprotrusion with actin dynamics[J]. J Biol Chem,2008,283(29):20454-72.
    [134]Chauhan BK, Disanza A, Choi SY, Faber SC, Lou M, Beggs HE, Scita G, ZhengY, Lang RA.Cdc42- and IRSp53-dependent contractile filopodia tetherpresumptive lens and retina to coordinate epithelial invagination[J].Development,2009,136(21):3657-67.
    [135]Dan C, Kelly A, Bernard O, Minden A.Cytoskeletal changes regulated by thePAK4 serine/threonine kinase are mediated by LIM kinase 1 and cofilin[J]. BiolChem,2001,276(34):32115-21.
    [136]Wilkinson S, Paterson HF, Marshall CJ.Cdc42-MRCK and Rho-ROCKsignalling cooperate in myosin phosphorylation and cell invasion[J]. Nat CellBiol,2005,7(3):255-61.
    [137]Gaggioli C, Hooper S, Hidalgo-Carcedo C,et al.Fibroblast-led collectiveinvasion of carcinoma cells with differing roles for RhoGTPases in leading andfollowing cells[J].Nat Cell Biol,2007,9(12):1392-400.
    [138]Damiano Serio, Eric Ispanovic and Tara L Haas Cdc42 increases activation ofMMP-2 in endothelial cells[J].The FASEB Journal,2008,22:925-9.
    [139]Eric Ispanovic, Damiano Serio, Tara L,et al.Cdc42 and RhoA have opposingroles in regulating membrane type 1-matrix metalloproteinase localization andmatrix metalloproteinase-2 activation[J].Am J Physiol Cell Physiol, 2008,295(3):C600-C610.
    [140]Saníger ML, Oya R, Macías D, Domínguez JN, Aránega A, Luque F.c-Junkinase mediates expression of VEGF induced at transcriptional level by Rac1and Cdc42Hs but not by RhoA[J]. J Cell Biochem, 2006,98(3):650-60.
    [141]薛妍,毕锋,刘娜等.Rho GTPases对肿瘤血管生成相关分子的作用[J].中国生物化学与分子生物学报,2004,20(5):664-669.
    [142]黄应桂,吴冬梅.胃癌组织中p53、PCNA及c-erbB-2表达与淋巴结转移的关系实用癌症杂志[J].2002,17(3):250-251.
    [143]郑美桦.田晓东.李虎.等.咽喉鳞癌组织中增殖细胞核抗原和核仁组成区相关嗜银蛋白的表达[J].临床耳鼻咽喉科杂志.2002,16(12):653-655.
    [144]Shen LJ,zhang Hx,zhang ZJ,et a1.Detection of HBV, PCNA and GST-πinhepatocellular carcinoma and chronic liver diseases[J].World J GastroenteroI,2003,9(3):459-462.
    [145]Ebelt J,Neid M,Tannapfel A,etal.Prognostic significance of proliferation markersin hepatocellular carcinoma(HCC)[J].J Gastroenterol Hepatol,1998,13(6) :666-670.
    [146]Kato T,Kimura T,Takami N,etal.New prognostic factors associated withlong-term survival IN node-negative breast cancer patients[J].Breast Cancer,1996, 6(4):370-377.
    [147]朱彩荣,曾祥凤,陈亚进,等.MMP-2、TIMP-2和VEGF-C在肝细胞性肝癌中的表达及其与淋巴结转移关系的研究[J].中国病理生理杂志,2005,2l(7):1336-1339.
    [148] Ishii Y,Nakasato Y,Kobayashi S,et a1.A study on angiogenesis-related matrixmetalloproteinase networks in primary hepatocellular carcinoma[J].J Exp ClinCancer Res,2003,22(3):461-470.
    [149]Chao Y,Li C P,Chau G Y,et a1.Prognostic significance of vascular endothelialgrowth factor,basic fibroblast growth factor,and angiogenin in patients withrespectable hepatocellular carcinoma after surgery[J].Ann Surg Oncol,2003, l0(4):355-362.
    [150]崇,郭荣平,石明,等.VEGF与MMP-9在肝癌组织中的表达及其临床意义[J].《癌症》Chinese Journal of Cancer,2006,25(5):599-603.
    [151]Damiano Serio, Eric Ispanovic,Tara L Haas,et al.Cdc42 increases activation ofMMP-2 in endothelial cells[J].The FASEB Journal, 2008;22:925-9.
    [152]Eric Ispanovic, Damiano Serio, and Tara L. Haas.Cdc42 and RhoA haveopposing roles in regulating membrane type 1-matrix metalloproteinaselocalization and matrix metalloproteinase-2 activation[J]. Am J Physiol CellPhysiol,2008,295(3): C600-C610,.
    [153]Ritter M,Ali M Y,Grimm C F,et a1.Immunoregulation of dendritic and T cellsby alpha-fetoprotein in patients with hepatocellular carcinoma[J].J Hepatology,2004,41(6):999-1007.
    [154]Nagao M,Nakajima Y,Kanehiro H,et a1.The impact of interferon gammareceptor expression on the mechanism of escape from host immune surveillancein hepatocellular carcinomar[J].Hepatology,2000,32(3):491-500.
    [155]Elena Dudich,Lidia Semenkova,Igor Dudich,et a1.α-Fetoprotein causesapoptosis in tumor cells via a pathway independent of CD95,TNFRl and TNFR2through activation of caspase-3-like proteases[J].Eur J Biochem,1999,266(3):750-761.
    [156]殷晓煜,吕明德,梁力建等,全身性化学生物治疗晚期原发性肝癌(附20例临床分析)[J].中华肝胆外科杂志,2004,10(10):665-667.
    [157]Chang CS,Huang SM,Lin HH,etal.Different expression of apoptotic proteinsbetween HBV-infected and non-HBV-infected hepatocellular carcinoma[J].Hepatogastroenterology,2007,54(79):2061-8.
    [158]Cooper AB, Wu J, Lu D, Maluccio MA.Is autotaxin (ENPP2) the link betweenhepatitis C and hepatocellular cancer? J Gastrointest Surg,2007,11(12): 1628-34.
    [159]Wu F, Chen Y, Li Y, et al.RNA-interference-mediated Cdc42 silencingdown-regulates phosphorylation of STAT3 and suppresses growth in humanbladder-cancer cells.Biotechnol Appl Biochem,2008,49(Pt 2):121-8.

© 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