卵巢癌组织中NOB1的表达及其在卵巢癌发生中作用机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
卵巢癌是女性生殖系统最常见的恶性肿瘤之一,在美国卵巢癌已居妇科恶性肿瘤死亡原因的首位,我国卵巢癌的发病率也呈逐年上升的趋势。即使实施理想的肿瘤细胞减灭术及铂类为主的化疗,但卵巢癌5年生存率徘徊在25%~30%。近几年来,寻找新的卵巢癌的治疗方法已经成为迫在眉睫的问题。
     RNA干扰又称为转录后基因沉默,通过将与mRNA序列相对应的正义和反义RNA组成的21-23个双链RNA导入靶细胞,从而使细胞内特异性基因的mRNA降解和基因沉默。目前国内外研究学者已常规利用RNAi抑制或下调在卵巢癌发生、发展过程中起关键作用的癌基因、抑癌基因、凋亡相关基因等卵巢癌相关基因的表达,从而达到基因治疗的目的。
     人NOB1基因是近来发现的基因,在白血病等多种肿瘤组织和卵巢癌的多药耐药性细胞中发现有过表达。进一步研究发现,NOB1能够抑制细胞增殖,使细胞周期停留在G1期,调节细胞生长和增殖。提示NOB1在癌症发生和转移过程中可能发挥着重要作用。
     目前国内外尚无卵巢癌组织中NOB1的表达及其二者关系的研究,故而本文研究的目的是验证NOB1基因的过表达可以促进卵巢癌的发生,并通过RNAi抑制NOB1的表达。采用半定量RT- PCR方法检测NOB1在卵巢癌组织、良性肿瘤组织及正常组织中的表达情况。设计并化学合成针对NOB1的siRNA,用慢病毒载体转染法将其转SKOV3、HEY细胞内,通过MTT、细胞克隆形成实验,观察NOB1-siRNA对SKOV3、HEY生长活性和增殖能力的影响;流式细胞术观察NOB1-siRNA对SKOV3、HEY细胞分裂能力的影响。建立SKOV3裸鼠皮下移植瘤模型,体内观察发现NOB1-siRNA对卵巢癌生长的影响。半定量RT- PCR实验结果发现NOB1在卵巢癌组织中的表达明显高于良性肿瘤组织及正常组织,且其表达与肿瘤的大小、细胞分化程度相关,与年龄、组织学分型、肿瘤分期无相关性。MTT、细胞克隆形成实验结果发现转染NOB1-siRNA的SKOV3、HEY细胞生长活性和克隆形成率明显低于对照,流式细胞术实验结果发现转染NOB1-siRNA的SKOV3、HEY细胞阻滞于G0/G1期,细胞分裂减缓;成功建立SKOV3裸鼠皮下移植瘤模型,体内观察实验结果发现注射NOB1-siRNA的小鼠的转移瘤体积及重量均低于对照组。由此得出结论:NOB1在卵巢癌组织中的表达明显上调,同时NOB1-siRNA可有效抑抑制SKOV3、HEY的细胞生长活性、增殖能力并阻滞细胞分裂,同时显著抑制裸鼠的成瘤能力,提示NOB1可能与卵巢癌发生、发展相关,NOB1有望成为卵巢恶性肿瘤基因靶向治疗的新靶点。
Ovarian cancer is one of the most common malignanc tumors in the female genital organs, which was about 4% of the total malignant tumors and 5% of the cancers. The incidence of ovarian cancer is lower than the cervical cancer and endometrial cancer and the mortality mortality rate was rank at the top of gynecologic cancer. With a high degree of malignancy of ovarian cancer and poor prognosis, the ovarian cancer was a serious threat to women's health.
     Since the embryonic development, anatomy organization and endocrine function is more complicated, the clinical early diagnosis of ovarian cancer on the type and nature is very difficult. Most patients diagnosed were found in the advanced phase of ovarian cancer. Moreover, the majority of ovarian cancer had spread to the bilateral uterine attachmentuterus, omentum and pelvic organs by laparotomy. Therefor, both in the diagnosis and treatment effects for ovarian cancer were not good enough. For patients with advanced ovarian cancer, even after surgery and chemotherapy, the majority of them recurred two or three years later. So far, the five-year survival rate of ovarian cancer was only 20% to 30% from the clinical data on the domestic and international statistics, significantly lower than that of cervical cancer and endometrial cancer. Every year, the death number of ovarian cancers was even more than the sum of all other gynecological tumors. In recent years, the clinical treatment of ovarian cancer has made some progress as the surgery and adjuvant therapy improved, but the late survival rate of ovarian patients did not significantly increased.
     Ovarian was exposed to the abdominal cavity, which instantly after Penetrating, ovarian cancer cells can extensively distributed, landing onto the surface of main organs in abdominal cavity, namely diaphragm, liver, intestine, mesentery, omentum and other abdominal viscera which consequently lead to difficulties to lesions complete resection by Surgery. The residual lesions and a large number of sub-clinical lesions mainly rely on chemotherapy after Surgery, but chemotherapy drugs can not be completely eliminated the ovarian cancer which was moderate sensitive to the chemotherapy drugs. The remaining tumor cells exposed to chemotherapy drugs got multi-drug resistance for long period of time, which made the treatment for ovarian cancer more difficult. Therefore, to search for a more effective treatment method and alleviatethe side effects was a intractable problem for researchers in gynecologic oncology.
     Tumor-specific molecular targeted therapy provides a new way for the treatment of ovarian cancer. It greatly enhanced the efficiency and patient survival for its special role in tumor cells and less toxicity to normal tissue. Those drugs targeted to the tumors were applied to the clinic therapy, including epidermal growth factor receptor (EGFR) inhibitors, monoclonal antibody for specific cell marker, drugs with genetics signs and against certain cancer genes and cells, anti-angiogenesis drugs. These drugs have brought a good therapeutic effect in cancer therap, however, there are still some limitations for their widely use. It was mainly due to that different tumors have different genetic backgrounds and biological properties, which affected the scope of application of molecular targeted drugs. Thus, in the treatment of ovarian cancer, it shouldn’t simply rely on migration from other tumors. It is of important practical significance to independent research cancer-causing genes and find suitable molecular target of the ovarian cancer.
     Oncogenes of ovarian cancer involve in the cell proliferation, cell cycle regulation, apoptosis signal, invasion, metastasis and other aspects. A new gene NOB1 (NIN1/RPN12 binding protein 1 homolog) was cloned in 2005, which is a RNA-binding protein. This type of protein is often associated with the occurrence of malignant tumors. Structurally, NOB1 is a component of 26S proteasome which is a protelytic complex of eukaryotes. It comprises two major complexes: the catalytic 20S proteasome and the 19S regulatory particle, which degrades proteins that have been targeted for destruction by the ubiquitination (Ub) pathway. It has been found that an increasing number of proteins were included in the subunits of 26S protease. Among them, yeast Nob1p (Nin one binding protein) was first separated by the yeast two-hybrid screening method using Rpn12, a subunit of the 19S regulatory particle. Nob1p played an important role in the function of protease and RNA metabolism process.
     In the past studies, Nob1p was found to be associated with the proteasome in the cells with exponential growth period, and is degraded during the transition to the stationary phase. Nob1p can also form a complex with nucleoprotein Pno1, which is required for nuclear transfer of the 20S proteasome. Nob1p is a molecular chaperone combined with 20S proteasome and 19S regulatory particle, as well as facilitated to the maturation of 26S proteasome.
     Yeast Nob1p equally involved in the process of ribosome RNA (rRNA) development and assembling and rRNA is one of the most important structural and functional components in the ribosome assembly process. Nob1p can be co-precipitated with 20S pre-rRNA and pre-40S. The precipitated 20S pre-rRNA is dimethylated at the 3 'region. Depletion of Nob1p will damage the synthesis of 18S rRNA, accompanied by an accumulation of 20S pre-rRNA at a high level.
     In humans, Nob1p homologous gene NOB1 and its encoding product were first cloned and analyzed from human kidney cDNA library in 2005. The human NOB1 gene consisted of nine exons and eight introns and located at 16q22.3. The assembled cDNA of NOB1 was 1749 bp in length, containing an ORF of 1239 bp. The ORF encoded a protein with a predicted molecular weight of 50 kDa through the western experiments. While the results of RT-PCR showed that the expression of NOB1 was mainly detected in adult human lung, liver, and spleen. In vitro cultured mammalian cells, the expression of NOB1 protein mainly distributed in nucleus, and some can be detected in cytoplasm. This information provides valuable reference for further study of NOB1 gene function. However, up to date, there was no tumor-related report about NOB1 gene. The role of the NOB1 gene in ovarian cancer occurrence and development process is still unknown. The function of NOB1 and the correlation with the prognosis of ovarian cancer need further validation, which will undoubtedly contribute to the clinic application of NOB1 and the treatment of ovarian cancer.
     In the present study, the expression of NOB1 in ovarian cancer and its relevance to the clinical pathological feature was explored, which was not reported previously. The inhibition effect of NOB1 on the cell proliferation and cell cycle in vitro and the tumor-forming ability in nude mice was investigated. NOB1 is expected to become the new molecule targeted to ovarian tumor in gene therapy. However, the function and application of NOB1 gene in human tumor therapy need further investigation. Together with our results, it provides new ideas and references for the functional mechanism of NOB1 and its therapeutic applications for malignant tumors, especially ovarian cancer.
     Objective: To explore the role of NOB1 gene in the ovarian cancer and its relationship with clinical pathological characteristics, the expression of NOB1 gene in the ovarian cancer was investigated. To elucidate the effect of NOB1 gene on the cell proliferation and cell cycle, RNAi technology was applied to silence NOB1 gene in ovarian cancer cell lines SKOV3 and HEY. Moreover, to further confirm the effect of decreased NOB1 levels on tumorigenesis in vivo, tumorigenicity experiments were performed in the ovarian cancer cells following lentivirus infection.
     Methods:
     In the present study, we tested the expression of NOB1 in 50 ovarian tumor samples and 50 non-ovarian tumor samples by quantitative PCR, and explored the correlation between the expression of NOB1 and the ovarian clinic-pathologic feature.
     We designed several sequences of siRNA which targeted NOB1 gene and screened the most effective silence site for RNAi, and pack it with lentiviral particles.
     Using the packed lentiviral vector to infect human ovarian tumor SKOV3 and HEY cell line, MTT assay was performed to evaluate the cell proliferation. DNA synthesis in proliferating cells was determined by BrdU incorporation assay. Colony formation ability and cell cycle change was also investigated after the NOB1 gene inhibition.
     Nude mice transplanted human ovarian cancer model was established. The weight of the tumor weight and size were measuredafter the targeted shRNA lentiviral particles and control lentiviral particles were injected into the mice. The inhibition rate was used as an index to calculate the effect NOB1 gene silence on the ovarian cancer tumorigenesis capacity in vivo.
     Results:
     The high expression of NOB1 gene in ovarian cancer was significant correlated with the tumor size and the cell differentiation, while no correlated with the age of patients, tumor histological types and staging from the quantitative PCR and statistical analysis results.
     The recombinant lentiviral vector of shRNA targeted NOB1 was successfully constructed and was infected into HEK293 cells together with an over-expression vector. Western blot results proved that the 3rd siRNA target sequence was the most effective on NOB1 silencing. The NOB1 mRNA and protein expression were significantly inhibited in two human ovarian cancer cell lines, SKOV3 and HEY cells, when infected by shRNA lentiviral vector.
     It was showed that the cell proliferation, DNA replication and colony formation ability were inhibited. Both cells were significantly blocked at G1 phase following the flow cytometric analysis. In the xenografts assays in nude mice, the tumor size and weight was measured. It was revealed that the tumorigenic capacity was significantly inhibited following the knockdown of NOB1 in ovarian cancer cells.
     Conclusions: The high expression of NOB1 in ovarian cancer was relevant to the clinical pathological feature, which may be a molecular incentive of malignant tumor formation. The NOB1 gene expression in ovarian cancer cells was effectively down-regulated by lentivirus-mediated RNA interference. Inhibition the NOB1 expression can inhibit the cell proliferation in vitro and the tumor-forming ability in nude mice, and affect the cell cycle. NOB1 is expected to become the new molecule targeted to tumor in gene therapy.
引文
[1] Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics [J]. CA Cancer J Clin, 2009, 59: 225-249.
    [2] WHO. Cancer epidemiology databases. France: International Agency for Research on Cancer; 2001 (http://www.iarc.fr)
    [3] Jin F, Devesa SS, Chow WH, Zheng W, Ji BT, Fraumeni Jr JF, et al. Cancer incidence trends in urban Shanghai, 1972-1994: an update [J]. Int J Cancer, 1999, 83:435-440.
    [4] orkka K, Blomqvist C, Rissanen P, et al. Salvage therapies in women who fail To respond to first-line treatmen twith fluorouraeil, epirubiein, and cyclophosphamide fo radvanced breast cancer [J]. J Clin Oneol, 1994, 12:1639-1647.
    [5] Kramm CM, Niehuse T, Rainov NG. Experimental strategies for combined suicide and immune cancer gene therapy. An overview. Methods Mol Biol, 2003, 215(6):137-152.
    [6] Fire A, Xu S, Montgomery MK, et al. Potent and specific interference by double- atranded RNA in Caenorhabditis elegans [J]. Nature, 1998, 391(6669): 806-811.
    [7] Nieth C, Priebsch A, Stege A, et al. Modulation of the classical multidrug resistance (MDR) phenotype by RNA interfenrence (RNAi) [J]. FEBS Lett, 2003, 545(2-3): 144-150.
    [8] Zhao LJ, Jian H, Zhu H. Specific gene inhibition by adenovirus-mediated expression of small interfering RNA [J]. Gene, 2003, 316:137-141.
    [9]Urban-Klein B, Werth S, Abuharbeid S, et al. RNAi-medi-ated gene-targeting through systemic application ofpolyethylenimine (PEI)-complexed siRNA in vivo. GeneTher, 2005, 12(5): 461 -466.
    [10] Choudhurg A,Charo J,Parapuram SK,et al. Small interfering RNA (siRNA)inhibits the expression of the Her2/neu gene,upregulated HLA class I and induces apoptosis of Her2/neu positive tumor eell lines[J].Int J Cancer,2004,108(1):71-77.
    [11] Menendez JA,Velon L,Mehmi I,et a1.Inhibition of fatty acid synthase(FAS)suppresses HER2/nue (erbB2) oncogene overexpression in cancer cells[J]. Proc Natl Acad SciUSA,2004,101(29):10715-10720.
    [12] Noske A, Kaszubiak A, Weichert W, et al.Specific inhibition of AKT2 by RNA interference results in reduction of ovarian cancer cell proliferation: increased expression of AKT in advanced ovarian cancer[J]. Cancer Lett , 2007,246(1-2):190-200.
    [13] Numnum TM, Makhija S, Lu B, et al.Improved anti-tumor therapy based upon infectivity-enhanced adenoviral delivery of RNA interference in ovarian carcinoma cell lines [J].Gynecol Oncol,2008,108(1):34-41.
    [14] He X, Pool M, Darcy KM, et al.Knockdown of polypyrimidine tractbinding protein suppresses ovarian tumor cell growth and invasiveness in vitro[J]. Oncogene,2007,26(34):4961-4968.
    [15]赖晓红,翁光华. VEGF的RNAi对卵巢癌细胞SKOV3生物学活性的影响作用[J].中国医疗前沿,2008,3(22):23-25.
    [16]陈诗书,戴冰冰.基因治疗的研究现状和评价[J].中华肿瘤,2002,24:313 -315.
    [17]Peixin Dong, Kazuki Nabeshima, Naoko Nishimura, et al.Overexpression diffuse expression pattern of IQGAP1 at invasion fronts are independent prognostic parameters in ovarian carcinomas. [J].Cancer Letter. 2006,243:120-127.
    [18] Dong Pei-Xin, Jia Nan, Xu Zhu-Jie, et al.Silencing of QGAP1 by shRNA inhibits the invasion of ovarian carcinoma HO-8910PM cells in vitro [J]. Journal of Experimental & Clinical Cancer Research, 2008, 27(1):77-82.
    [19] Maldonado E, Reinberg D. News on initiation and elongation of transcription by RNA polymerase II [J]. Curr Opin Cell Biol, 1995, 7: 352-361.
    [20] Y. Zhang, J. Ni, G. Zhou, et al. ,Cloning expression and characterization of the human NOB1 gene [J]. Mol Biol Rep, 2005, 32(3):185-189.
    [21] Ferrell K , Wilkinson CRM, Dubiel W, et al. Regulatory subunit interactions of the26S proteasome, a complex problem [J]. Trends Biochem Sci, 2000, 25: 83-88.
    [22] Zachariae W, Nasmyth K. Whose end is destruction: cell division and the anaphase-promoting complex [J]. Gene Dev, 1999, 19: 2039-2058.
    [23] Fujimuro M, Takada H, Saeki Y, et al. Growth-dependent change of the 26S proteasome in budding yeast [J]. Biochem Biophy Res Com,1998, 251: 818-823.
    [24] Vierstra RD. The ubiquitin/26S proteasome pathway, the complex last chapter in the life of many plant proteins [J]. Trends Plant Sci, 2003, 8: 135-142.
    [25] Yoshiko T, Nobuyuki T, Keiji T, et al. Nob1p, a new essential protein, associates with the 26S proteasome of growing Saccharomyces cerevisiae cells [J]. Gene, 2000, 243:37-45.
    [26] Tone Y, Tohe A. Nob1p is required for biogenesis of the 26S proteasome and degraded upon its maturation in Saccharomyces cerevisiae [J]. Gene Dev, 2002, 6: 3142-3157.
    [27] Gerbi SA, Borovjagin AV, Lange TS. The nucleolus: a site of ribonucleoprotein maturation [J]. Curr Opin Cell Biol, 2003, 15: 318-325.
    [28] Fatica A , Oeffinger M, Dlakic M, et al. Nob1p is required for cleavage of the 3 ' end of 18S rRNA [J]. Mol Cell Biol, 2003, 23: 1798-1807.
    [29] Alessandro F, David T, MensurD. PIN domain of Nob1p is required for D-site cleavage in 20S pre-rRNA [J]. RNA, 2004, 10:1698-1701.
    [30] Orphanides, G., T. Lagrange, and D. Reinberg. The general transcription factors of RNA polymerase II [J]. Genes Dev, 1996, 10: 2657-2683.
    [31] Ghosh M, Elsby LM, Mal TK, et al. Probing Zn2+-binding effects on the zinc-ribbon domain of human general transcription factor TFIIB [J]. Biochem, 2004, 378: 317-324.
    [32] Hong L, Han Y, Han Y, et al.Mechanisms of growth arrest by zinc ribbon domain-containing 1 (ZNRD1) in gastric cancer cells [J]. Carcinogenesis, 2007, 28(8):1622-1628.
    [33] Qian X, Jeon C, Yoon H, et al. Structure of a new nucleic-acid-binding motif in eukaryotic transcriptional elongation factor TFIIS. Nature, 1993, 365: 277-279.
    [34] Hong L, Zhao Y, Wang J, et al. Reversal of multidrug resistance of adriamycin- resistant gastric adenocarcinoma cells through the up-regulation of DARPP-32 [J]. Dig Dis Sci, 2008, 53(1):101-107.
    [35] Hong L, Han Y, Shi R, et al. ZNRD1 gene suppresses cell proliferation through cell cycle arrest in G1 phase [J]. Cancer Biol Ther, 2005, 4(1): 60-64.
    [36] Wang B, Jones DN, Kaine BP, et al. High-resolution structure of an archaeal zinc ribbon defines a general architectural motif in eukaryotic RNA polymerases [J]. Structure, 1998, 6: 555-569.
    [37] Han Y, Hong L, Qiu J, et al. Preparation and characterization of a novel monoclonal antibody specific to human NOB1 protein, Hybridoma [J]. Larchmt, 2008, 27 (3): 187-190.
    [38] Hong L, Wang J, Han Y, et al. Reversal of multidrug resistance of vincristine- resistant gastric adenocarcinoma cells through up-regulation of DARPP-32 [J]. Cell Biol Int, 2007, 31(9):1010-1015.
    [39] Hong L, Zhang Y, Liu N, et al. Suppression of the cell proliferation in stomach cancer cells by the ZNRD1 gene [J]. Biochem Biophy Res Com, 2004, 321(3): 611-616.
    [40] Tubon TC, Tansey WP, Herr W. A Nonconserved Surface of the TFIIB Zinc Ribbon Domain Plays a Direct Role in RNA Polymerase II Recruitment [J]. Mol Cell Biol, 2004, 24: 2863-2874.
    [41] Kira S, Makarova L, Aravind MYG, et al. Comparative Genomics of the Archaea (Euryarchaeota): Evolution of Conserved Protein Families, the Stable Core, and the Variable Shell [J]. Genome Res, 1999, 9:608-628.
    [42] Han Y, Hong L, Zhong C, et al. The expression of NOB1 in spiral ganglion cells of guinea pig [J]. Int J Pediatr Otorhi, 2009, 73: 315-319.
    [43] Zhao Y, Hong L, Wang R, et al. Expression and prognostic value of ZNRD1 in esophageal squamous cell carcinoma [J]. Dig Dis Sci , 2009, 54:586-592.
    [44] Hong L, Piao Y, Han Y, et al. Zinc ribbon domain-containing 1 (ZNRD1) mediates multidrug resistance of leukemia cells through regulation of P-glycoprotein andBcl-2 [J]. Mol Cancer Ther, 2005, 4 (12): 1936-1942.
    [45] Hong L, Qiao T, Han Y, et al. ZNRD1 mediates resistance of gastric cancer cells to methotrexate by regulation of IMPDH2 and Bcl-2 [J]. Biochem Cell Biol, 2006, 84 (2): 199-206.
    [46] Hong L, Ning X, Shi Y, et al. Reversal of multidrug resistance of gastric cancer cells by down-regulation of ZNRD1 with ZNRD1 siRNA [J]. Br J Biomed Sci, 2004, 61 (4): 206-210.
    [47] Hong L, Han Y, Shi R, et al. ZNRD1 gene suppresses cell proliferation through cell cycle arrest in G1 phase [J]. Cancer Biol Ther, 2005, 4: 60-64.
    [48] Hammond SM, Bernstein E, Beach D, et al. An RNA-directed nuclease mediates post-transcriptional gene silencing in drosophela cells [J]. Nature, 2000, 404: 293-296.
    [49] Cogonic MG. Post-transcriptional gene silencing aculss kingdoms. Curr Opinion Genet Dev, 2000, 10: 638-643.
    [50] Guo S, Kemphues KJ. par-1, a gene required for establishing polarity in C.elehans embryos,encodes a putative Ser/Thr kinase that is asymmetrically distributed [J]. Cell, 1995, 81: 611-620.
    [51] Elbashir SM, Harborth J, Weber K, et al. Analysis of gene function in somatic mammalian cells using small interfering RNAs [J]. Methods, 2002, 26(2):100-213.
    [52] Schiffelers RM, Ansari A, Xu J, et al. Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle [J]. Nucleic Acids Res, 2004, 32(19):e149.
    [53] Elbashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-ntcleotide RNAs mediate RNA interference in cultured mammalian cells [J]. Nature, 2001, 411(6 836): 494-498.
    [54] Sanchez A, Newmark PA. Double-stranded RNA specifically disrupts gene expression during planarian regeneration [J]. Proc Natl Acad Sci USA, 1999, 96(9): 5049-5054.
    [55] Chuang CF, Meyerowitz EM. Specific herltable genetic interference by double-strand RNA in Arabidopsis thallane [J]. Proc Natl Acid Sci USA, 2000, 97: 4985-4900.
    [56] Hammond SM, Boettcher S, Caudy AA, et al. Argonaute 2, a link between genetic and biochemical analyses of RNAi. Science, 2001, 293(5532):1146-1150.
    [57] Sijen T, Fleenor J, Simmer F, et al. On the role of RNA amplification in dsRNA-triggered gene silencing [J]. Cell, 2001,107(4): 465-476.
    [58] Nykanen A, Haley B, Zamore PD. ATP requirements and small interfering RNA structure in the RNA interference pathway [J]. Cell, 2001,107(3): 309-321.
    [59] Elbashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells [J]. Nature, 2001, 411(6836): 494-498.
    [60] Chen T, Wang Z, Wang R, et al. Polyethylenimine-DNA solid particles for gene delivery [J]. J Drug Target, 2007, 15 (10): 714- 720.
    [61]张明,邵宁生. RNA阻抑和基因沉默[J].军事医学科学院院刊,2002,26(1): 61-65.
    [62] Sabine B. Antisense-RNA regulation and RNA inter-ference [J]. Biochemica et Biophysica Acta, 2002, 1575:15-25.
    [63] Tang G, Reinhart BJ, Bartel DP, et al. A biochemical framework for RNA silencing in plants [J] .Genes Development, 2003, 17(1): 49- 63.
    [64] Samuel CE. Knockdown by RNAi- proceed with caution [J]. Nature Biotechnol, 2004, 22(3): 280- 282.
    [65] Morel JB, Mourrain P, Beclin C, et al. DNA methylation and chromatin structure affect transcriptional and posttranscriptional transgene silencing in arabidopsis [J]. Curr Biol, 2000, 10(24): 1591- 1594.
    [66] Philltp DZ, Thomas T. RNAi: double-stranded RNA directs the ATP- dependent cleavage of mRNA at 21 to 23 nucleotide intervals [J] .Cell, 2000, 101(31) :25- 33.
    [67] Fraster AG, Kamath RS, Zipperlen P, et al. Functional genomic analysis of C. elegans chromosome I by systematic RNA interference [J]. Nature, 2000, 408(6810): 325-330.
    [68] Gonczy PE, Cheverri C, Oegema K, et al. Functional genomic analysis of cell division in C.elegans using RNAi of genes on chromosomeⅢ[J]. Nature, 2000, 6810: 331-336.
    [69] Jacque JM, Triques K, Stevenson M, et al. Modulation of HIV-1 replication by RNA interference [J]. Nature, 2002, 418 (6896): 435-438.
    [70] Novina CD, MurrayMF, Dykxhoorn DM, et al. siRNA-directed inhibition of HIV-1 infection [J]. Nat Med, 2002, 8 (7): 681-686.
    [71] Wannenes F, Ciafre SA, Niola F, et al. Vector-based RNA interference against vascular endothelial growth factor—a significantly limits vascularization and growth of prostate cancer in vivo [J]. Cancer Gene Ther, 2005, 12 (12): 926-934.
    [72] Ray KM, PaulA. RNA interference: from gene silencing to gene-specific therapeutics [J]. Pharmacol Ther, 2005, 107 (2): 222-239.
    [73] Yang Z, Cloud A, Hughes D, et al. Stable inhibition of human thymidy late synthase exp ression following retroviral introduction of an siRNA gene [J]. Cancer Gene Ther, 2006, 13 (1): 107-114.
    [74] Aloy MT, Hadchity E, Bionda C, et al. Protective role of Hsp27 protein against gamma radiation-induced apoptosis and radiosensitization effects of Hsp27 gene silencing in different human tumor cells [J]. Int J Radiat Oncol Biol Phys, 2008, 70 (2): 543-553.
    [75] Brake OT, Hooft K, Liu YP, et al. Lentiviral vector design for multiple shRNA expression and durable H IV-1 inhibition [J]. Mol Ther, 2008,16 (3) : 557-564.
    [76] Ketting RF. A genetic link between co- suppression and RNA interference in celegans [J] .Nature, 2000, 404( 6778) :296- 298.
    [77] Kim K, Zang R, Choi SC, Ryu SY, Kim JW. Current status of gynecological cancer in China [J]. J Gynecol Oncol, 2009, 20(2):72-76.
    [78] Holsehneider CH, Berek JS. Ovarian cancer: epidemiology . Biology, and prognostic faclom [J]. Seminars in Surgieal Oncology, 2000, 19: 3-10.
    [79] Markman M. Intraperitoneal chemotherapy as primary treatment of advanced ovarian cancer: efficacy, toxicity, and future directions [J]. Rev Recent Clin Trials,2007, 2(3):169-173.
    [80] Reibenwein J, Krainer M. Targeting signaling pathways in ovarian cancer [J]. Expert Opin Ther Targets, 2008, 12(3):353-365.
    [81] Tolmachev V. Imaging of HER-2 overexpression in tumors for guiding therapy [J]. Curr Pharm Des, 2008, 14(28): 2999-3019.
    [82] Lafky JM, Wilken JA, Baron AT, et al. Clinical implications of the ErbB/epidermal growth factor (EGF) receptor family and its ligands in ovarian cancer [J]. Biochim Biophys Acta, 2008, 1785(2):232-265.
    [83] Kamath S, Buolamwini JK. Targeting EGFR and HER-2 receptor tyrosine kinases for cancer drug discovery and development [J]. Med Res Rev, 2006, 26(5): 569-594.
    [84] Ortega E, Marti RM, Yeramian A, et al. Targeted therapies in gynecologic cancers and melanoma [J]. Semin Diagn Pathol, 2008, 25(4):262-273.
    [85] Corney DC, Flesken-Nikitin A, Choi J, et al. Role of p53 and Rb in ovarian cancer [J]. Adv Exp Med Biol, 2008, 622: 99-117.
    [86] Tachibana M, Watanabe J, Matsushima Y, et al. Independence of the prognostic value of tumor suppressor protein expression in ovarian adenocarcinomas: A multivariate analysis of expression of p53, retinoblastoma, and related proteins [J]. Int J Gynecol Cancer, 2003, 13(5):598-606.
    [87] Shaw PA, Rouzbahman M, Pizer ES, et al. Candidate serous cancer precursors in fallopian tube epithelium of BRCA1/2 mutation carriers [J]. Mod Pathol, 2009, 22(9):1133-1138.
    [88] Zagorianakou N, Stefanou D, Makrydimas G, et al. CD44s expression, in benign, borderline and malignant tumors of ovarian surface epithelium. Correlation with p53, steroid receptor status, proliferative indices (PCNA, MIB1) and survival [J]. Anticancer Res, 2004, 24(3a):1665-1670.
    [89] Talvensaari-Mattila A, Santala M, Soini Y, Turpeenniemi-Hujanen T. Prognostic value of matrix metalloproteinase-2 (MMP-2) expression in endometrial endometrioid adenocarcinoma [J]. Anticancer Res, 2005, 25(6B):4101-4115.
    [90] Henic E, Sixt M, Hansson S, H?yer-Hansen G, Casslén B. EGF-stimulated migration in ovarian cancer cells is associated with decreased internalization, increased surface expression, and increased shedding of the urokinase plasminogen activator receptor [J]. Gynecol Oncol, 2006, 101(1):28-39.
    [91] Ferrell K, Wilkinson CR, Dubiel W, Gordon C. Regulatory subunit interactions of the 26S proteasome, a complex problem [J]. Trends Biochem Sci, 2000, 25: 83-88.
    [92] Zachariae W, Nasmyth K. Whose end is destruction: cell division and the anaphase-promoting complex [J]. Gene Dev, 1999, 19: 2039-2058.
    [93] Fujimuro M, Takada H, Saeki Y, et al. Evidence for an Interaction between Ubiquitin-Conjugating Enzymes and the 26S Proteasome [J]. Biochem Biophys Res Com, 1998, 251: 818-823.
    [94] Vierstra RD. The ubiquitin/26S proteasome pathway, the complex last chapter in the life of many plant proteins [J]. Trends Plant Sci, 2003, 8:135-142.
    [95] Tone Y, Tanahashi N, Tanaka K, Fujimuro M, Yokosawa H & Toh-e A. Nob1p, a new essential protein, associates with the 26S proteasome of growing saccharomyces cerevisiae cells [J]. Gene, 2000, 243:37-45.
    [96] Tone Y, Toh-E A. Nob1p is required for biogenesis of the 26S proteasome and degraded upon its maturation in Saccharomyces cerevisiae [J]. Gene Dev, 2002, 16: 3142-3157.
    [97] Gerbi SA, Borovjagin AV, Lange TS. The nucleolus: a site of ribonucleoprotein maturation [J]. Curr Opin Cell Biol, 2003, 15:318-325.
    [98] Fatica A, Oeffinger M, Dlakic? M, et al. Nob1p Is Required for Cleavage of the 3’End of 18S rRNA [J]. Mol Cell Biol, 2003, 23:1798-1807.
    [99] Zhang Y, Ni J, Zhou GJ, et al. Cloning, expression and characterization of the human NOB1 gene [J]. Mol Biol Rep, 2005, 32:185-189.
    [100] Canevari S, Gariboldi M, Reid JF, et al. Molecular predictors of response and outcome in ovarian cancer [J]. Crit Rev Oncol Hemat,2005, 60(1): 19-37.
    [101]Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2-△△C T Method[J]. Methods, 2001, 25:402-408.
    [102]Duan ZF, Ames RY, Ryan M, et al. CDDO-Me, a synthetic triterpenoid, inhibits expression of IL-6 and Stat3 phosphorylation in multi-drug resistant ovarian cancer cells [J]. Cancer Chemoth Pharm, 2009, 63(4): 681-689.
    [103]Odening KE, Li W, Rutz R, et al. Enhanced complement resistance in drug-selected P-glycoprotein expressing multi-drug-resistant ovarian carcinoma cells [J]. Clin Exp Immunol, 2009, 155(2): 239-248.
    [104] Hong L, Qiao T, Han Y, et al. ZNRD1 mediates resistance of gastric cancer cells to methotrexate by regulation of IMPDH2 and Bcl-2 [J]. Biochem Cell Biol, 2006, 84 (2): 199-206.
    [105]Ghataorhe P, Kurian AW, Pickart A, et al. A carrier of both MEN1 and BRCA2 mutations: case report and review of the litera2 ture [ J ]. Cancer Genet Cytogenet, 2007, 179 (2) : 89 - 92.
    [106]Schmutzler RK. Molecular genetics and clinicsof hereditary breast cancer [J]. Verh Dtsch Ges Pathol,2005,89:25 - 34.
    [107]Marchini S, Marabese M, Marrazzo E, et al. Deltan p53 expresion is associated with poor survival in ovarian cancer [J ]. Ann Oncol,2007,12(5):367 - 369.
    [108]Gatcliffe TA, MonkBJ, Planutis K, et al. Wnt signaling in ovarian tumorigenesis [J]. Int J Gynecol Cancer,2007,6 (3): 265 -269.
    [109]Wu HJ,Wu HT,WengDH,et al. Reversalof drug resistance in human ovarian cancer cells by wild - type PTEN gene and its mechanisms [J ]. Zhonghua Fu Chan Ke Za Zhi, 2007, 42 (9):612 - 616.
    [110]Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer incidence and mortality in Europe in 2008 [J]. Eur J Cancer, 2010, 46(4): 765-781.
    [111]Wu RH ,Cheng TL ,Lo SR ,et al. A tightly regulated and reversibly inducible siRNA expression system for conditional RNAi2mediated gene silencing in mammalian cells[J ] . J Gene Med ,2007 ,9 (7) :620.
    [112]Gao J , Su L ,Qin R ,et al. Transfection of antisense oligodeoxynucleotide inhibit s heparanase gene expression and invasive ability of human pancreatic cancer cell invitro[J ] . J Huazhong Univ Sci Technolog Med Sci ,2006 ,26 (1) :72.
    [113]Cesarone G, Garofalo C ,Abrams MT ,et al. RNAi-mediated silencing of insulin receptor subst rate 1 ( IRS21) enhances tamoxifen- nduced cell death in MCF27 breast cancer cells[J ] . J Cell Biochem ,2006 ,98 (2) :440.
    [114]Vandersickel V, Mancini M, Marras E, et al. Lentivirus-mediated RNA interference of Ku70 to enhance radiosensitivity of human mammary epithelial cells [J]. Int J Radiat Biol, 2010, 86(2): 114-124.
    [115]Sankpal NV, Fleming TP, Gillanders, WE. Dual Expression Lentiviral Vectors for Concurrent RNA Interference and Rescue [J]. J Surg Res, 2009, 156(1): 50-56.
    [116]Greenlee RT, Hill-Harmon MB, Murray T, Thum M. Cancer statistics, 2001.CA Cancer J Clin 2001:51:15-36.
    [117]Oehler VG, Yeung KY, Choi YE. The derivation of diagnostic markers of chronic myeloid leukemia progression from microarray data [J]. Blood, 2009, 114(15): 3292-3298.
    [118]Bai Y, Deng HX, Yang Y, et al. VEGF-Targeted Short Hairpin RNA Inhibits Intraperitoneal Ovarian Cancer Growth in Nude Mice [J]. Oncology, 2005,77(6): 385-394.
    [119]Wei ZT, Zhang X, Wang XY. PDCD4 inhibits the malignant phenotype of ovarian cancer cells [J]. Cancer Sci, 2009, 100(8): 1408-1413.
    [120]Racz Z, Hamar P. Can siRNA technology provide the tools for gene therapy of the future? [J]. Curr Med Chem, 2006, 13(19): 2299-2307.
    [121]Graat HCA, van Beusechem VW, Schagen FHE, et al. Intravenous administration of the conditionally replicative adenovirus Ad5-Delta 24RGD induces regression of osteosarcoma lung metastases [J]. Mol Cancer, 2008, 7:9.
    [122]Ito T, Okada T, Miyashita H, et al. Prevention of monocrotaline-induced pulmonary arterial hypertension by an intramuscular injection of an adeno- associated virus vector expressing prostacyclin synthase [J]. Circulation, 2006, 114(18):82-82
    [123]Sheets RL, Stein J, Manetz TS, et al. Biodistribution of DNA plasmid vaccinesagainst HIV-1, Ebola, severe acute respiratory syndrome, or West Nile virus is similar, without integration, despite differing plasmid backbones or gene inserts [J]. Toxicol Sci, 2006, 91(2): 610-619.
    [124]Ganesh S, Gonzalez-Edick M, Gibbons D, et al. Evaluation of Biodistribution of a Fiber-Chimeric, Conditionally Replication-Competent (Oncolytic) Adenovirus in CD46 Receptor Transgenic Mice [J]. Human Gene Ther, 2009, 20 (10): 1201-1213.
    [125]Okada Y, Okada N, Mizuguchi H, et al. Transcriptional targeting of RGD fiber-mutant adenovirus vectors can improve the safety of suicide gene therapy for murine melanoma [J]. Cancer Gene Ther, 2005, 12(7): 608-616.

© 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