MicroRNA-520b调节乳腺癌细胞免疫逃逸作用和分子机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
MicroRNAs (miRNAs)是一类非编码蛋白的短序列RNA片段,长度大约为21-23个核苷酸。研究表明miRNAs参与了肿瘤发生、发展和转移等诸多过程,最近研究发现microRNA-520b(miR-520b)与免疫调节有关。CD46, CD55和CD59是一簇膜结合补体调节蛋白(mCRPs),在补体级联反应中可以通过抑制C5转化酶阻止膜攻击复合物(MAC)在肿瘤细胞表面的形成,抑制了补体介导的细胞毒(complement-dependent cytotoxicity, CDC)作用,保护肿瘤细胞免受补体攻击。但是,调节CRP的分子机制尚不清楚,miR-520b是否参与了补体介导的肿瘤免疫逃逸也未见报道。为了阐明乳腺癌细胞中调节mCRPs的分子机制,我们研究了miR-520b或miR-520e在乳腺癌细胞中参与免疫调节的作用,研究应用不同转移能力的乳腺癌细胞系MCF-7和LM-MCF-7(获国家发明专利,ZL.0310107316)探讨了miR-520b与CRP的关系及其分子调控机制,主要研究内容如下:
     1. miR-520b和miR-520e靶向CD46影响补体介导的细胞毒作用及其分子机制研究
     本实验室前期通过对肝癌细胞与正常肝细胞的microRNA芯片分析发现,microRNA-520b在肝癌细胞中下调。为了进一步研究microRNA-520b及与其高度同源的miR-520e在乳腺癌细胞中的作用及其与补体介导的肿瘤免疫逃逸的关系,我们首先应用实时定量反转录聚合酶链式反应(quantitative RT-PCR)的方法检测分析了miR-520b和miR-520e在正常的乳腺细胞HBL-100以及三株乳腺癌细胞系MCF-7, LM-MCF-7和MDA-MB-231细胞系中的表达水平,实验结果表明,与正常的乳腺细胞HBL-100相比,乳腺癌细胞MCF-7, LM-MCF-7和MDA-MB-231细胞系中miR-520b和miR-520e的基因表达均下调。我们进一步在8例正常乳腺组织和7例乳腺癌组织临床标本中比较了miR-520b的表达水平,发现miR-520b在乳腺癌组织中的表达水平与正常乳腺组织中的表达水平相比显著下调,并发现乳腺癌细胞系抵抗CDC的能力均高于正常乳腺细胞。为了进一步研究miR-520b或miR-520e在乳腺癌细胞中对补体依赖的细胞毒作用的影响,我们外源转染了合成的miR-520b或miR-520e的成熟体(mimics),以及miR-520b或miR-520e的抑制子(anti-mimics), CDC实验表明,在MCF-7或MDA-MB-231乳腺癌细胞系中过表达miR-520b或miR-520e均可增强CDC的作用,而在干扰了miR-520b或miR-520e的乳腺癌细胞中则抑制了其对CDC的敏感性。综上结果表明,miR-520b和miR-520e参与了乳腺癌细胞中补体介导的免疫抑制作用。
     为进一步阐明miR-520b和miR-520e在乳腺癌细胞中参与补体介导的免疫抑制的分子机理,我们应用生物信息学软件在TargetScan和DIANA microT 3.0预测microRNA靶基因的数据库中对miR-520b和miR-520e的靶基因进行了预测筛选。结果发现,miR-520b和miR-520e均可以靶向与补体激活级联调节有关的靶基因——膜辅助因子(membrane co-factor, MCP, CD46)。本研究发现在MCF-7乳腺癌细胞中分别过表达miR-520b或miR-520e成熟体可以下调CD46蛋白质表达水平,但不能影响CD46 mRNA转录水平;当分别过表达miR-520b或miR-520e的抑制子时可见CD46蛋白质表达水平上调。这一结果表明CD46是miR-520b和miR-520e两者共同的靶基因。同时也提示miR-520b和miR-520e可能是通过结合CD46 3’非翻译区(3’UTR)抑制其翻译调控CD46蛋白质表达。为了进一步证明这个假设,我们克隆了CD46 3'UTR miR-520b和miR-520e结合区并构建了报告基因载体。报告基因结果表明miR-520b和miR-520e可以直接靶向结合CD46 3'UTR相应的结合区。ELISA结果表明,在乳腺癌细胞中过表达miR-520b或miR-520e可以增加补体成分C3b在肿瘤细胞上的积累,从而激活补体级联反应途径,促进CDC对肿瘤细胞的裂解作用。以上研究阐明乳腺癌细胞中miR-520b和miR-520e均可直接靶向CD46影响补体介导的细胞毒作用。
     2.miR-520b靶向HBXIP影响补体介导的细胞毒作用及其分子机制研究
     我们前期发现HBXIP在肿瘤细胞中促进增殖、抑制凋亡等方面发挥重要作用。本研究中我们探讨了HBXIP在乳腺癌细胞中对CDC的作用。我们首先检测了HBL-100, MCF-7, LM-MCF-7和MDA-MB-231细胞系中HBXIP的表达水平,发现三株乳腺癌细胞系HBXIP的表达水平均高于HBL-100细胞系,并发现HBXIP的表达水平与膜结合补体调节蛋白(mCRPs)的主要成员CD46, CD55和CD59的表达水平呈正相关。因此,我们设想HBXIP可能与调节CD46, CD55和CD59有关。
     报告基因实验结果显示,在过表达HBXIP基因的乳腺癌细胞系中,CD46, CD59报告基因活性升高,而RNA干扰HBXIP基因后其报告基因活性降低。应用Real-time PCR实验检测发现过表达HBXIP可以上调CD46, CD55和CD59的mRNA转录水平,而在干扰的乳腺癌细胞系CD46, CD55和CD59的mRNA水平下调。我们进一步发现在乳腺癌细胞系中瞬时过表达或干扰HBXIP可以以剂量依赖的方式上调或下调mCRPs的蛋白质表达水平。以上结果表明HBXIP在乳腺癌细胞中可以调节CD46, CD55和CD59的表达。
     为了进一步阐明HBXIP调节mCRPs的分子机制,我们通过报告基因、Western blot分析、免疫荧光染色方法发现HBXIP在乳腺癌细胞中可以激活NF-κB。双荧光素酶报告基因以及Western blot分析表明,用NF-κB特异性抑制剂PDTC处理MCF-7-HBXIP乳腺癌细胞后可拮抗CD46, CD55和CD59的表达,当RNA干扰NF-κB活性时也可获得相似的结果。上述结果表明HBXIP可以上调CD46, CD55和CD59可以通过NF-κB信号通路。我们还发现HBXIP也可以上调ERK1/2的磷酸化水平,而对丝裂原激活蛋白激酶(MAPK)其他两个成员p38和c-Jun N末端激酶(JNK)的磷酸化水平无影响。发现磷酸化的ERK1/2也可以上调CD46, CD55和CD59的表达水平。ERK1/2上游激酶MEK1的特异性抑制剂PD98059也可有效抑制mCRPs的蛋白质表达水平。进一步通过Western blot、双荧光素酶报告基因实验和免疫荧光染色等实验研究表明,ERK1/2的活化可以促进NF-κB的活性,并使其入核,进而促进mCRPs的表达。上述结果表明,HBXIP可以通过ERK1/2/NF-κB信号转导途径上调CD46,CD55和CD59的表达水平,进而降低乳腺癌细胞对CDC作用的敏感性,发挥乳腺癌细胞的免疫逃逸作用。
     为了探讨HBXIP上游调控因子,我们通过TargetScan软件预测发现HBXIP为miR-520b的靶基因。于是,我们克隆并且构建了含有HBXIP 3'UTR的报告基因载体,双荧光素酶报告基因实验以及Western blot分析结果表明miR-520b可以直接结合HBXIP 3'UTR从而抑制HBXIP蛋白质的翻译。表明HBXIP在参与调节CDC作用中可以被miR-520b调控。
     综上所述,本研究通过在乳腺癌细胞中对miR-520b或miR-520e的深入研究,发现miR-520b或miR-520e在乳腺癌免疫抑制方面发挥重要的作用,主要阐明了如下分子机制:1)miR-520b或miR-520e通过直接靶向结合补体调节蛋白CD46,影响乳腺癌细胞对补体攻击的效应;2)miR-520b靶蛋白HBXIP可以通过ERK1/2/NF-κB信号转导途径上调CD46, CD55和CD59的表达水平,进而降低乳腺癌细胞对CDC作用的敏感性,发挥乳腺癌细胞的免疫逃逸作用。本研究揭示了乳腺癌免疫抑制调节的一个新的分子机制,为乳腺癌的诊断、治疗提供了更丰富的理论依据。
MicroRNAs (miRNAs) are small approximately 21-23 nucleotide RNA species that are expressed from specialized genes. It has been reported that miRNAs participate cell growth, cell death, apoptosis, and metastasis during the carcinogenesis and tumor progression. Recently, it was reported that miR-520b was involved in immune regulation. CD46, CD55 and CD59 are clusters of membrane-bound complement-regulatory proteins (mCRPs), which are able to inhibit the complement-dependent cytotoxicity (CDC) through inhibiting C5 convertase during the complement activation cascade. Therefore, they play important roles in protecting tumor cells from complement attack. However, the mechanism of regulation for mCRPs remains unclear. It is unclear whether miR-520b is involved in the tumor immunosuppression. In the present study, we investigated the correlation between miR-520b or miR-520e and CRP using parallel breast cancer cell lines, such as MCF-7 and LM-MCF-7 (National patent of invent, China) that have different metastatic capabilities. The investigation contains two parts as follows:
     Part one:miR-520b and miR-520e are involved in complement-dependent cytotoxicity in breast cancer cells by directly targeting CD46
     According to our previous data in microarray comparing hepatoma cells and normal liver cells, we found that miR-520b was downreguated in hepatoma cells. Furthermore, to investigate the function of miR-520b and miR-520e in immunosuppression during breast cancer progression, we firstly examined the expression levels of miR-520b or miR-520e in normal breast HBL-100 cell line and other three breast cancer cell lines, including MCF-7, LM-MCF-7, and MDA-MB-231, by quantitative real-time PCR.. The results indicated that the expression levels of miR-520b and miR-520e in breast cancer MCF-7, LM-MCF-7, and MDA-MB-231 cells were downregulated relative to HBL-100 cells. Futhermore, we also examined the expression level of miR-520b in normal breast tissue and breast tumor tissue. And we found that the expression level of miR-520b between normal breast tissue and breast tumor tissue was significangtly different. We also found that the resistant capability of the breast cancer cells to CDC was stronger than that of normal breast cell line. To further elucidate the effect of miR-520b and miR-520e on CDC of breast cancer cells, we transfected miR-520b or miR-520e mimics as well as its anti-mimics into MCF-7 or MDA-MB-231 cells. The complement-mediated cytolysis assay revealed that the overexpression of miR-520b or miR-520e mimics was able to enhance the CDC sensitivity in breast cancer cells. Conversely, the suppression of miR-520b or miR-520e in breast cancer cells could decrease the sensitivity of breast cancer cells to CDC. Thus, we conclude that miR-520b and miR-520e are involved in immunosuppressive regulation mediated by complement.
     Furthermore, to elucidate the mechanism of immunosuppressive regulation mediated by miR-520b and miR-520e in breast cancer cells, we identified the candidate targets of miR-520b and miR-520e using program online, TargetScan and DIANA microT3.0. We found that both miR-520b and miR-520e could target a complement cascade regulatory target, such as the membrane co-factor (MCP, CD46). Overexpression of miR-520b or miR-520e mimics could decrease the protein expression of CD46 rather than the mRNA level of CD46. Conversely, overexpression of anti-miR-520b or anti-miR-520e could increase the protein expression level of CD46. These data demonstrated that CD46 indeed acted as a target of miR-520b or miR-520e. We also indicated that miR-520b or miR-520e was able to regulate CD46 by directly targeting 3'UTR of CD46 mRNA. To further confirm the finding, we constructed a dual-luciferase reporter gene vector containing the 3'UTR targeting sites of miR-520b or miR-520e mRNA. The luciferase reporter gene assay showed that miR-520b or miR-520e was able to directly target the 3'UTR of CD46 mRNA. And the ELISA assay indicated that overexpression of miR520b or miR-520e could increase the deposition of C3b on breast cancer cells, so that it could promot the CDC effect on breast cancer cells. Thus, we conclude that both miR-520b and miR-520e are involved in CDC by directly targeting CD46.
     Part two:Effect of miR-520b targeting HBXIP on complement-dependent cytotoxicity in breast cancer cells
     Previous studies revealed that hepatitis B X-interacting protein (HBXIP) was involved in cancer cell growth and inhibiting apoptosis. In this study, we investigated the relationship between HBXIP and CDC in breast cancer cells. We firstly examined the expression levels of HBXIP in HBL-100, MCF-7, LM-MCF-7 and MDA-MB-231 cell lines by RT-PCR and Western blot analysis. We found that the expression levels of HBXIP in the three breast cancer cell lines were higher than that in normal breast cell line. Moreover, Western blot analysis showed that HBXIP expression was positively correlated with the expression levels of CD46, CD55 and CD59. Thus, we supposed that HBXIP might act as a regulator of mCRPs. Then, Dual-luciferase reporter gene assay revealed that the CD46 and CD59 promoter activities were increased in HBXIP stably transfected breast cancer cell line. Contrary, the promoter activities of CD46 and CD59 were decreased by RNAi targeting HBXIP mRNA in breast cancer cells. Furthermore, we showed that HBXIP upregulated the mRNA expression levels of CD46, CD55 and CD59 in breast cancer cells by real-time PCR as well as the protein expression level by Western blot. These data indicate that HBXIP is able to regulate the expression of CD46, CD55, and CD59 in breast cancer cells. We further found that HBXIP could activate NF-κB in breast cancer cells by reporter gene assay, Western blot analysis and immunofluorescence staining. The treatment with NF-κB specific inhibitor PDTC could inhibit the expression of CD46, CD55 and CD59 in breast cancer cells by luciferase reporter gene assay and Western blot analysis. Thus, we conclude that HBXIP is able upregulate the expression of CD46, CD55 and CD59 through NF-κB signaling pathway. Moreover, we found that HBXIP could increase the phosphorylated level of ERK1/2 rather than p38 and c-Jun N-terminal kinase (JNK). The activated ERK1/2 could upregulate the expression levels of CD46, CD55 and CD59. Contrarily, the specific inhibitor of MEK1 (upstream kinase of ERK1/2) PD98059 was able to decrease the expression levels of CD46, CD55 and CD59. Western blot analysis, dual-luciferase reporter gene assay and immunofluorescence staining assay showed that the activation of ERK1/2 could stimulate NF-κB translocation so that promoting the expression levels of CD46, CD55 and CD59. Therefore, we conclude that HBXIP is able to regulate the CDC sensitivity to protect the breast cancer cells from complement attack through upregulating CD46, CD55 and CD59 involving ERK1/2-NF-κB cascade signaling pathways. To investigate the upstream regulator of HBXIP, we constructed a luciferase reporter gene vector containing full-length 3'UTR of HBXIP mRNA according to the predicted bound sites of miR-520b using TargetScan program. Dual-luciferase reporter gene assay and Western blot analysis manifested that miR-520b was able to downregulate the expression of HBXIP at protein level by targeting its 3'UTR, suggesting that miR-520b targeting HBXIP is involved in the regulation of complement-dependent cytotoxicity in breast cancer cells.
     In summary, we found that miR-520b and miR-520e were involved in immunosuppression in breast cancer cells. Our finding showed that miR-520b and miR-520e regulated the complement attack in breast cancer cells by directly targeting CD46. Moreover, HBXIP, one of the targets of miR-520b, could upregulate the expression levels of mCRPs to protect breast cancer cells from complement attack involving ERK1/2-NF-κB cascade signaling pathways. Our data provide a novel regulatory mechanism in immunosuppression of breast cancer cells.
引文
[1]Kaaks R, F Berrino, T Key, et al., Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst,2005,97:755-765.
    [2]Baker L, PR Quinlan, N Patten, et al., p53 mutation, deprivation and poor prognosis in primary breast cancer. Br J Cancer,102:719-726.
    [3]Fukunaga E, Y Inoue, S Komiya, et al., Smurf2 induces ubiquitin-dependent degradation of Smurfl to prevent migration of breast cancer cells. J Biol Chem, 2008,283:35660-35667.
    [4]Khalid S. D Hwang, Y Babichev, et al., Evidence for a tumor promoting effect of high-fat diet independent of insulin resistance in HER2/Neu mammary carcinogenesis. Breast Cancer Res Treat,2009.
    [5]Shou J, S Massarweh, CK Osborne, et al.. Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu cross-talk in ER/HER2-positive breast cancer. J Natl Cancer Inst,2004,96:926-935.
    [6]Calin GA, CD Dumitru, M Shimizu, et al., Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia.ProcNatl Acad Sci USA,2002,99:15524-15529.
    [7]Jovanovic M, MO Hengartner, miRNAs and apoptosis:RNAs to die for. Oncogene,2006,25:6176-6187.
    [8]Lee Y, M Kim, J Han, et al., MicroRNA genes are transcribed by RNA polymerase Ⅱ. Embo J,2004,23:4051-4060.
    [9]Lim LP, ME Glasner, S Yekta, et al., Vertebrate microRNA genes. Science,2003, 299:1540.
    [10]Houbaviy HB, MF Murray, PA Sharp, Embryonic stem cell-specific MicroRNAs. Dev Cell,2003,5:351-358.
    [11]Lagos-Quintana M, R Rauhut, J Meyer, et al., New micro RNAs from mouse and human. Rna,2003,9:175-179.
    [12]Tanzer A, PF Stadler, Molecular evolution of a microRNA cluster. J Mol Biol, 2004,339:327-335.
    [13]Yadav D, J Ngolab, RS Lim, et al., Cutting edge:down-regulation of MHC class I-related chain A on tumor cells by IFN-gamma-induced microRNA. J Immunol, 2009,182:39-43.
    [14]Durrant LG, MA Chapman, DJ Buckley, et al., Enhanced expression of the complement regulatory protein CD55 predicts a poor prognosis in colorectal cancer patients. Cancer Immunol Immunother,2003,52:638-642.
    [15]Inoue T, M Yamakawa, T Takahashi, Expression of complement regulating factors in gastric cancer cells. Mol Pathol,2002,55:193-199.
    [16]Gorter A, S Meri, Immune evasion of tumor cells using membrane-bound complement regulatory proteins. Immunol Today,1999,20:576-582.
    [17]Maio M, LI Brasoveanu, S Coral, et al., Structure, distribution, and functional role of protectin (CD59) in complement-susceptibility and in immunotherapy of human malignancies (Review). Int J Oncol,1998,13:305-318.
    [18]Rooney IA, JE Heuser, JP Atkinson, GPI-anchored complement regulatory proteins in seminal plasma. An analysis of their physical condition and the mechanisms of their binding to exogenous cells. J Clin Invest,1996, 97:1675-1686.
    [19]Gorter A, VT Blok, WH Haasnoot, et al., Expression of CD46, CD55, and CD59 on renal tumor cell lines and their role in preventing complement-mediated tumor cell lysis. Lab Invest,1996,74:1039-1049.
    [20]Rushmere NK, JM Knowlden, JM Gee, et al., Analysis of the level of mRNA expression of the membrane regulators of complement, CD59, CD55 and CD46, in breast cancer. Int J Cancer,2004,108:930-936.
    [21]Qiao L, TJ Zhao, FZ Wang, et al., NF-kappaB downregulation may be involved the depression of tumor cell proliferation mediated by human mesenchymal stem cells. Acta Pharmacol Sin,2008,29:333-340.
    [22]You J, D Mi, X Zhou, et al., A positive feedback between activated extracellularly regulated kinase and cyclooxygenase/lipoxygenase maintains proliferation and migration of breast cancer cells. Endocrinology,2009, 150:1607-1617.
    [23]Qiao L, Z Xu, T Zhao, et al., Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res,2008,18:500-507.
    [24]Qin X, H Zhang, X Zhou, et al., Proliferation and migration mediated by Dkk-1/Wnt/beta-catenin cascade in a model of hepatocellular carcinoma cells. Transl Res,2007,150:281-294.
    [25]Figueroa JD, KC Flanders, M Garcia-Closas, et al., Expression of TGF-beta signaling factors in invasive breast cancers:relationships with age at diagnosis and tumor characteristics. Breast Cancer Res Treat,2009.
    [26]Schmidt MK, J Tommiska, A Broeks, et al., Combined effects of single nucleotide polymorphisms TP53 R72P and MDM2 SNP309, and p53 expression on survival of breast cancer patients. Breast Cancer Res,2009,11:R89.
    [27]Jiang BH, LZ Liu, PI3K/PTEN signaling in angiogenesis and tumorigenesis. Adv Cancer Res.2009,102:19-65.
    [28]Roux PP. J Blenis, ERK and p38 MAPK-activated protein kinases:a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev,2004, 68:320-344.
    [29]Kraus S, R Seger, Z Fishelson, Involvement of the ERK mitogen-activated protein kinase in cell resistance to complement-mediated lysis. Clin Exp Immunol,2001,123:366-374.
    [30]Sun Z, R Andersson, NF-kappaB activation and inhibition:a review. Shock, 2002,18:99-106.
    [31]Chen J, XM Xu, CB Underhill, et al., Tachyplesin activates the classic complement pathway to kill tumor cells. Cancer Res,2005.65:4614-4622.
    [32]Li Q, IM Verma, NF-kappaB regulation in the immune system. Nat Rev Immunol,2002,2:725-734.
    [33]Ravi R, GC Bedi, LW Engstrom, et al., Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-kappaB. Nat Cell Biol,2001. 3:409-416.
    [34]Melegari M, PP Scaglioni, JR Wands, Cloning and characterization of a novel hepatitis B virus x binding protein that inhibits viral replication. J Virol,1998, 72:1737-1743.
    [35]Wang FZ, L Sha, LH Ye, et al., Promotion of cell proliferation by HBXIP via upregulation of human telomerase reverse transcriptase in human mesenchymal stem cells. Acta Pharmacol Sin,2008,29:83-89.
    [36]Wang FZ, L Sha, WY Zhang, et al., Involvement of hepatitis B X-interacting protein (HBXIP) in proliferation regulation of cells. Acta Pharmacol Sin,2007, 28:431-438.
    [37]Fujii R, C Zhu, Y Wen, et al., HBXIP, cellular target of hepatitis B virus oncoprotein, is a regulator of centrosome dynamics and cytokinesis. Cancer Res, 2006,66:9099-9107.
    [38]Marusawa H, S Matsuzawa, K Welsh, et al., HBXIP functions as a cofactor of survivin in apoptosis suppression. Embo J,2003,22:2729-2740.
    [39]Schmittgen TD, KJ Livak, Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc,2008,3:1101-1108.
    [40]Uliasz TF, SJ Hewett, A microtiter trypan blue absorbance assay for the quantitative determination of excitotoxic neuronal injury in cell culture. J Neurosci Methods,2000,100:157-163.
    [41]Kumar MS, J Lu, KL Mercer, et al., Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet,2007,39:673-677.
    [42]Iorio MV, P Casalini, C Piovan, et al., microRNA-205 regulates HER3 in human breast cancer. Cancer Res,2009,69:2195-2200.
    [43]Mertens-Talcott SU, S Chintharlapalli, X Li, et al., The oncogenic microRNA-27a targets genes that regulate specificity protein transcription factors and the G2-M checkpoint in MDA-MB-231 breast cancer cells. Cancer Res,2007,67:11001-11011.
    [44]Valastyan S, F Reinhardt, N Benaich, et al., A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell,2009,137:1032-1046.
    [45]Du J, S Yang, D An, et al., BMP-6 inhibits microRNA-21 expression in breast cancer through repressing deltaEFl and AP-1. Cell Res,2009,19:487-496.
    [46]Frankel LB, NR Christoffersen, A Jacobsen, et al., Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem,2008,283:1026-1033.
    [47]Miller TE, K Ghoshal, B Ramaswamy, et al., MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kipl. J Biol Chem,2008, 283:29897-29903.
    [48]Li W, R Duan, F Kooy, et al., Germline mutation of microRNA-125a is associated with breast cancer. J Med Genet,2009,46:358-360.
    [49]Kovalchuk O, J Filkowski, J Meservy, et al., Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol Cancer Ther,2008,7:2152-2159.
    [50]Gee HE, C Camps, FM Buffa, et al., MicroRNA-10b and breast cancer metastasis. Nature,2008,455:E8-9; author reply E9.
    [51]Saito Y, JM Friedman, Y Chihara, et al., Epigenetic therapy upregulates the tumor suppressor microRNA-126 and its host gene EGFL7 in human cancer cells. Biochem Biophys Res Commun,2009,379:726-731.
    [52]Shenouda SK. SK Alahari, MicroRNA function in cancer:oncogene or a tumor suppressor? Cancer Metastasis Rev,2009,28:369-378.
    [53]Friedman JM, G Liang, CC Liu, et al., The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2. Cancer Res,2009,69:2623-2629.
    [54]Lee KH, YG Goan, M Hsiao, et al., MicroRNA-373 (miR-373) post-transcriptionally regulates large tumor suppressor, homolog 2 (LATS2) and stimulates proliferation in human esophageal cancer. Exp Cell Res,2009, 315:2529-2538.
    [55]Lee YS, A Dutta, The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev,2007,21:1025-1030.
    [56]Gancz D, Z Fishelson, Cancer resistance to complement-dependent cytotoxicity (CDC):Problem-oriented research and development. Mol Immunol,2009. 46:2794-2800.
    [57]van Meerten T, RS van Rijn, S Hol, et al., Complement-induced cell death by rituximab depends on CD20 expression level and acts complementary to antibody-dependent cellular cytotoxicity. Clin Cancer Res,2006,12:4027-4035.
    [58]Vose JM, Antibody-targeted therapy for low-grade lymphoma. Semin Hematol, 1999,36:15-20.
    [59]Rheins LA, JJ Nordlund, Melanoma and immunity. Immunol Ser,1989, 46:769-787.
    [60]Descamps B, R Gagnon, R van der Gaag, et al., Antibody dependent cell mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC) in 229 sera from human renal allograft recipients. J Clin Lab Immunol,1979, 2:303-309.
    [61]Russell S, CD46:a complement regulator and pathogen receptor that mediates links between innate and acquired immune function. Tissue Antigens,2004, 64:111-118.
    [62]Fishelson Z, N Donin, S Zell, et al., Obstacles to cancer immunotherapy: expression of membrane complement regulatory proteins (mCRPs) in tumors. Mol Immunol,2003,40:109-123.
    [63]Migliore C, S Giordano, MiRNAs as new master players. Cell Cycle,2009, 8:2185-2186.
    [64]Davis BN, A Hata, Regulation of MicroRNA Biogenesis:A miRiad of mechanisms. Cell Commun Signal,2009,7:18.
    [65]Xia H, Y Qi, SS Ng, et al., MicroRNA-15b regulates cell cycle progression by targeting cyclins in glioma cells. Biochem Biophys Res Commun,2009, 380:205-210.
    [66]Bartel DP, MicroRNAs:genomics, biogenesis, mechanism, and function. Cell, 2004,116:281-297.
    [67]He L, JM Thomson, MT Hemann, et al., A microRNA polycistron as a potential human oncogene. Nature,2005,435:828-833.
    [68]Calin GA, CM Croce, MicroRNA signatures in human cancers. Nat Rev Cancer, 2006,6:857-866.
    [69]Liszewski MK, TC Farries, DM Lublin, et al., Control of the complement system. Adv Immunol,1996,61:201-283.
    [70]Kraus D, ME Medof, C Mold, Complementary recognition of alternative pathway activators by decay-accelerating factor and factor H. Infect Immun, 1998,66:399-405.
    [71]Liu J, MA Carmell, FV Rivas, et al., Argonaute2 is the catalytic engine of mammalian RNAi. Science,2004,305:1437-1441.
    [72]Buettner R, M Huang, T Gritsko, et al., Activated signal transducers and activators of transcription 3 signaling induces CD46 expression and protects human cancer cells from complement-dependent cytotoxicity. Mol Cancer Res, 2007,5:823-832.
    [73]Tone M, LE Diamond, LA Walsh, et al., High level transcription of the complement regulatory protein CD59 requires an enhancer located in intron 1. J Biol Chem,1999,274:710-716.
    [74]Walport MJ, Complement. Second of two parts. N Engl J Med,2001, 344:1140-1144.
    [75]Li G, J Domenico, Y Jia, et al., NF-kappaB-dependent induction of cathelicidin-related antimicrobial peptide in murine mast cells by lipopolysaccharide. Int Arch Allergy Immunol.2009,150:122-132.
    [76]Park PH, MR McMullen, H Huang, et al., Short-term treatment of RAW264.7 macrophages with adiponectin increases tumor necrosis factor-alpha (TNF-alpha) expression via ERK1/2 activation and Egr-1 expression:role of TNF-alpha in adiponectin-stimulated interleukin-10 production. J Biol Chem,2007. 282:21695-21703.
    [77]Carey AM, R Pramanik, LJ Nicholson, et al., Ras-MEK-ERK signaling cascade regulates androgen receptor element-inducible gene transcription and DNA synthesis in prostate cancer cells. Int J Cancer,2007,121:520-527.
    [78]Fan Y, H Chen, B Qiao, et al., Opposing effects of ERK and p38 MAP kinases on HeLa cell apoptosis induced by dipyrithione. Mol Cells,2007,23:30-38.
    [79]Kunisch E, M Gandesiri, R Fuhrmann, et al., Predominant activation of MAP kinases and pro-destructive/pro-inflammatory features by TNF alpha in early-passage synovial fibroblasts via TNF receptor-1:failure of p38 inhibition to suppress matrix metalloproteinase-1 in rheumatoid arthritis. Ann Rheum Dis, 2007,66:1043-1051.
    [80]Burk U, J Schubert, U Wellner, et al., A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep,2008,9:582-589.
    [81]Gregory PA, AG Bert, EL Paterson, et al., The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol,2008,10:593-601.
    [82]Kong W, H Yang, L He, et al., MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol Cell Biol,2008,28:6773-6784.
    [83]Paterson EL, N Kolesnikoff, PA Gregory, et al., The microRNA-200 family regulates epithelial to mesenchymal transition. ScientificWorldJournal,2008, 8:901-904.
    [84]Mellstedt H, Monoclonal antibodies in human cancer. Drugs Today (Bare),2003, 39Suppl C:1-16.
    [85]Murakami F, [Cytotoxic action of monoclonal antibodies OC125 and SH-9 on ovarian cancer cell lines]. Nippon Sanka Fujinka Gakkai Zasshi,1996, 48:125-132.
    [86]Power B, P Hudson, Keystone symposia:antibody-based therapeutics for cancer. Expert Opin Biol Ther,2003,3:385-389.
    [87]Wang SY, G Weiner, Complement and cellular cytotoxicity in antibody therapy of cancer. Expert Opin Biol Ther,2008,8:759-768.
    [88]Spiller OB, O Criado-Garcia, S Rodriguez De Cordoba, et al., Cytokine-mediated up-regulation of CD55 and CD59 protects human hepatoma cells from complement attack. Clin Exp Immunol,2000,121:234-241.
    [89]Varsano S, L Rashkovsky, H Shapiro, et al., Cytokines modulate expression of cell-membrane complement inhibitory proteins in human lung cancer cell lines. Am J Respir Cell Mol Biol,1998,19:522-529.
    [90]Bjorge L, J Hakulinen, T Wahlstrom, et al., Complement-regulatory proteins in ovarian malignancies. Int J Cancer,1997,70:14-25.
    [91]Ellison BS, MK Zanin, RJ Boackle, Complement susceptibility in glutamine deprived breast cancer cells. Cell Div,2007,2:20.
    [92]Hara T, A Kojima, H Fukuda, et al., Levels of complement regulatory proteins, CD35 (CR1), CD46 (MCP) and CD55 (DAF) in human haematological malignancies. Br J Haematol,1992,82:368-373.
    [93]Tak PP, GS Firestein, NF-kappaB:a key role in inflammatory diseases. J Clin Invest,2001,107:7-11.
    [94]Tak PP, DM Gerlag, KR Aupperle, et al., Inhibitor of nuclear factor kappaB kinase beta is a key regulator of synovial inflammation. Arthritis Rheum,2001, 44:1897-1907.
    [95]Lindstrom TM, PR Bennett, The role of nuclear factor kappa B in human labour. Reproduction,2005,130:569-581.
    [96]Schaaf MJ, L Willetts, BP Hayes, et al., The relationship between intranuclear mobility of the NF-kappaB subunit p65 and its DNA binding affinity. J Biol Chem,2006.281:22409-22420.
    [97]Gaundar SS. KF Bradstock, LJ Bendall, p38MAPK inhibitors attenuate cytokine production by bone marrow stromal cells and reduce stroma-mediated proliferation of acute lymphoblastic leukemia cells. Cell Cycle,2009, 8:2975-2983.
    [98]Dumka D, P Puri, N Carayol, et al., Activation of the p38 Map kinase pathway is essential for the antileukemic effects of dasatinib. Leuk Lymphoma,2009, 50:2017-2029.
    [99]Kumamoto H, K Ooya, Immunohistochemical detection of phosphorylated.JNK. p38 MAPK, and ERK5 in ameloblastic tumors. J Oral Pathol Med,2007, 36:543-549.
    [100]Astuti P, T Pike, C Widberg, et al., MAPK pathway activation delays G2/M progression by destabilizing Cdc25B. J Biol Chem,2009,284:33781-33788.
    [101]Wang J, JY Zhou, GS Wu, ERK-dependent MKP-1-mediated cisplatin resistance in human ovarian cancer cells. Cancer Res,2007,67:11933-11941.
    [102]Liang FP, CH Lin, CD Kuo, et al., Suppression of v-Src transformation by andrographolide via degradation of the v-Src protein and attenuation of the Erk signaling pathway. J Biol Chem,2008,283:5023-5033.
    [103]Beck SE, JM Carethers, BMP suppresses PTEN expression via RAS/ERK signaling. Cancer Biol Ther,2007,6:1313-1317.
    [104]Khavari TA, J Rinn, Ras/Erk MAPK signaling in epidermal homeostasis and neoplasia. Cell Cycle,2007,6:2928-2931.
    [105]Yang SH, ZZ Wu, CM Chien, et al., JNK and ERK mitogen-activated protein kinases mediate THDA-induced apoptosis in K562 cells. Cell Biol Toxicol,2008, 24:291-302.
    [106]Ethier C, Y Labelle, GG Poirier, PARP-1-induced cell death through inhibition of the MEK/ERK pathway in MNNG-treated HeLa cells. Apoptosis,2007, 12:2037-2049.
    [107]Rivas MA, RP Carnevale, CJ Proietti, et al., TNF alpha acting on TNFR1 promotes breast cancer growth via p42/P44 MAPK, JNK, Akt and NF-kappa B-dependent pathways. Exp Cell Res,2008,314:509-529.
    [108]Olive V, MJ Bennett, JC Walker, et al., miR-19 is a key oncogenic component of mir-17-92. Genes Dev,2009,23:2839-2849.
    [109]Si ML, S Zhu, H Wu, et al., miR-21-mediated tumor growth. Oncogene,2007, 26:2799-2803.
    [1]Bartel DP, MicroRNAs:genomics, biogenesis, mechanism, and function. Cell, 2004,116:281-297.
    [2]Bartel DP, CZ Chen, Micromanagers of gene expression:the potentially widespread influence of metazoan microRNAs. Nat Rev Genet,2004, 5:396-400.
    [3]Lee RC, RL Feinbaum, V Ambros, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell,1993, 75:843-854.
    [4]Smallridge R, A small fortune. Nat Rev Mol Cell Biol,2001,2:867.
    [5]Griffiths-Jones S, The microRNA Registry. Nucleic Acids Res,2004, 32:D109-111.
    [6]He L, JM Thomson, MT Hemann, et al., A microRNA polycistron as a potential human oncogene. Nature,2005,435:828-833.
    [7]Calin GA, CD Dumitru, M Shimizu, et al., Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocyticleukemia. Proc Natl Acad Sci USA,2002,99:15524-15529.
    [8]Yi R, Y Qin, IG Macara, et al., Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev,2003,17:3011-3016.
    [9]Gregory RI, R Shiekhattar, MicroRNA biogenesis and cancer. Cancer Res,2005, 65:3509-3512.
    [10]Liu J, MA Carmell, FV Rivas, et al., Argonaute2 is the catalytic engine of mammalian RNAi. Science,2004,305:1437-1441.
    [11]Sen GL, HM Blau, Argonaute 2/RISC resides in sites of mammalian mRNA decay known as cytoplasmic bodies. Nat Cell Biol,2005,7:633-636.
    [12]Li SC, P Tang, WC Lin, Intronic microRNA:discovery and biological implications. DNA Cell Biol,2007,26:195-207.
    [13]Aukerman MJ, H Sakai, Regulation of flowering time and floral organ identity by a MicroRNA and its APETALA2-like target genes. Plant Cell,2003, 15:2730-2741.
    [14]Ambros V, The functions of animal microRNAs. Nature,2004,431:350-355.
    [15]Calin GA, CM Croce, MicroRNA signatures in human cancers. Nat Rev Cancer, 2006,6:857-866.
    [16]Calin GA, CM Croce, MicroRNAs and chromosomal abnormalities in cancer cells. Oncogene,2006,25:6202-6210.
    [17]Calin GA, R Garzon, A Cimmino, et al., MicroRNAs and leukemias:how strong is the connection? Leuk Res,2006,30:653-655.
    [18]Chen CZ, L Li, HF Lodish, et al., MicroRNAs modulate hematopoietic lineage differentiation. Science,2004,303:83-86.
    [19]Fazi F, A Rosa, A Fatica, et al., A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis. Cell,2005,123:819-831.
    [20]El Ouaamari A, N Baroukh, GA Martens, et al., miR-375 targets 3'-phosphoinositide-dependent protein kinase-1 and regulates glucose-induced biological responses in pancreatic beta-cells. Diabetes,2008,57:2708-2717.
    [21]Xin M, EM Small, LB Sutherland, et al., MicroRNAs miR-143 and miR-145 modulate cytoskeletal dynamics and responsiveness of smooth muscle cells to injury. Genes Dev,2009,23:2166-2178.
    [22]Lagos-Quintana M, R Rauhut, W Lendeckel, et al., Identification of novel genes coding for small expressed RNAs. Science,2001,294:853-858.
    [23]Zhang B, X Pan, GP Cobb, et al., microRNAs as oncogenes and tumor suppressors. Dev Biol,2007,302:1-12.
    [24]Hutvagner G, MJ Simard, CC Mello, et al., Sequence-specific inhibition of small RNA function. PLoS Biol,2004,2:E98.
    [25]Meister G, M Landthaler, Y Dorsett, et al., Sequence-specific inhibition of microRNA- and siRNA-induced RNA silencing. Rna,2004,10:544-550.
    [26]Krutzfeldt J, N Rajewsky, R Braich, et al., Silencing of microRNAs in vivo with 'antagomirs'. Nature,2005,438:685-689.
    [27]Lewis BP, RE Green, SE Brenner, Evidence for the widespread coupling of alternative splicing and nonsense-mediated mRNA decay in humans. Proc Natl Acad Sci USA,2003,100:189-192.
    [28]Wightman B, I Ha, G Ruvkun, Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell, 1993,75:855-862.
    [29]Schmittgen TD, J Jiang, Q Liu, et al., A high-throughput method to monitor the expression of microRNA precursors. Nucleic Acids Res,2004,32:e43.
    [30]Jiang J, EJ Lee, Y Gusev, et al., Real-time expression profiling of microRNA precursors in human cancer cell lines. Nucleic Acids Res,2005,33:5394-5403.
    [31]Babak T, W Zhang, Q Morris, et al., Probing microRNAs with microarrays: tissue specificity and functional inference. Rna,2004,10:1813-1819.
    [32]Thomson JM, J Parker, CM Perou, et al., A custom microarray platform for analysis of microRNA gene expression. Nat Methods,2004,1:47-53.
    [33]Lu J, G Getz, EA Miska, et al., MicroRNA expression profiles classify human cancers. Nature,2005,435:834-838.
    [34]Cummins JM, Y He, RJ Leary, et al., The colorectal microRNAome. Proc Natl Acad Sci USA,2006,103:3687-3692.
    [35]Michael MZ, OC SM, NG van Holst Pellekaan, et al., Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res,2003,1:882-891.
    [36]Murakami Y, T Yasuda, K Saigo, et al., Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene,2006,25:2537-2545.
    [37]Takamizawa J, H Konishi, K Yanagisawa, et al., Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res,2004,64:3753-3756.
    [38]Calin GA, C Sevignani, CD Dumitru, et al., Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA,2004,101:2999-3004.
    [39]Ota A, H Tagawa, S Karnan, et al., Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma. Cancer Res,2004,64:3087-3095.
    [40]Saito Y, G Liang, G Egger, et al., Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell,2006,9:435-443.
    [41]Karube Y, H Tanaka, H Osada, et al., Reduced expression of Dicer associated with poor prognosis in lung cancer patients. Cancer Sci,2005,96:111-115.
    [42]Calin GA, F Trapasso, M Shimizu, et al., Familial cancer associated with a polymorphism in ARLTS1. N Engl J Med,2005,352:1667-1676.
    [43]Duan R, C Pak, P Jin, Single nucleotide polymorphism associated with mature miR-125a alters the processing of pri-miRNA. Hum Mol Genet,2007, 16:1124-1131.
    [44]Bottoi A, MC Zatelli, M Ferracin, et al., Identification of differentially expressed microRNAs by microarray:a possible role for microRNA genes in pituitary adenomas. J Cell Physiol,2007,210:370-377.
    [45]Lee CH, DC Wu, JM Lee, et al., Anatomical subsite discrepancy in relation to the impact of the consumption of alcohol, tobacco and betel quid on esophageal cancer. Int J Cancer,2007,120:1755-1762.
    [46]Geiger TR, DS Peeper, Metastasis mechanisms. Biochim Biophys Acta,2009, 1796:293-308.
    [47]Voulgari A, A Pintzas, Epithelial-mesenchymal transition in cancer metastasis: mechanisms, markers and strategies to overcome drug resistance in the clinic. Biochim Biophys Acta,2009,1796:75-90.
    [48]Korpal M, ES Lee, G Hu, et al., The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem,2008, 283:14910-14914.
    [49]Kong W, H Yang, L He, et al., MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol Cell Biol,2008,28:6773-6784.
    [50]Gregory PA, AG Bert, EL Paterson, et al., The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol,2008,10:593-601.
    [51]Burk U, J Schubert, U Wellner, et al., A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep,2008,9:582-589.
    [52]Hurteau GJ, JA Carlson, SD Spivack, et al., Overexpression of the microRNA hsa-miR-200c leads to reduced expression of transcription factor 8 and increased expression of E-cadherin. Cancer Res,2007,67:7972-7976.
    [53]Tryndyak VP, FA Beland, IP Pogribny, E-cadherin transcriptional down-regulation by epigenetic and microRNA-200 family alterations is related to mesenchymal and drug-resistant phenotypes in human breast cancer cells. Int J Cancer,2009,126:2575-2583.
    [54]Gebeshuber CA, K Zatloukal, J Martinez, miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep,2009, 10:400-405.
    [55]Ma L, J Teruya-Feldstein, RA Weinberg, Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature,2007,449:682-688.
    [56]Huang Q, K Gumireddy, M Schrier, et al., The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol,2008, 10:202-210.
    [57]Negrini M, GA Calin, Breast cancer metastasis:a microRNA story. Breast Cancer Res,2008,10:203.
    [58]Meng F, R Henson, H Wehbe-Janek, et al., MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology,2007,133:647-658.
    [59]Zhu S, H Wu, F Wu, et al., MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res,2008,18:350-359.
    [60]Asangani IA, SA Rasheed, DA Nikolova, et al., MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene,2008, 27:2128-2136.
    [61]Suzuki M, I Yoshino, Identification of microRNAs caused by DNA methylation that induce metastasis. Future Oncol,2008,4:775-777.
    [62]Bhaumik D, GK Scott, S Schokrpur, et al., Expression of microRNA-146 suppresses NF-kappaB activity with reduction of metastatic potential in breast cancer cells. Oncogene,2008,27:5643-5647.
    [63]Scott GK, A Goga, D Bhaumik, et al., Coordinate suppression of ERBB2 and ERBB3 by enforced expression of micro-RNA miR-125a or miR-125b. J Biol Chem,2007,282:1479-1486.
    [64]Li N, H Fu, Y Tie, et al., miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett,2009,275:44-53.
    [65]Liang Z, H Wu, S Reddy, et al., Blockade of invasion and metastasis of breast cancer cells via targeting CXCR4 with an artificial microRNA. Biochem Biophys Res Commun,2007,363:542-546.
    [66]Voorhoeve PM, C le Sage, M Schrier, et al., A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell, 2006,124:1169-1181.
    [67]Johnson SM, H Grosshans, J Shingara, et al., RAS is regulated by the let-7 microRNA family. Cell,2005,120:635-647.
    [68]Iorio MV, M Ferracin, CG Liu, et al., MicroRNA gene expression deregulation in human breast cancer. Cancer Res,2005,65:7065-7070.
    [69]Chan JA, AM Krichevsky, KS Kosik, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res,2005,65:6029-6033.
    [70]Lawrie CH, S Soneji, T Marafioti, et al., MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int J Cancer,2007,121:1156-1161.
    [71]Loffler D, K Brocke-Heidrich, G Pfeifer, et al., Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood,2007, 110:1330-1333.
    [72]Si ML, S Zhu, H Wu, et al., miR-21-mediated tumor growth. Oncogene,2007, 26:2799-2803.
    [73]Zhu S, ML Si, H Wu, et al., MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem,2007,282:14328-14336.
    [74]Baffa R, M Fassan, S Volinia, et al., MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. J Pathol,2009,219:214-221.
    [75]Sasayama T, M Nishihara, T Kondoh, et al., MicroRNA-10b is overexpressed in malignant glioma and associated with tumor invasive factors, uPAR and RhoC. Int J Cancer,2009,125:1407-1413.
    [76]Cimmino A, GA Calin, M Fabbri, et al., miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA,2005,102:13944-13949.
    [77]Raveche ES, E Salerno, BJ Scaglione, et al., Abnormal microRNA-16 locus with synteny to human 13q14 linked to CLL in NZB mice. Blood,2007, 109:5079-5086.
    [78]Linsley PS, J Schelter, J Burchard, et al., Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol,2007,27:2240-2252.
    [79]Tam W, SH Hughes, WS Hayward, et al., Avian bic, a gene isolated from a common retroviral site in avian leukosis virus-induced lymphomas that encodes a noncoding RNA, cooperates with c-myc in lymphomagenesis and erythroleukemogenesis. J Virol,2002,76:4275-4286.
    [80]Kluiver J, A van den Berg, D de Jong, et al., Regulation of pri-microRNA BIC transcription and processing in Burkitt lymphoma. Oncogene,2007, 26:3769-3776.
    [81]Kluiver J, S Poppema, D de Jong, et al., BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J Pathol, 2005,207:243-249.
    [82]Eis PS, W Tam, L Sun, et al., Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc Natl Acad Sci USA,2005,102:3627-3632.
    [83]Kluiver J, E Haralambieva, D de Jong, et al., Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma. Genes Chromosomes Cancer,2006,45:147-153.
    [84]Costinean S, N Zanesi, Y Pekarsky, et al., Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc Natl Acad Sci USA,2006,103:7024-7029.
    [85]O'Connell RM, KD Taganov, MP Boldin, et al., MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA,2007, 104:1604-1609.
    [86]Rodriguez A, E Vigorito, S Clare, et al., Requirement of bic/microRNA-155 for normal immune function. Science,2007,316:608-611.
    [87]Thai TH, DP Calado, S Casola, et al., Regulation of the germinal center response by microRNA-155. Science,2007,316:604-608.
    [88]Vigorito E, KL Perks, C Abreu-Goodger, et al., microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells. Immunity,2007, 27:847-859.
    [89]Chang TC, EA Wentzel, OA Kent, et al., Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell,2007, 26:745-752.
    [90]Tazawa H, N Tsuchiya, M Izumiya, et al., Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci USA,2007,104:15472-15477.
    [91]Bommer GT, I Gerin, Y Feng, et al., p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol,2007,17:1298-1307.
    [92]Raver-Shapira N, E Marciano, E Meiri, et al., Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell,2007,26:731-743.