HMGA2在维持人骨肉瘤细胞恶性表型中的关键作用及分子机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本研究以体外培养的人骨肉瘤U2OS细胞为实验模型,利用RNA干扰技术下调高迁移率蛋白A2 (HMGA2)表达,分析其对骨肉瘤细胞表型的影响,确定HMGA2在维持骨肉瘤细胞恶性表型中的作用;探讨其作用的分子机制及靶向HMGA2的RNA干扰技术在骨肉瘤治疗中的应用前景。结果:成功地构建了针对HMGA2的,基于DNA的shRNA干扰质粒载体(HMGA2-shRNA);将这一载体稳定转染人骨肉瘤U2OS细胞,明显地下调了其HMGA2的表达水平。细胞表型研究显示,抑制HMGA2表达明显地改变了该细胞的恶性表型;表现为细胞增殖和成集落能力明显下降;自发凋亡率则明显增加;同时细胞迁移速度、侵袭能力及粘附、铺展能力均明显受到抑制;并显著增加了细胞对化疗药物的敏感性。这些结果表明HMGA2在维持骨肉瘤U2OS细胞恶性表型及对化疗药物抗性方面起关键作用。应用实时定量RT-PCR对相关基因表达水平分析发现:下调HMGA2表达使细胞凋亡基因Caspase-3和Caspase-9表达上调,而凋亡抑制基因Bcl-2、Survivin表达水平降低;MMP-2和MMP-9表达降低,TIMP-3表达增高;CDKN1A表达增高,提示HMGA2是通过调控一系列靶基因表达水平来维持骨肉瘤U2OS细胞恶性表型的。本研究还首次发现下调HMGA2使该细胞的SOX2表达水平降低,PTEN表达水平提高。SOX2曾被报道为干细胞的标记物,这一发现提示在U2OS细胞群中有骨肉瘤干细胞样细胞的存在,且与HMGA2表达水平相关。而PTEN则曾被描述为肿瘤抑制基因,进一步证实了HMGA2在维持骨肉瘤细胞恶性表型的决定作用,提示靶向HMGA2的RNA干扰在骨肉瘤治疗中的应用前景。
High mobility group protein A2 (HMGA2) is a member of the HMGA family. HMGA2 was composed with three AT-hook binding domain and an acidic C-terminal domain. AT-hook can help HMGA2 recognize and bind to the AT-rich regions of DNA small groove specifically, resulting in DNA bending, stretching, looping or melting, regulating replication, transcription and DNA repair, also known as architectural transcription factor. HMGA2 showed high expression at the embryonic period, whereas in the normal mature tissues almost no expression, but in some malignant tumors showed high expression, such as breast cancer, non-small cell lung cancer, pancreatic cancer, retinoblastoma, squamous cell carcinoma, suggesting that HMGA2 play an important role in malignant transformation of tumors. More and more studies have shown that cancer stem cells (CSC) are not sensitive to the chemotherapy and radiotherapy, resulting in cancer recurrence, metastasis and hard cure. It has been reported, overexpression HMGA2 and H-Ras gene can inhibit the differentiation of breast cancer stem cells in order to facilitate the maintenance of their self-renewal, but the miRNA let-7 can promote the differentiation of breast cancer stem cells to treat breast cancer by inhibiting the expression of HMGA2 and H-Ras gene.
     Osteosarcoma incidence growths rapidly and tends to younger, can occur lung metastasis within a few months. It has high degree of malignancy and poor prognosis, and chemotherapy or radiotherapy is ineffective. Therefore, searching for effective treatment for osteosarcoma is the focus of osteosarcoma study, and targeted gene therapy is considered one of the best treatments, so to find effective target genes for treatment are extremely important. In view of differential expression of HMGA2 in tumor cells and normal cells, RNAi gene therapy has certain advantages. In this study, we culture human osteosarcoma U2OS cells, utilize RNAi to inhibit the expression of HMGA2, and to observe the biological behaviors of U2OS cells after silencing of HMGA2, and to investigate the postential mechanisms, and to explore the possible relationship between HMGA2 and cancer stem cells.
     PartⅠHMGA2-shRNA transfected into U2OS cells reduced the expression of HMGA2
     Objective:To construct an effective HMGA2-shRNA silencing plasmid, and detect its expression in human osteosarcoma cell U2OS. Methods: According to the full cDNA sequence of HMGA2 (N0. 003483) in GenBank, select the specific sequence 5'-CGCCAACGTTCGA TTTCAT-3 'for the target of the interference effect, and construct the sequence to the eukaryotic expression plasmid pGCsi-H1, containing the green fluorescent protein GFP. Select the scrambled plasmid which will not interfere with any human mRNA as negative control. HMGA2-shRNA and scrambled plasmids were transfected into U2OS cells, selected by G418 and obtained stable cell clone of the HMGA2-shRNA and scrambled respectively, RT-PCR detect silencing effect of HMGA2-shRNA. Results: Obtained stable transfection cell clones of HMGA2-shRNA and scrambled, RT-PCR results showed that HMGA2-shRNA plasmid reduced mRNA expression levels of HMGA2 significantly. Conclusion: Obtained effective stable cell clones of silencing HMGA2 gene successfull.
     PartⅡEffects on proliferation and apoptosis of HMGA2 in U2OS cells
     Objective:To study the effects of silencing HMGA2 on cell morphology, growth curve, colony forming ability, cell apoptosis and cell cycle of human osteosarcoma U2OS cell, to detect mRNA levels of related gene, and to explore its possible mechanism. Methods: Utilize microscopy observe cell morphology; CCK-8 kit detect cell growth curve; soft agar colony forming experiment testing the ability of cell cloning;Hoechst33342 dyes detect cell apoptosis; PI staining with flow cytometry detect cell cycle changes. Utilized real-time RT-PCR detecting mRNA level of relative genes. Results: compared with untreated and scrambled group, HMGA2 gene silencing group cell proliferation activity was decreased significantly (P<0.01); capacity of clonal growth was decreased; apoptotic cells increased significantly (P <0.05); the cell number of S phase decreased, most of the cell were blocked in the G0/G1 phase. The same culture conditions, untreated and scrambled group cells were spindle-type, cell body were more prominent after adheren, the majority of HMGA2 silencing group cells in shape of irregular polygons, cell body relatively flat after adherent. Real-time RT-PCR results show that the expression of the apoptosis gene caspase-3 and caspase-9 are increased,and the expression of the apoptosis inhibitor gene Bcl-2 and suvivin are decreased, and the expression of CDKN1A is increased. Conlusion: HMGA2 through inducted the expression of CDKN1A, overcomed the G1/S block to promote the proliferation of U2OS cells. HMGA2 inhibited spontaneous apoptosis of U2OS cells by reducing the expression of caspase-3 and caspase-9 and increasing the expression of Bcl-2 and suvivin.
     PartⅢEffects on migration and invasion of HMGA2 in U2OS cells
     Objective:To study the effects of HMGA2 gene on cell spreading, adhesion, migration, invasion in human osteosarcoma U2OS, to detect mRNA levels of related gene, and to explore its possible mechanism. Methods: Utilized microscope to observe cells spreading on FN; CCK-8 kit detect cells adhesion ability on FN and matrigel; scratch test detect cell migration; Boyden chamber observed cell invasion; Real-time RT-PCR detect related gene mRNA levels. Results: compared to untreated and scrambled group, HMGA2-shRNA group cells capacity of spreading on FN is diminished (P <0.05); and the capacity of adhesion on the FN (P <0.01) and Matrigel (P <0.05) were decreased significantly; migration ability of Cell injury repair and migrate to the chemokines were significantly reduced; Boyden chamber invasion assay results showed, the cell number of HMGA2-shRNA group reached the carbonate membrane is significantly lower than untreated and scrambled group(P <0.05). Real-time RT-PCR results showed that the expression of MMP-2 and MMP-9 were decreased significantly, while the expression of TIMP3 was increased. Conclusion: Reduced the expression of HMGA2 can decrease spreading and adhesion, migration and invasion of U2OS cell; real-time PCR results suggested that one important molecular mechanism of HMGA2 to maintain migration and invision of U2OS was increasing the expression of MMP-2 MMP-9 and reducing the expression of TIMP-3 expression.
     PartⅣthe role of HMGA2 in maintaining chemotherapy resistance of U2OS cells
     Objective:To study the effects of HMGA2 gene on chemotherapy sensitivity of U2OS cells; to detecte the mRNA level of stem cells markers, and to explore the relationship between HMGA2 and cancer stem cells. Methods: CCK-8 detected the response of U2OS cells to chemotherapeutic drugs vincristine; Real-time RT-PCR detected related gene mRNA levele of stem cells. Results:compared with untreated group(IC50=136.39ng/ml) and scrambled group (IC50=148.26 ng/ml), HMGA2-shRNA group(IC50=71.73 ng/ml) cells increased the sensitivity to chemotherapy vincristine(P<0.05). Real-time RT-PCR results showed that the expression of PTEN was increased and the expression of SOX2 was reduced. Conclusion: HMGA2 played an important role in maintaining the resistant to chemotherapy vincristine in U2OS cells, the results provided an experimental basis for the treatment of osteosarcoma by RNA interference targeted to HMGA2. For the first time, we found that HMGA2 can induce the expression of stem cell marker SOX2, suggesting that in the osteosarcoma U2OS cell populations exist stem cell-like cells, and associated with the expression level of HMGA2. The tumor suppressor gene PTEN, further confirmed the important role of HMGA2 in maintaining malignant phenotype of osteosarcoma cells, suggesting that HMGA2 is a promsing target gene in the RNA interference treatment osteosarcoma.
     PartⅤIsolation and biological behaviour analysis of prostatic carcinoma PC3 cell subpopulation
     Objective:To identify the characteristic of cancer stem-like cells in prostatic carcinoma PC3 cell line, and analyze the morphous and clone growth, investigate the method of discriminating the different tumor cell subpopulation by Rhodamin123 dye. Methods:Flow cytometry analyze the PC3 cells stained with Rhodamin123 dye; using fluorescent microscope to observe cells morphous and clone growth stained with Rhodamin123 and DAPI. Results:Two different tumor cell subpopulation exist in PC3 cell line, and the morphous, reproductive activity and colon growth are significant different in the two cell subpopulation. Conclusion:Cancer stem-like cell subpopulation exist in prostatic carcinoma PC3 cell line, the character of some cells exclude Rhodamin123 dye can be used to discriminate tumor stem cells and other tumor cells.
引文
[1]]Yu F, Yao H, Zhu P, et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells [J]. Cell, 2007, 131(6):1109-1123.
    [2]Langelotz C, Schmid P, Jakob C, et al. Expression of high-mobility group- protein HMGI-C mRNA in the peripheral blood is an independent poor prognostic indicator for survival in metastatic breast cancer [J]. Br J Cancer, 2003, 88(9): 1406-1410.
    [3]Young A R, Narita M. Oncogenic HMGA2: short or small[J]? Genes Dev, 2007, 21:1005–1009.
    [4]Miyazawa J, Mitoro A, Kawashiri S, et al. Expression of mesenchyme-specific gene HMGA2 in squamous cell carcinomas of the oral cavity [J]. Cancer Res, 2004, 64: 2024–2029.
    [5]Thuault S, Valcourt U, Petersen M, et al. Transforming growth factor-beta employs HMGA2 to elicit epithelial– mesenchymal transition [J]. J Cell Biol, 2006, 174: 175–183.
    [6]Pentimalli F, Dentice M, Fedele M, et al. Suppression of HMGA2 protein synthesis could be a tool for the therapy of well differentiated liposarcomas overexpressing HMGA2. Cancer Res, 2003, 63:7423–7.
    [7]Malek A, Bakhidze E, Noske A, et al. HGMA2 gene is a promising target for ovarian cancer silencing therapy. Int J Cancer, 2008, 123:348-356.
    [8]Bustin M, Reeves R. High-mobility-group chromosomal proteins: architectural components that facilitate chromatin function [J]. Prog Nucleic Acid Res Mol Biol, 1996, 54: 35-100.
    [9]Bustin M. Revised nomenclature for high mobility group (HMG) chromosomal proteins [J]. Trends Biochem Sci, 2001, 26: 152-153.
    [10]Frank O, Schwanbeck R, Wisniewski J R. Protein footprinting reveals specific binding modes of a high mobility group protein I to DNAs of different conformation [J]. J Biol Chem, 1998, 273: 20015-20020.
    [11]Yie J, Liang S, Merika M , et al. Intra- and intermolecular cooperative binding of high-mobility-group protein I(Y) to the beta-interferon promoter [J]. Mol Cell Biol, 1997, 17: 3649-3662.
    [12]Noro B, Licheri B, Sgarra R, et al. Molecular dissection of the architectural transcription factor HMGA2 [J]. Biochemistry, 2003, 42: 4569-4577.
    [13]Chin MT, Pellacani A, Wang H, et al. Enhancement of serum-response factor-dependent transcription and DNA binding by the architectural transcription factor HMG-I(Y) [J]. JBiol Chem, 1998, 273: 9755-9760.
    [14]Beitzel B, Bushman F. Construction and analysis of cells lacking the HMGA gene family [J]. Nucleic Acids Res, 2003, 31(17):5025-5032.
    [15]Martinez Hoyos J, Fedele M, Battista S, et al. Identification of the genes up- and down-regulated by the high mobility group A1 (HMGA1) proteins: tissue specificity of the HMGA1-dependent gene regulation [J]. Cancer research, 2004, 64(16):5728-5735.
    [16]Zhao K, Kas E, Gonzalez E, et al. SAR-dependent mobilization of histone H1 by HMG-I/Y in vitro: HMG-I/Y is enriched in H1-depleted chromatin [J]. EMBO J, 1993, 12: 3237-3247.
    [17]Reeves R, Leonard WJ, Nissen MS. Binding of HMG-I(Y) imparts architectural specificity to a positioned nucleosome on the promoter of the human interleukin-2 receptor alpha gene [J]. Mol Cell Biol, 2000, 20: 4666-4679.
    [18]Lee Y S, Dutta A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene [J]. Genes Dev, 2007, 21: 1025–1030.
    [19]Mayr C, Hemmann M T, Bartel D P. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation [J]. Science, 2007, 315: 1576–1579.
    [20]Kazmierczak B, Dal Cin P, Wanschura S, et al. HMGIY is the target of 6p21.3 rearrangements in various benign mesenchymal tumors [J]. Genes Chromosomes Cancer, 1998, 23: 279-285.
    [21] Lund T, Holtlund J, Laland S G. On the phosphorylation of low molecular mass HMG (high mobility group) proteins in Ehrlich ascites cells [J]. FEBS Lett, 1985, 180(2): 275-279.
    [22]Nissen M S, Langan T A, Reeves R. Phosphorylation by cdc2 kinase modulates DNA binding activity of high mobility group I nonhistone chromatin protein [J]. J Biol Chem, 1991, 266:19945–19952.
    [23]Reeves R, Langan T A, Nissen M S. Phosphorylation of the DNA-binding domain of nonhistone high-mobility group I protein by cdc2 kinase: reduction of binding affinity [J]. Proc Nat Acad Sci USA, 1991, 88:1671–1675.
    [24]Banks G C, Li Y, Reeves R. Differential in vivo modifications of the HMGI(Y) nonhistone chromatin proteins modulate nucleosome and DNA interactions [J]. Biochem, 2000, 39(28):8333-8346.
    [25]Sgarra R, Lee J, Tessari M A, et al. The AT-hook of the chromatin architectural transcription factor high mobility group A1a is arginine-methylated by protein arginine methyltransferase 6 [J]. J Biol Chem, 2006, 281(7): 3764-3772.
    [26]Fedele M, Battista S, Manfioletti G, et al. Role of the high mobility group A proteins in human lipomas [J]. Carcinogenesis, 2001, 22(10):1583-1591.
    [27]Schwanbeck R, Manfioletti G, Wisniewski JR. Architecture of high mobility group protein I-C.DNA complex and its perturbation upon phosphorylation by Cdc2 kinase. J Biol Chem, 2000, 275: 1793-1801.
    [28]Goodwin G. The high mobility group protein, HMGI-C. Int J Biochem Cell Biol, 1998, 30: 761-766.
    [29]Di Agostino S, Fedele M, Chieffi P, et al. Phosphorylation of high-mobility group protein A2 by Nek2 kinase during the first meiotic division in mouse spermatocytes. Mol Biol Cell, 2004, 15: 1224-1232.
    [30]Tamimi Y, Van der poel H G, Denyn M M, et al. Increased Expression of High Mobility Group Protein I(Y) in High Grade Prostatic Cancer Determined by in Situ Hybridization [J]. Cancer Res, 1993, 53(22):5512-5516.
    [31]Edberg D D, Adkins J N, Springer D L,et al. Dynamic and differential in vivo modifications of the isoform HMGA1a and HMGA1b chromatin proteins [J]. J Biol Chem, 2005, 280(10):8961-8973.
    [32]Edberg D D, Bruce J E, Siems W F, et al. In Vivo Posttranslational Modifications of the High Mobility Group A1a Proteins in Breast Cancer Cells of Differing Metastatic Potential [J]. 2004, 43(3):11500-11515.
    [33]Zou Y, Webb K, Perna A D, et al. A mass spectrometric study on the in vitro methylation of HMGA1a and HMGA1b proteins by PRMTs: methylation specificity, the effect of binding to AT-rich duplex DNA, and the effect of C-terminal phosphorylation [J]. Biochemistry, 2007, 46: 7896-7906.
    [34]Green MC. Catalog of mutant genes and polymorphic loci. In: Genetic Variants and Strains of the Laboratory Mouse. Lyon M and Searle A (eds.) Oxford University Press, 1989, pp12-403.
    [35]Zhou X, Benson KF, Ashar HR, et al. Mutation responsible for the mouse phenotype in the developmentally regulated factor HMGI-C [J]. Nature, 1995, 376 (6543):771-774
    [36]Benson K F, Chada K. Mini-mouse: phenotypic characterization of a transgenic insertional mutant allelic to pygmy [J]. Genet Res, 1994, 64: 27-33.
    [37]Brants J R, Ayoubi T A, Chada K, et al. Differential regulation of the insulin-like growth factorⅡmRNA-binding protein genes by architectural transcription factor HMGA2 [J]. FEBS Lett, 2004, 569(1-3):277-283.
    [38]Cleynen I, Brants J R, Peeters K, et al. HMGA2 regulates transcription of the Imp2 gene via an intronic regulatory element in cooperation with nuclear factor-kappaB [J]. Cancer Res, 2007, 5(4):363-372.
    [39]Chiappetta G, Avantaggiato V, Visconti R, et al. High level expression of the HMGI (Y) gene during embryonic development [J]. Oncogene, 1996, 13: 2439- 2446.
    [40]Hirning-Folz U, Wilda M, Rippe V, et al. The expression pattern of the Hmgic gene during development [J]. Genes Chromosomes Cancer, 1998, 23: 350-357.
    [41]Zhou X, Benson KF, Przybysz K, et al.Genomic structure and expression of the murine Hmgi-c gene [J]. Nucleic Acids Res, 1996, 24: 4071-4077.
    [42]Vartiainen E, Palvimo J, Mahonen A, et al. Selective decrease in low-Mr HMG proteins HMG I and HMG Y during differentiation of mouse teratocarcinoma cells [J]. FEBS Lett, 1988, 228: 45-48.
    [43]Giancotti V, Pani B, D'Andrea P, et al. Elevated levels of a specific class of nuclear phosphoproteins in cells transformed with v-ras and v-mos oncogenes and by cotransfection with c-myc and polyoma middle T genes [J]. EMBO J, 1987, 6: 1981-1987.
    [44]Battista S, Fedele M, Hoyos JM, et al. High-mobility-group A1 (HMGA1) proteins down-regulate the expression of the recombination activating gene 2 (RAG2)[J]. Biolchem J, 2005, 389(1):91-97.
    [45]Battista S, Pentimalli F, Baldassarre G, et al. Loss of HMGA1 gene function affects embryonic stem cell lympho-hematopoietic differentiation [J]. FASEB J, 2003, 17(11): 1496-1498.
    [46]Melillo R M, Pierantioni G M, Scala S, et al. Critical role of the HMGI(Y) proteins in adipocytic cell growth and differentiation [J]. Mol Cell Biol, 2001, 21(7):2485-2495.
    [47]Caron L, Bost F, Prot M, et al. A new role for the oncogenic high-mobility group A2 transcription factor in myogenesis of embryonic stem cells [J]. Oncogene, 2005, 24: 6281-6291.
    [48]Fedele M, Fidanza V, Battista S, et al. Haploinsufficiency of the Hmga1 gene causes cardiac hypertrophy and myelo-lymphoproliferative disorders in mice [J]. Cancer Res, 2006, 66: 2536-2543.
    [49]Foti D, Chiefari E, Fedele M, et al. Lack of the architectural factor HMGA1 causes insulin resistance and diabetes in humans and mice [J]. Nat Med, 2005, 11: 765-773.
    [50]Bussemakers M J G, Van de Ven W J M, Debruyne F M J, et al. Identification of high mobility group protein I(Y) as potential progression marker for prostate cancer by differential hybridization analysis [J]. Cancer Res, 1991, 51:606–611.
    [51]Donato G, Martinez Hoyos J, Amorosi A, et al. High mobility group A1 expression correlates with the histological grade of human glial tumors [J]. Oncol Rep, 2004, 11:1209– 1213.
    [52]Abe N, Watanabe T, Lzumisato Y, et al. High mobility group A1 is expressed in metastatic adenocarcinoma to the liver and intrahepatic cholangiocarcinoma, but not in hepatocellular carcinoma: its potential use in the diagnosis of liver neoplasms [J]. J Gastroenterol, 2003, 38:1144–1149.
    [53]Chuma M, Saeki N, Yamamoto Y, et al. Expression profiling in hepatocellular carcinoma with intrahepatic metastasis: identification of high-mobility group I(Y) protein as a molecular marker of hepatocellular carcinoma metastasis [J]. Keio J Med, 2004, 53:90–97.
    [54]Chang Z G, Yang LY, Wang W, et al. Determination of high mobility group A1 (HMGA1) expression in hepatocellular carcinoma: a potential prognostic marker [J]. Dig Dis Sci, 2005, 50:1764–1770.
    [55]Tesfaye A, Di-Cello F, Hillion J, et al. The high-mobility group A1 gene up-regulates cyclooxygenase 2 expression in uterine tumorigenesis [J]. Cancer Res, 2007, 67:3998–4004.
    [56]Pierantoni G M, Agosti V, Fedele M, et al. High mobility group A1 proteins are overexpressed in human leukemias [J]. Biochem J, 2003, 372(Pt1):145–150.
    [57]Petit M M, Mols R, Schoenmakers E F, et al. LPP, the preferred fusion partner gene of HMGIC in lipomas, is a novel member of the LIM protein gene family [J]. Genomics, 1996, 36:118-129.
    [58]Schoenmakers E F, Huysmans C, van de Ven W J. Allelic knockout of novel splice variants of human recombination repair gene RAD51B in t(12;14) uterine leiomyomas [J]. Cancer Res, 1999, 59:19-23.
    [59]Rogalla P, Kazmierczak B, Meyer-Bolte K, et al. The t(3;12)(q27;q14-q15) with underlying HMGIC-LPP fusion is not determining an adipocytic phenotype [J]. Genes Chromosomes Cancer, 1998, 22:100-104.
    [60]Schoenberg Fejzo M, Ashar HR, Krauter KS, et al. Translocation breakpoints upstream of the HMGIC gene in uterine leiomyomata suggest dysregulation of this gene by a mechanism different from that in lipomas [J]. Genes Chromosomes Cancer, 1996, 17:1-6.
    [61]Quade B J, Weremowicz S, Neskey D M, et al. Fusion transcripts involving HMGA2 are not a common molecular mechanism in uterine leiomyomata with rearrangements in 12q15 [J]. Cancer Res, 2003, 63:1351-1358.
    [62]Wood L J, Maher J F, Bunton T E, et al. The oncogenic properties of the HMG-I gene family [J]. Cancer Res, 2000, 60:4256-4261.
    [63]Wood L J, Mukherjee M, Dolde CE, et al. HMG-I/Y, a new c-Myc target gene and potential oncogene [J]. Mol Cell Biol, 2000, 20:5490-5502.
    [64]Fedele M, Battista S, Kenyon L, et al. Overexpression of the HMGA2 gene in transgenic mice leads to the onset of pituitary adenomas [J]. Oncogene, 2002, 21:190-3198.
    [65]Finelli P, Pierantoni G M, Giardino D, et al. The high mobility group A2 gene is amplified and overexpressed in human prolactinomas [J]. Cancer Res, 2002, 62: 2398-2405.
    [66]Fedele M, Pierantoni G M, Visone R, et al. E2F1 activation is responsible for pituitary adenomas induced by HMGA2 gene overexpression [J]. Cell Div, 2006, 1:17.
    [67]Muller H, Bracken A P, Vernell R, et al: E2Fs regulate the expression of genes involved in differentiation, development, proliferation, and apoptosis [J]. Genes Dev, 2001, 15:267-285.
    [68]Subramanian D, Griffith J D. Interactions between p53, hMSH2-hMSH6 and HMGI(Y) on Holliday junctions and bulged bases [J]. Nucleic Acids Res, 2002, 30(11): 2427–2434.
    [69]Reeves R, Adair J E. Role of high mobility group (HMG) chromatin proteins in DNA repair [J]. DNA Repair, 2005, 4(8): 926–938.
    [70]Borrmann L, Schwanbeck R, Heyduk T, et al. High mobility group A2 protein and its derivatives bind a specific region of the promoter of DNA repair gene ERCC1 and modulate its activity [J]. Nucleic Acids Res, 2003, 31(23): 6841–6851.
    [71]Baldassarre G, Battista S, Belletti B, et al. Negative regulation of BRCA1 gene expression by HMGA1 proteins accounts for the reduced BRCA1 protein levels in sporadic breast carcinoma [J]. Mol Cell Biol, 2003, 23(7):2225–2238.
    [72]Baldassarre G, Belletti B, Battista S, et al. HMGA1 protein expression sensitizes cells to cisplatin-induced cell death [J]. Oncogene,2005, 24: 6809–6819.
    [73]Boo L M, Lin H H, Chung V, et al. High mobility group A2 potentiates genotoxic stress in part through the modulation of basal and DNA damage- dependent phospha- tidylinositol 3-kinase-related protein kinase activation [J]. Cancer Res, 2005, 65: 6622–663.
    [74]Muller-Tidow C, Ji P, Diederichs S, et al. The cyclin A1-CDK2 complex regulates DNA double-strand break repair [J]. Mol Cell Biol, 2004, 24(20):8917–8928.
    [75]Huber M A, Kraut N, Beug H. Molecular requirements for epithelial- mesenchymal transition during tumor progression [J]. Curr Opin Cell Biol, 2005, 17(5):548–558.
    [76]Reeves R, Edberg D D, Li Y. Architectural transcription factor HMGI(Y) promotes tumor progression and mesenchymal transition of human epithelial cells [J]. Mol Cell Biol, 2001,21:575–594.
    [77]Thuault S, Valcourt U, Petersen M, et al. Transforming growth factor-βemploys HMGA2 to elicit epithelial-mesenchymal transition [J]. J Cell Biol, 2006, 174(2):175–183.
    [78]Pagano M, Pepperkok R, Verde F, et al. Cyclin A is required at two points in the human cell cycle [J]. EMBO J, 1992, 11(3):961–971.
    [79]Fedele M, Visone R, De Martino I, et al. HMGA2 induces pituitary tumorigenesis by enhancing E2F1 activity [J]. Cancer Cell, 2006, 9(6):459–471.
    [80]Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell proliferation and transformation [J]. Biochim Biophys Acta, 1991, 1072:129–157.
    [81]Karin M, Liu Z, Zandi E. AP-1 function and regulation [J]. Curr Opin Cell Biol, 1997, 9:240–246.
    [82]Vallone D, Battista S, Pierantoni G M, et al. Neoplastic transformation of rat thyroid cells requires the JunB and Fra1 gene induction which is dependent on the HMGI-C gene products [J]. EMBO J, 1997, 17:5310–5321.
    [83]Casalino L, Bakiri L, Talotta F, et al. Fra-1 promotes growth and survival in RAS-transformed thyroid cells by controlling cyclin A transcription [J]. EMBO J, 2007, 26:1878–1890.
    [84]Pierantoni G M, Rinaldo C, Esposito F, et al. High Mobility Group A1 (HMGA1) proteins interact with p53 and inhibit its apoptotic activity [J]. Cell Death Differ, 2006, 13(9): 1554–1563.
    [85]Frasca F, Rustighi A, Malaguarnera R, et al. HMGA1 inhibits the function of p53 family members in thyroid cancer cells [J]. Cancer Res, 2006, 66(6): 2980–2989.
    [86]Pierantoni G M, Rinaldo C, Mottolese M, et al. High-mobility group A1 inhibits p53 by cytoplasmic relocalization of its proapoptotic activator HIPK2 [J]. J Clin Invest, 2007, 117 (3):693–702.
    [87]Mantovani A. Cancer: inflammation by remote control [J]. Nature, 2005, 435(7043): 752–753.
    [88]Zhang X M, Verdine G L. A small region in HMG I(Y) is critical for cooperation with NF-B on DNA [J]. J Biol Chem, 1999, 274 (29): 20235–20243.
    [89]Bogdan C. Nitric oxide and the immune response [J]. Nature Immunol, 2001, 2: 907–916.
    [90]Geller D A, Billiar T R. Molecular biology of nitric oxide synthases [J]. Cancer Metastasis Rev, 1998, 17 (1):7–23.
    [91]Tsujii M, Kawano S, DuBois R N. Cycloxygenase-2 expression in human colon cancer cells increases metastatic potential [J]. Proc Natl Acad Sci USA, 1997, 94(7): 3336–3340.
    [92]Huber M A, Kraut N, Beug H. Molecular requirements for epithelial- mesenchymal transition during tumor progression [J]. Curr Opin Cell Bio, 2005, 17 (5):548–558.
    [93]Tallini G, Dal Cin P, Rhoden KJ, et al. Expression of HMGI-C and HMGI(Y) in ordinary lipoma and atypical lipomatous tumors: immunohistochemical reactivity correlates with karyotypic alterations [J]. Am J Pathol, 1997, 151: 37-43.
    [94]Kazmierczak B, Dal Cin P, Sciot R, et al. Inflammatory myofibroblastic tumor with HMGIC rearrangement [J]. Cancer Genet Cytogenet, 1999, 112:156-160.
    [95]Abe N, Watanabe T, Suzuki Y, et al. An increased high-mobility group A2 expression level is associated with malignant phenotype in pancreatic exocrine tissue [J]. Br J Cancer, 2003, 89(11): 2104-2109.
    [96]Reya T, Morrison S J, Clarke M F, et al. Stem cells, cancer, and cancer stem cells [J]. Nature, 2001, 414:105–111.
    [97]Pardal R, Clarke M F, Morrison S J. Applying the principles of stem-cell biology to cancer [J]. Nature Rev Cancer, 2003, 3:895–902.
    [98]Molofsky A V, Pardal R, Morrison S J. Diverse mechanisms regulate stem cell self-renewal [J]. Curr Opin Cell Biol, 2004, 16:700–-707.
    [99]Taipale J, Beachy P A. The Hedgehog and Wnt signalling pathways in cancer [J]. Nature, 2001, 411:349–-354.
    [100]Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukemia after transplantation into SCID mice [J]. Nature, 1994, 17:645–648.
    [101]Bonnet D, Dick J E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell [J]. Nature Med, 1997, 3: 730–737.
    [102]Warner J K, Wang J C, Hope K J, et al. Concepts of human leukemic development [J]. Oncogene, 2004, 23:7164–7177.
    [103]Park I-K, Qian D, Kiel M, et al. Bmi-1 is required for the maintenance of adult self-renewing haematopoietic stem cells [J]. Nature, 2003, 423:302–305.
    [104]Lessard J, Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells [J]. Nature, 2003, 423:255–260.
    [105]Singh S K, Clarke I D, Terasaki M, et al. Identification of a cancer stem cell in human brain tumors [J]. Cancer Res, 2003, 63:5821–5828.
    [106]Hemmati H D, Nakano I, Lazareff J A, et al. Cancerous stem cells can arise from pediatric brain tumors [J]. Proc Natl Acad Sci USA, 2003, 100:15178–15183.
    [107]Molofsky A V, He S H, Bydon M,et al. Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a andp19Arf senescence pathways. Genes Dev, 2005, 19:1432–1437.
    [108]Lowe S W, Sherr C J. Tumor suppression by Ink4a–-Arf: progress and puzzles. Curr Opin Genet Dev, 2003, 13:77–83.
    [109]Stiles B, Groszer M, Wang S, et al. PTENless means more [J]. Dev Biol, 2004, 273:175–184.
    [110]Di Cristofano A, Pandolfi P P. The multiple roles of PTEN in tumor suppression [J]. Cell, 2000, 100:387–390.
    [111]Zhang J, Grindley J C, Yin T, et al. PTEN maintains haematopoitic stem cells and acts in lineage choice and leukaemia prevention [J]. Nature, 2006, 441:518–522.
    [112]Yilmaz OH, Valdez R, Theisen BK, et al. Pten dependence distinguishes haematopoietic stem cells form leukaemia-initiating cells [J]. Nature, 2006, 441:475–482.
    [113]Mitsui K, Tokuzawa Y, Itoh H, et al. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells [J]. Cell, 2003, 113:631–642.
    [114]Vaish M. Mismatch repair deficiencies transforming stem cells into cancer stem cells and therapeutic imp lications [J]. Mol Cancer, 2007, 2 (6):26.
    [115]Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice [J]. Nature, 1994, 367:645–648.
    [116]Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. Prospective identification of tumorigenic breast cancer cells [J]. Proc Natl Acad Sci USA, 2003, 100:3983– 3988.
    [117]Singh S K, Hawkins C, Clarke I D, et al. Identification of human brain tumour initiating cells [J]. Nature, 2004, 432(7015):396–401.
    [118]O’Brien C A, Pollett A, Gallinger S, et al. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice [J]. Nature, 2007, 445:106–110.
    [119]Ricci-Vitiani L, Lombardi D G, Pilozzi E, et al. Identification and expression of human colon-cancer-initiating cells [J]. Nature, 2007, 445:111–115.
    [120]Prince M E, Sivanandan R, Kaczorowski A, et al. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma [J]. Proc Natl Acad Sci USA, 2007, 104(3):973–978.
    [121]Li C, Heidt D G, Dalerba P, et al. Identification of pancreatic cancer stem cells [J]. Cancer Res, 2007, 67(3):1030–1037.
    [122]Kondo T, Setoguchi T, Taga T. Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line [J]. Proc Natl Acad Sci USA, 2004, 101(3):781–786.
    [123]Patrawala L, Calhoun T, Schneider-Broussard R, et al. Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2- cancer cells aresimilarly tumorigenic [J]. Cancer Res, 2005, 65(14):6207–6219.
    [124]Hirschmann-Jax C, Foster A E, Wulf G G, et al. A distinct "side population" of cells with high drug efflux capacity in human tumor cells [J]. Proc Natl Acad Sci USA, 2004, 101(39):14228–14233.
    [125]Barnhart B C, Simon M C. Metastasis and stem cell pathways [J]. Cancer Metastasis Rev, 2007, 26 (2):261-271.
    [126]Goodell M A, Brose K, Paradis G, et al. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo[J]. J Exp Med, 1996, 183:1797-1806.
    [127]Asakura A, Rudnicki M A. Side population cells from diverse adult tissues are capable of in vitro hematopoietic differentiation [J]. Experimental Hematology, 2002, 30:1339-1345.
    [128]Behbod F, Xian W, Shaw C A, et al. Transcriptional profiling of mammary gland side population cells [J]. Stem Cells, 2006, 24(4):1065–1074.
    [129]Chen J, Hersmus N, Van D, et al. The adult pituitary contains a cell population displaying stem /progenitor cell and early embryonic characteristics [J]. Endocrinology, 2005, 146 (9):3985 -3998.
    [130]Xu Y X, He Z Y, Zhu H Y, et al. Murine fertilized ovum, blastomere and morula cells lacking SP phenotype [J]. Sci China C Life Sci, 2007, 50(6): 762-765.
    [131]Zhou S, Schuetz J D, Bunting K D, et al. The ABC transpoter Bcrp1/ABCG2 is expression in a wide variety of stem cells and is a molecular determinant of the side-population phenotype [J]. Nat Med, 2001, 7:1028–1034.
    [132]Woodward W A, Chen M S, Behbod F, et al. WNT/beta– catenin mediates radiation resistance of mouse mammary progenitor cells [J]. Proc Natl Acad Sci USA, 2007, 104(2):618-623.
    [133]Zen Y, Fujii T, Yoshikawa S, et al. Histological and culture studies with respect to ABCG2 expression support the existence of a cancer cell hierarchy in human hepatocellular carcinoma [J]. Am J Pathol, 2007, 170(5):1750-1762.
    [134]Robey R W, PolgarO, Deeken J, et al. ABCG2: determining its relevance in clinical drug resistance [J]. Cancer Metastasis Rev, 2007, 26(1): 39-57.
    [135]Hadnagy A, Gaboury L, Beaulieu R, et al. SP analysis may be used to identify cancer stem cell populations [J]. Exp Cell Res, 2006, 312(19):3701-3710.
    [136]Sales-Pardo I, Avendaìo A, Martinez-Muìoz V, et al. Flow Cytometry of the side population: tips & tricks [J]. Cell Oncol, 2006, 28(1-2):37-53.
    [137]Malek A, Bakhidze E, Noske A, et al. HMGA2 gene is a promising target for ovarian cancer silencing therapy [J]. Int J Cancer , 2008 ,123: 348–356.
    [138]Chau K Y, Manfioletti G, Cheung-Chau KW, et al. Derepression of HMGA2 gene expression in retinoblastoma is associated with cell proliferation [J]. Mol Med, 2003, 9(5-8):154-165.
    [139]司徒镇强,吴军正。《细胞培养》。世界图书出版公司, 2007,181-192。
    [140]Wolf M. Influence of matrigel on biodistribution studies in cancer research [J]. Pharmazie, 2008, 63(1):43-48.
    [141]韩锐。《抗癌药物研究与实验技术》。北京医科大学和中国协和医科大学联合出版社。1997,324-341。
    [142]Deryugina E I, Quigley J P. Matrix metalloproteinases and tumor metastasis [J]. Cancer Metastasis Rev, 2006, 25(1):9-34.
    [143]Stetler-Stevenson W G. Matrix metalloproteinases in angiogenesis:a movin target for therapeutic intervention [J]. J Clin Invest, 1999, 103(9):1237-12.
    [144]Habich A, J urga M, Markiewicz I, et al. Early appearance of stem/ progenitor cells wit h neural– like characteristics in human cord blood mononuclear fraction cultured in vitro [J]. Exp Hematol, 2006, 34 :914 - 925.
    [145]Samuel J, Hart C, Gilmore P, et a1. Enrichment of the prostate side population the quest for the prostate epithelial stem cell [J]. Eur Urol Suppl, 2006, 5:62-62.
    [146]Challen G A, Little M H. A side order of stem cells: the SP phenotype [J ] . Stem Cells, 2006, 24:3-12.
    [147]张玥,张凤春,张雁云.乳腺癌MCF27肿瘤细胞SP亚群初步分析[J ] .上海交通大学学报(医学版) ,2006 ,26 (5) :507 - 510.
    [148]Wang Y,Hao D,Stein WD,Wang L.A kinetic study of Rhodamine123 pumping by P-glycoprotein.Biochim Biophys Acta[J].2006,1758:1671-1675.