用户名: 密码: 验证码:
Wnt通路抑制分子DKK1在肝细胞癌中异常高表达的分子机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
发现肝癌(Human hepatocellular careinoma,HCC)相关基因的重要步骤之一是获得在肝癌组织中异常表达的基因。我们实验室运用cDNA芯片方法筛选到了一批在肝癌中异常表达的已知和未知基因。对这些基因的功能进行进一步研究将有助于对肝癌发病分子机理的深入了解。我们首先挑选了6个在芯片结果显示有差异表达的较有意义的基因,运用Northern blot方法做进一步验证。本课题对其中一个基因Dickkopf1(DKK1)与肝癌的相关性进行较深入的研究。
     我们的结果发现在肝癌细胞株中分泌蛋白DKK1对Wnt/β-catenin信号通路具有抑制作用,这种抑制作用由于β-catenin基因的突变而丧失;而在肝癌细胞株中DKK1的表达水平和β-catenin定位相关,部分DKK1表达病例β-catenin存在突变,两套临床病例组化分析显示DKK1高表达与β-catenin胞浆/核累积具有相关性;在肝癌细胞中活化的Wnt信号可以反馈调节DKK1的表达。大样本量肝癌组织(来自314例外科切除和192例肝移植肝癌病人)免疫组化实验显示DKK1高表达肝癌病人,包括AFP阴性和TNM stage I病人,存在较差的预后;DKK1和β-catenin双阳性病人存在更差的预后;并且多因素分析显示DKK1是肝癌病人总体生存和无瘤生存的独立预后指标。我们的分析显示DKK1基因在侵袭性肝癌细胞株和发生复发的肝癌病人中高表达;体外运动侵袭等相关实验和裸鼠体内实验显示干扰DKK1表达后显著抑制肝癌细胞侵袭转移的发生;我们进一步探讨了DKK1促进肝癌细胞侵袭转移过程的具体信号途径和分子机制。上述结果提示在肝癌中DKK1存在高表达并与Wnt信号的活化相关,同时DKK1可以作为肝癌病人的预后指标。
     第一部分DKK1在肝癌细胞Wnt通路中作用
     【目的】探讨DKK1在Wnt信号途径中的抑制作用是否由于β-catenin基因的突变而丧失和是否由于β-catenin基因的突变引起Wnt信号途径的活化反馈调节DKK1基因的表达。【方法】用Western blot和免疫荧光方法检测肝癌细胞株中DKK1表达和β-catenin定位的情况。选用不同细胞株分别干扰β-catenin基因的表达和转入β-catenin突变体提高β-catenin基因的表达,Western blot方法检测DKK1基因表达水平的改变。分别选用存在β-catenin基因突变和不存在β-catenin基因突变的肝癌细胞株,以TOPflash/FOPflash作为报告质粒,pRL-SV40质粒作为内对照,用双荧光素报告系统、Western blot和免疫荧光方法检测DKK1对Wnt信号变化的影响。用组织芯片免疫组织化学方法分析临床病例中DKK1表达和β-catenin定位的关系。【结果】在肝癌细胞株中DKK1的表达水平和β-catenin定位相关;干扰和提高β-catenin基因的表达可相应降低和上调DKK1基因的表达水平。DKK1能够抑制HuH-7细胞中Wnt-3a活化信号,不抑制β-catenin突变体活化信号,同时DKK1不抑制β-catenin存在突变和胞浆/核累积的肝癌细胞株HepG2、MHCC97L中活化的Wnt信号;在这两类细胞株中DKK1也相应的改变或不改变β-catenin表达和定位的情况。部分DKK1表达病例β-catenin存在突变,大样本量临床病例中组化分析显示DKK1表达与β-catenin胞浆/核累积具有相关性。【结论】在肝癌细胞中,由于β-catenin基因的突变使DKK1对于Wnt信号途径的抑制作用丧失;而Wnt信号途径的活化可以反馈调节DKK1基因的表达。
     第二部分DKK1在临床肝癌病人预后分析中的应用
     【目的】探讨DKK1表达情况与肝癌病人预后(总体生存和无瘤生存)及肝癌病人临床病理特征间的关系。【方法】应用免疫组织化学染色方法研究肝癌组织芯片中DKK1表达及β-catenin定位的情况。单因素方差分析比较组间差异,卡方检验或Fisher精确概率法比较不同DKK1和β-catenin表达组间阳性率的差别,Kaplan-Meier法计算各组间不同生存率,Log-rank检验比较生存期差别。用Cox比例风险模型进行多因素分析以了解各个相关因素对总体生存率和无瘤生存率的影响。【结果】临床病例中DKK1组化分析显示DKK1表达与肝癌病人总体生存和无瘤生存相关,DKK1高表达肝癌病人,包括AFP阴性和TNMⅠ期病人,存在较差的预后。多因素分析显示DKK1是肝癌病人总体生存和无瘤生存的独立预后指标。DKK1高表达与β-catenin胞浆/核定位显著相关,DKK1/β-catenin双阳性的肝癌病人更倾向于多结节或弥散型复发和肝外转移,而这部分病人也存在更差的预后;DKK1/β-catenin也是影响肝癌病人总体生存率和无瘤生存率的独立因素。【结论】DKK1高表达肝癌病人存在较差的预后,DKK1可以作为肝癌的预后指标。
     第三部分DKK1对肝癌细胞侵袭转移的影响及其分子机制
     【目的】探讨DKK1基因在肝癌侵袭和转移中的作用和DKK1参与肝癌侵袭转移的分子机制。【方法】用定量PCR和Western blot方法检测肝癌细胞株和临床肝癌标本中DKK1的表达情况。通过化学合成RNA干扰片段瞬时干扰和质粒载体稳定转染干扰HCCLM3细胞DKK1的表达,体外损伤修复实验、侵袭实验和裸鼠体内实验检测下调DKK1表达前后肝癌细胞侵袭转移能力的改变情况。对3种肝癌细胞株DKK1高、低表达情况下的细胞进行表达谱芯片分析,利用配对t检验比较DKK1高表达组和DKK1低表达组之间的差异基因,分析这些差异基因相关的癌相关信号通路,进一步采用定量PCR对芯片结果进行验证,并在临床病例中检测这些分子的表达与DKK1表达的相关性。【结果】DKK1基因在侵袭性肝癌细胞株和发生复发的肝癌病人中高表达。体外损伤修复实验、侵袭实验证实,当DKK1被干扰后,肝癌细胞增殖能力没有发生改变而细胞侵袭被抑制。裸鼠体内实验显示干扰DKK1表达后显著抑制肝癌细胞肺转移的发生。DKK1高表达组对比DKK1低表达组在基因表达谱上存在显著差异,涉及VEGF、NF-κB、P13K和Integrin等癌相关信号通路;MMP16、KDR、PTPRA这3个在两组中差异显著的基因与肿瘤侵袭转移密切相关,定量PCR验证结果与基因芯片结果一致。在66例临床组织标本中MMP16、KDR、PTPRA的mRNA表达水平都与DKK1的表达水平成正相关,因而DKK1可能通过影响MMP16降解细胞外基质和通过KDR促进肿瘤血管形成等途径参与肿瘤侵袭转移过程。【结论】DKK1通过影响细胞外基质的降解和肿瘤血管的形成等促进肝癌侵袭和转移的发生。
In order to identify novel genes related to the carcinogenesis of human hepatocellular carcinoma(HCC),we utilized the eDNA microarray technology to discover differential gene expression profile between HCC tissue and its corresponding non-cancerous liver tissue.We found a cluster of genes with abnormal expression pattern in HCCs.Furthermore,the upregulated expression of 6 genes in HCC samples was confirmed using Northern blot.Among them,we selected Dickkopf-1(DKK1) for further investigation.
     Our research showed that DKK1 inhibited Wnt/β-catenin signaling in HCC cell lines withoutβ-catenin mutation,whereas DKK1 in HCC cell lines withβ-catenin mutations had no antagonistic effect on elevated Wnt/β-catenin signaling.The expression level of DKK1 was associated with the staining pattern ofβ-catenin in HCC cell lines,and DKK1 overexpression correlated withβ-catenin cytoplasmic/nuclear accumulation in clinical HCC samples.Meanwhile,the expression of DKK1 is regulated by Wnt/β-eatenin signaling in HCC cell lines.High DKK1 expression predicted unfavorable prognosis in HCC patients,especially in early stage patients and those with normal AFP levels.The HCC patients with high DKK1 expression and cytoplasmic/nuclearβ-catenin accumulation had very poor prognosis.In multivariate analyses,DKK1 was an independent predictor for overall survival(OS) and disease free survival(DFS) of HCC patients.In our studies,we found that DKK1 was preferentially overexpressed in recurrent HCC patients and metastatic HCC cell lines.Depletion of DKK1 caused a notable decrease in cell migration and invasiveness both in vitro and in vivo.We further explored the signal pathway and molecular mechanism triggered by DKK1 for the promotion of cell motility and metastasis.Our data demonstrated that DKK1 expression can be induced by an active Wnt/β-catenin signaling pathway and DKK1 is a novel prognostic predictor for patients with HCC.
     SectionⅠRole of DKK1 in the Wnt Pathway in HCC Cells
     【Objective】To determine whether extracellular DKK1 exerts a suppressive effect and the relationship between its expression and cytoplasmic/nuclearβ-catenin accumulation in HCC.【Methods】The expression patterns of DKK1 andβ-catenin were assessed by Western blotting and immunofluorescence in HCC cell lines. Regulation of DKK1 expression was also examined by transfectingβ-catenin mutant or small interfering RNAs directed againstβ-catenin into HCC cells.The effect of DKK1 on Wnt signaling was analyzed by TOPflash/FOPflash luciferase reporter assay,Western blotting and immunofluorescence in HCC cell lines with either wild-type or mutantβ-catenin.The expression patterns of DKK1 andβ-catenin were assessed by immunohistochemistry in tissue microarrays.【Results】The level of DKK1 in HCC cell lines is associated with staining pattern ofβ-catenin and its expression is regulated by Wnt/β-catenin signaling.Meanwhile,in HCC cell lines withoutβ-catenin mutation,DKK1 inhibited the Wnt-3a-activated Wnt/β-catenin signaling,whereas DKK1 in HCC cell lines withβ-catenin mutations had no antagonistic effect on elevated Wnt/β-catenin signaling.Immunohistochemical staining using tumor tissue microarrays consisting of 314 HCC patients showed that a high level of DKK1 expression was associated withβ-catenin cytoplasmic/nuclear accumulation.【Conclusion】DKK1 expression can be induced by an active Wnt/β-catenin signaling pathway and the suppressive effect of extracellular DKK1 on Wnt signaling is lost when cytoplasmicβ-catenin mutations occur in HCC cells.
     SectionⅡPrognostic Significance of DKK1 for HCC Patients
     【Objective】To measure the level of DKK1 expression in patients with HCC to evaluate its correlation with clinicopathological features and significance on prognosis.【Methods】Tissue microarrays containing 314 HCC patients who underwent surgical resection between 1997 and 2000 were used to detect the expression patterns of DKK1 andβ-catenin by immunohistochemistry. Clinicopathologic data for these patients were evaluated.The prognostic significance was assessed by using Kaplan-Meier survival estimates and log-rank tests.【Results】In the large cohort of HCC patients with long-term follow-up,DKK1 expression predicted unfavorable prognosis in HCC patients,especially in normal AFP and early stage HCC patients.In multivariate analyses,DKK1 was an independent predictor for OS and DFS of HCC patients.Furthermore,the high level of DKK1 expression was correlated withβ-catenin cytoplasmic/nuclear staining in clinical HCC samples,and patients with DKK1~+/β-catenin~+ had higher rates of diffused/distant recurrence.HCC patients with high DKK1 expression and cytoplasmic/nuclearβ-catenin staining had very poor prognosis,and the the co-index of DKK1/β-catenin also was an independent prognosticator for both OS and DFS of patients with HCC.【Conclusion】DKK1 expression predicted unfavorable prognosis and is a novel prognostic predictor for HCC patients.
     SectionⅢImpact of DKK1 on the Invasion and Metastasis of HCC Cells
     【Objective】To explore the role of DKK1 in HCC cell migration and metastasis and investigate the molecular mechanism triggered by DKK1 for the promotion of HCC metastasis.【Methods】DKK1 expression in HCC cell lines and in tumor tissues derived from HCC patients was investigated using quantitative real-time PCR (qRT-PCR) and Western blot analysis.The role of DKK1 in HCC was investigated by DKK1 depletion using transient or stable transfection of small interfering RNAs directed against DKK1.The in vitro and in vivo effects of DKK1 on the cell invasion and metastasis were examined by wound healing assay,cell invasion assay and metastasis model on mice.We examined the expression profile of 3 paired DKK1-high and DKK1-low expression HCC cell lines.Differentially expressed genes were identified based on paired t-test and the involved signaling pathways were analysed among these genes.We further confirmed the data of cDNA microarray by qRT-PCR and verified the correlation among these genes and DKK1 expression in clinical HCC samples.【Results】DKK1 was preferentially overexpressed in recurrent HCC patients and metastatic HCC cell lines.Depletion of DKK1 caused a notable decrease in cell migration and invasiveness both in vitro and in vivo.Differentially expressed genes were identified between DKK1-high and DKK1-low expression group,and these genes were involved in the VEGF,NF-κB,PI3K and Integrin signaling pathways.The expression of 3 selected-genes was analyzed by qRT-PCR,and the results were in agreement with the microarray data.The expression of MMP16,KDR,and PTPRA was positively correlated with DKK1 mRNA expression in 66 HCC samples.
     【Conclusion】DKK1 might be critically involved in the metastasis of HCCs through the activation of MMP 16 and promotion of angiogenesis.
引文
1.Parkin DM,Bray F,Ferlay J,et al.Global cancer statistics,2002[J].CA Cancer J Clin,2005,55(2):74-108.
    2.Tang ZY,Ye SL,Liu YK,et al.A decade's studies on metastasis of hepatocellular carcinoma[J].J Cancer Res Clin Oncol,2004,130(4):187-196.
    3.Teufel A,Staib F,Kanzler S,et al.Genetics of hepatocellular carcinoma[J].World J Gastroenterol,2007,13(16):2271-2282.
    4.Villanueva A,Newell P,Chiang DY,et al.Genomics and signaling pathways in hepatocellular carcinoma[J].Semin Liver Dis,2007,27(1):55-76.
    5.Laurent-Puig P,Zucman-Rossi J.Genetics ofhepatocellular tumors[J].Oncogene,2006,25(27):3778-3786.
    6.Thompson MD,Monga SP.WNT/beta-catenin signaling in liver health and disease[J].Hepatology,2007,45(5):1298-1305.
    7.Krupnik VE,Sharp JD,Jiang C,et al.Functional and structural diversity of the human Dickkopf gene family[J].Gene,1999,238(2):301-313.
    8.Glinka A,Wu W,Delius H,et al.Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction[J].Nature,1998,391(6665):357-362.
    9.Fedi P,Bafico A,Nieto Sofia A,et al.Isolation and biochemical characterization of the human DKK1 homologue,a novel inhibitor of mammalian Wnt signaling[J].J Biol Chem,1999,274(27):19465-19472.
    10.Mao B,Wu W,Li Y,et al.LDL-receptor-related protein 6 is a receptor for Dickkopf proteins[J].Nature,2001,411(6835):321-325.
    11.Mao B,Wu W,Davidson G,et al.Kremen proteins are Dickkopf receptors that regulate Wnt/β-catenin signalling[J].Nature,2002,417(6889):664-667.
    12.Bafico A,Liu G.Yaniv A,et al.Novel mechanism of Wnt signalling inhibition mediated by Dickkopf-1 interaction with LRP6/Arrow[J].Nat Cell Biol,2001,3(7):683-686.
    13.Gonzalez-Sancho JM,Aguilera O,Garcia JM,et al.The Wnt antagonist DICKKOPF-1 gene is a downstream target of beta-catenin/TCF and is downregulated in human colon cancer[J].Oncogene,2005,24(6):1098-1103.
    14.Kuphal S,Lodermeyer S,Bataille F,et al.Expression of Dickkopf genes is strongly reduced in malignant melanoma[J].Oncogene,2006,25(36):5027-5036.
    15.Wirths O,Waha A,Weggen S,et al.Overexpression of human Dickkopf-1,an antagonist of wingless/WNT signaling,in human hepatoblastomas and Wilms'tumors[J].Lab Invest,2003,83(3):429-434.
    16.Forget MA,Turcotte S,Beauseigle D,et al.The Wnt pathway regulator DKK1 is preferentially expressed in hormone-resistant breast tumours and in some common cancer types[J].Br J Cancer,2007,96(4):646-653.
    17.Yamabuki T,Takano A,Hayama S,et al.Dikkopf-1 as a Novel Serologic and Prognostic Biomarker for Lung and Esophageal Carcinomas[J].Cancer Res,2007,67(6):2517-2525.
    18.Patil MA,Chua MS,Pan KH,et al.An integrated data analysis approach to characterize genes highly expressed in hepatocellular carcinoma[J].Oncogene,2005,24(23):3737-3747.
    19.Niida A,Hiroko T,Kasai M,et al.DKK1,a negative regulator of Wnt signaling,is a target of the beta-catenin/TCF pathway[J].Oncogene,2004,23(52):8520-8526.
    20.Lustig B,Jerchow B,Sachs M,et al.Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors[J].Mol Cell Biol,2002,22(4):1184-1193.
    21.Nusse R,Varmus HE.Wnt genes[J].Cell,1992,69(7):1073-1087.
    22.Cadigan KM,Nusse R.Wnt signaling:A common theme in animal development[J].Genes Dev,1997,11(24):3286-3305.
    23.Giles RH,van Es JH,Clevers H.Caught up in a Wnt storm:Wnt signaling in cancer[J].Biochim Biophys Acta,2003,1653(1):1-24.
    24.Polakis P.Wnt signaling and cancer[J].Genes Dev,2000,14(15):1837-1851.
    25.Gospodarowicz MK,Miller D,Groome PA,et al.The process for continuous improvement of the TNM classification[J].Cancer,2004,100(1):1-5.
    26.Terris B,Pineau P,Bregeaud L,et al.Close correlation between beta-catenin gene alterations and nuclear accumulation of the protein in human hepatocellular carcinomas[J].Oncogene,1999,18(47):6583-6588.
    27.Shimada M,Hamatsu T,Yamashita Y,et al.Characteristics of multicentric hepatocellular carcinomas:comparison with intrahepatic metastasis[J].World J Surg,2001,25(8):991-995.
    28.孟奎,石群立,周晓军等.组织芯片技术在免疫组化及原位杂交阳性对照中的应用[J].诊断病理学杂志,2004,11(6):435-436.
    29.Arimura E,Kotoh K,Nakamuta M,et al.Local recurrence is an important prognostic factor of hepatocellular carcinoma[J].Word J Gastroenterol,2005,11(36):5601-5606.
    30.Qin LX,Tang ZY.Recent progress in predictive biomarkers for metastatic recurrence of human hepatocellular carcinoma:a review of the literature[J].J Cancer Res Clin Oncol,2004,130(9):497-513.
    31.Peng SY,Chen WJ,Lai PL,et al.High alpha-fetoprotein level correlates with high stage,early recurrence and poor prognosis of hepatocellular carcinoma:significance of hepatitis virus infection,age,p53 and beta-catenin mutations[J].Int J Cancer,2004,112(1):44-50.
    32.Llovet JM,Di Bisceglie AM,Bruix J,et al.Design and endpoints of clinical trials in hepatocellular carcinoma[J].J Natl Cancer Inst,2008,100(10):698-711.
    33.Inagawa S,Itabashi M,Adachi S,et al.Expression and prognostic roles of beta-catenin in hepatocellular carcinoma:correlation with tumor progression and postoperative survival[J].Clin Cancer Res,2002,8(2):450-456.
    34.Schmitt-Gr(a|¨)ff A,Ertelt V,Allgaier HP,et al.Cellular retinol-binding protein-1 in hepatocellular carcinoma correlates with beta-catenin,Ki-67 index,and patient survival[J].Hepatology,2003,38(2):470-480.
    35.Gustavson MD,Crawford HC,Fingleton B,et al.Tcf binding sequence and position determines beta-catenin and Lef-1 responsiveness of MMP-7promoters[J].Mol Carcinog,2004,41(3):125-139.
    36.Marchenko GN,Marchenko ND,Leng J,et al.Promoter characterization of the novel human matrix metalloproteinase-26 gene:regulation by the T-cell factor-4implies specific expression of the gene in cancer cells of epithelial origin[J].Biochem J,2002,363(Pt 2):253-262.
    37.Takahashi M,Tsunoda T,Seiki M,et al.Identification of membrane-type matrix metalloproteinase-1 as a target of the beta-catenin/Tcf4 complex in human colorectal cancers[J].Oncogene,2002,21(38):5861-5867.
    38.Hanahan D,Weinberg RA.The hallmarks of cancer[J].Cell,2000,100(1):57-70.
    39.Liotta LA.Tumor invasion and metastases--role of the extracellular matrix:Rhoads Memorial Award lecture[J].Cancer Res,1986,46(1):1-7.
    40.T(?)m(?)r J,Csuka O,Orosz Z,et al.Molecular pathology of tumor metastasis.I.Predictive pathology[J].Pathol Oncol Res,2001,7(3):217-230.
    41.Lowy AM,Clements WM,Bishop J,et al.beta-Catenin/Wnt signaling regulates expression of the membrane type 3 matrix metalloproteinase in gastric cancer[J].Cancer Res,2006,66(9):4734-4741.
    42.Nakada M,Nakamura H,Ikeda E,et al.Expression and tissue localization of membrane-type 1,2,and 3 matrix metalloproteinases in human astrocytic tumors[J].Am J Pathol,1999,154(2):417-428.
    43.Ohnishi Y,Tajima S,Ishibashi A.Coordinate expression of membrane type-matrix metalloproteinases-2 and 3(MT2-MMP and MT3-MMP) and matrix metalloproteinase-2(MMP-2) in primary and metastatic melanoma cells[J].Eur J Dermatol,2001,11(5):420-423.
    44.Kato H,Yoshikawa M,Miyazaki T,et al.Expression of vascular endothelial growth factor(VEGF) and its receptors(Flt-1 and Flk-1) in esophageal squamous cell carcinoma[J].Anticancer Res,2002,22(6C):3977-3984.
    45.Hyder SM,Murthy L,Stancel GM.Progestin regulation of vascular endothelial growth factor in human breast cancer cells[J].Cancer Res,1998,58(3):392-395.
    46.Zhang H,Wu J,Meng L,et al.Expression of vascular endothelial growth factor and its receptors KDR and Fit-1 in gastric cancer cells[J].World J Gastroenterol,2002,8(6):994-998.
    47.Harada Y,Ogata Y,Shirouzu K.Expression of vascular endothelial growth factor and its receptor KDR(kinase domain-containing receptor)/Flk-1(fetal liver kinase-1) as prognostic factors in human colorectal cancer[J].Int J Clin Oncol,2001,6(5):221-228.
    48.An SJ,Nie Q,Chen ZH,et al.KDR expression is associated with the stage and cigarette smoking of the patients with lung cancer[J].J Cancer Res Clin Oncol,2007,133(9):635-642.
    49.Seto T,Higashiyama M,Funai H,et al.Prognostic value of expression of vascular endothelial growth factor and its fit-1 and KDR receptors in stage Ⅰnon-small-cell lung cancer[J].Lung Cancer,2006,53(1):91-96.
    50.Giatromanolaki A,Sivridis E,Brekken R,et al.The angiogenic "vascular endothelial growth factor/flk-1(KDR) receptor" pathway in patients with endometrial carcinoma:prognostic and therapeutic implications[J].Cancer,2001,92(10):2569-2577.
    51.Müller T,Choidas A,Reichmann E,et al.Phosphorylation and free pool of beta-catenin are regulated by tyrosine kinases and tyrosine phosphatases during epithelial cell migration[J].J Biol Chem,1999,274(15):10173-10183.
    52.Wu CW,Kao HL,Li AF,et al.Protein tyrosine-phosphatase expression profiling in gastric cancer tissues[J].Cancer Lett,2006,242(1):95-103.
    53.Berndt A,Luo X,B(o|¨)hmer FD,et al.Expression of the transmembrane protein tyrosine phosphatase RPTPalpha in human oral squamous cell carcinoma[J].Histochem Cell Biol,1999,111(5):399-403.
    [1]Jain S,Chakraborty G,Bulbule A,et al.Osteopontin:an emerging therapeutic target for anticancer therapy[J].Expert Opin Ther Targets,2007,11(1):81-90.
    [2]Rangaswami H,Bulbule A,Kundu GC.Osteopontin:role in cell signaling and cancer progression[J].Trends Cell Biol,2006,16(2):79-87.
    [3]E1-Tanani MK,Campbell FC,Kurisetty V,et al.The regulation and role of osteopontin in malignant transformation and cancer[J].Cytokine Growth Factor Rev,2006,17(6):463-474.
    [4]Katagiri YU,Sleeman J,Fujii H,et al.CD44 variants but not CD44s cooperate with betal-containing integrins to permit cells to bind to osteopontin independently of arginine-glycine-aspartic acid,thereby stimulating cell motility and chemotaxis[J].Cancer Res,1999,59(1):219-226.
    [5]Desai B,Rogers MJ,Chellaiah MA.Mechanisms of osteopontin and CD44 as metastatic principles in prostate cancer cells[J].Mol Cancer,2007,6:18.
    [6]Wai PY,Kuo PC.Osteopontin:regulation in tumor metastasis[J].Cancer Metastasis Rev,2008,27(1):103-118.
    [7]Bellahc(?)ne A,Castronovo V,Ogbureke KU,et al.Small integrin-binding ligand N-linked glycoproteins(SIBLINGs):multifunctional proteins in cancer[J].Nat Rev Cancer,2008,8(3):212-226
    [8]Tuck AB,Chambers AF,Allan AL.Osteopontin overexpression in breast cancer:knowledge gained and possible implications for clinical management[J].J Cell Biochem,2007,102(4):859-868.
    [9]Samant RS,Clark DW,Fillmore RA,et al.Breast cancer metastasis suppressor 1(BRMS1) inhibits osteopontin transcription by abrogating NF-kappaB activation.Mol Cancer,2007,6:6.
    [10]Vaidya KS,Welch DR.Metastasis suppressors and their roles in breast carcinoma.J Mammary Gland Biol Neoplasia,2007,12(2-3):175-190.
    [11]Lei H,Juan AH,Kim MS,et al.Identification of a Hoxc8-regulated transcriptional network in mouse embryo fibroblast cells[J].Proc Natl Acad Sci U S A,2006,103(27):10305-10309.
    [12]Rittling SR,Chambers AF.Role of osteopontin in tumour progression[J].Br J Cancer,2004,90(10):1877-1881.
    [13]Brown LF,Papadopoulos-Sergiou A,Berse B,et al.Osteopontin expression and distribution in human carcinomas[J].Am J Pathol,1994,145(3):610-623.
    [14]Kreunin P,Urquidi V,Lubman DM,et al.Identification of metastasis-associated proteins in a human tumor metastasis model using the mass-mapping technique[J].Proteomics,2004,4(9):2754-2765.
    [15]Jessen KA,Liu SY,Tepper CG,et al.Molecular analysis of metastasis in a polyomavirus middle T mouse model:the role of osteopontin[J].Breast Cancer Res,2004,6(3):R157-R169.
    [16]Denhardt DT,Mistretta D,Chambers AF,et al.Transcriptional regulation of osteopontin and the metastatic phenotype:evidence for a Ras-activated enhancer in the human OPN promoter[J].Clin Exp Metastasis,2003,20(1):77-84.
    [17]Eccles SA,Welch DR.Metastasis:recent discoveries and novel treatment strategies[J].Lancet,2007,369(9574):1742-1757.
    [18]Wise GE,King GJ.Mechanisms of tooth eruption and orthodontic tooth movement[J].J Dent Res,2008,87(5):414-434.
    [19]Ramaiah SK,Rittling S.Pathophysiological role of osteopontin in hepatic inflammation,toxicity,and cancer[J].Toxicol Sci,2008,103(1):4-13.
    [20]Wang D,You Y,Lin PC,et al.Bcl10 plays a critical role in NF-kappaB activation induced by G protein-coupled receptors[J].Proc Natl Acad Sci U S A,2007,104(1):145-150
    [21]Mi Z,Guo H,Wai PY,et al.Integrin-linked kinase regulates osteopontin-dependent MMP-2 and uPA expression to convey metastatic function in murine mammary epithelial cancer cells[J].Carcinogenesis,2006,27(6):1134-1145.
    [22]Das R,Mahabeleshwar GH,Kundu GC.Osteopontin induces AP-l-mediated secretion of urokinase-type plasminogen activator through c-Src-dependent epidermal growth factor receptor transactivation in breast cancer cells[J].J Biol Chem,2004,279(12):11051-11064.
    [23]Philip S,Kundu GC.Osteopontin induces nuclear factor kappa B-mediated promatrix metalloproteinase-2 activation through I kappa B alpha/IKK signaling pathways,and curcumin(diferulolylmethane) down-regulates these pathways[J].J Biol Chem,2003,278(16):14487-14497.
    [24]Scatena M,Almeida M,Chaisson ML,et al.NF-kappaB mediates alphavbeta3integrin-induced endothelial cell survival[J].J Cell Biol,1998,141(4):1083-1093.
    [25]Jain S,Chakraborty G,Kundu GC.The crucial role of cyclooxygenase-2 in osteopontin-induced protein kinase C alpha/c-Src/IkappaB kinase alpha/beta-dependent prostate tumor progression and angiogenesis[J].Cancer Res,2006,66(13):6638-6648.
    [26]Chakraborty G,Jain S,Behera R,et al.The multifaceted roles of osteopontin in cell signaling,tumor progression and angiogenesis[J].Curr Mol Med,2006,6(8):819-830.
    [27]Das R,Mahabeleshwar GH,Kundu GC.Osteopontin stimulates cell motility and nuclear factor kappaB-mediated secretion of urokinase type plasminogen activator through phosphatidylinositol 3-kinase/Akt signaling pathways in breast cancer cells[J].J Biol Chem,2003,278(31):28593-28606.
    [28]Rangaswami H,Bulbule A,Kundu GC.Nuclear factor-inducing kinase plays a crucial role in osteopontin-induced MAPK/IkappaBalpha kinase-dependent nuclear factor kappaB-mediated promatrix metalloproteinase-9 activation[J].J Biol Chem,2004,279(37):38921-38935.
    [29]Gao C,Mi Z,Guo H,et al.Osteopontin regulates ubiquitin-dependent degradation of Statl in murine mammary epithelial tumor cells[J].Neoplasia,2007,9(9):699-706.
    [30]McAllister SS,Gifford AM,Greiner AL,et al.Systemic endocrine instigation of indolent tumor growth requires osteopontin[J].Cell,2008,133(6):994-1005.
    [31]Hong H,McCullough CM,Stegemann JP.The role of ERK signaling in protein hydrogel remodeling by vascular smooth muscle cells[J].Biomaterials,2007,28(26):3824-3833.
    [32]Sun BS,Dong QZ,Ye QH,et al.Lentiviral-mediated miRNA against osteopontin suppresses tumor growth and metastasis of human hepatocellular carcinoma[J].Hepatology,2008,48(6):1834-1842.

© 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