SiRNA干扰β-catenin信号通路在肝癌治疗中的作用及机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
原发性肝癌是世界上最常见的十种恶性肿瘤之一,我国又是原发性肝癌的高发区。一旦发现多为中晚期,失去了手术机会,现有的治疗方法效果亦不理想。因此,长久以来肝癌的研究一直为基础和临床科研人员所关注。
     β-catenin是细胞质中一种重要的多功能蛋白,处于Wnt信号通路的活化中心,与形成肿瘤的多种因素密切相关,参与肿瘤发生、发展过程的多个环节。因此,本研究欲应用siRNA干扰β-catenin信号传导通路,以观察其对肝癌细胞生长、细胞周期、细胞凋亡相关因子、血管生成因子、端粒酶及其它信号通路的影响及siRNA能否用于肝癌的治疗。
     首先筛选能够有效沉默β-catenin基因表达的siRNA,然后用有效的siRNA转染入肝癌HepG2细胞,在不同的时间点用RT-PCR及western blot法检测β-catenin的mRNA及蛋白质表达,MTT法检测肝癌细胞的生长,72h和96h用流式细胞仪检测细胞周期。最后用western blot法检测细胞周期、细胞凋亡、血管生成因子、端粒酶及其它信号通路部分相关蛋白质的表达,用RT-PCR法检测VEGF-A、VEGF-C的mRNA表达。从而可知siRNA可以沉默β-catenin基因及蛋白质的表达,并使细胞周期阻滞于G0/G1期。一次转染可在一定时间内抑制肝癌细胞的生长,在肝癌的治疗中起一定的作用,但不能治愈肝癌。肝癌细胞中β-catenin信号传导通路可以调节肝癌的细胞生长、细胞周期、细胞凋亡、血管生成因子、端粒酶及其它信号通路等,从而抑制β-catenin信号通路在肝癌的治疗中起一定的作用。
     我们需要进一步深入研究,以了解肝癌的发生、发展过程,从而寻找更为有效的治疗肝癌的方法。
Hepatocellular carcinoma (HCC) is one of ten most common malignant tumors in the world and our country is its high incidence. HCC has the hidden genesis, the rapid development and the high malignance. Once it is found, it is in the late stage and the patient loses the chance of operation. The current treatment is also not satisfactory. Therefore, the study of HCC is always concerned by the basic and clinical researches for a long time.β-catenin, as a multifunctional protein in cytoplasm, is close to many factors that form tumors and participates in a lot of steps of tumor genesie and development. The current researches indicate that there are the abnormal expressions of cell cycle, apoptosis, angiogenesis, telomerase, signaling pathway, and so on, in HCC, but the relation betweenβ-catenin and these factors is not clear.
     Based on the above findings, in HepG2 cells, we silenceβ-catenin gene and observe the change of cell growth, cell cycle, apoptosis, angiogenesis, telomerase and signaling pathway, thereby studying the relation betweenβ-catenin and these factors and validating theβ-catenin role on HCC genesis and development and the siRNA role on HCC gene therapy. These will offer the basis of theory for HCC pathogenesis and therapy so that people may better study and treat HCC.
     (1) Bolting effective siRNA againstβ-catenin
     In our studies, after the siRNA marked by fluorescence was transfected into the HepG2 cells, the transfection efficiency was detected. The different siRNA againstβ-catenin was teansfected into the HepG2 cells,β-catenin protein expression was detected by western blot at 48 and 72h. We drew a conclusion that the first siRNA againstβ-catenin was the most effective. After the first siRNA againstβ-catenin was transfected into HepG2 cells, the mRNA and protein expressions ofβ-catenin were detected respectively by RT-PCR and western blot.β-catenin mRNA expression was most obviously inhibited at 24 h and increased gradually at 48, 72 and 96 h after the transfection.β-catenin protein expression decreased gradually at 24, 48 and 72 h and slightly increased at 96 hours after the transfection. These proved that the first siRNA againstβ-catenin could specifically silenceβ-catenin expression again. And we studied its time-varying pattern, thereby selecting 72 and 96h as the point time of future experiments. These lay a solid foundation for future experiments.
     (2) The influence of silencingβ-catenin on HCC cell growth
     In our studies, after the siRNA againstβ-catenin was transfected into the HepG2 cells, we observed the cell shape and measured HCC cell growth by MTT assay. After the transfection, the cell growth was inhibited at 24, 48 and 72 h and the weakened inhibition was manifested by slightly increased rate of cell growth at 96 h. Therefore, Only one time transient transfection may temporarily inhibit HCC cell growth, which palys a certain role in the therapy of HCC, but may not cure HCC. We will further study if multiple transfections or stable transfection would work as a powerful tool to cure HCC in the future. Our results also suggest that other signaling pathways may have some functions to compensate the inhibition ofβ-catenin signaling pathway. It needs to be further study in the future.
     (3) The influence of silencingβ-catenin on HCC cell cycle
     In our studies, after the siRNA againstβ-catenin was transfected into the HepG2 cells, HCC cell cycle was detected by flow cytometry and the protein expression of cyclin A and cyclin E was detected by western blot. After the transfection, the cell cycle was blocked in G0/G1 phase at 72 h and was slightly returned and proceeded to M phase at 96 h. HCC cells began to multiplied again at 96 hours. The result was similar to that of MTT. After the transfection, the protein expression of cyclin A and cyclin E increased at 72 h and decreased at 96 h, so we may know that they are related toβ-catenin signaling pathway and are also related to other factors, andβ-catenin may regulate cell growth through affecting cell cycle.
     (4) The influence of silencingβ-catenin on HCC apoptosis
     In our studies, after the siRNA againstβ-catenin was transfected into the HepG2 cells, the protein expression of apoptosis-related factors (caspase-3, XIAP, Grp78 and HSP27) was detected by western blot. The protein expression of phosphorylated (p)-caspase-3 and Grp78 increased at 72h and decreased at 96h. The protein expression of caspase-3 and XIAP decreased at 72h and increased at 96 h. The HSP27 protein expression showed no change following transfection. These indicate that Wnt/β-catenin signaling pathway can regulate the expression of these proteins (caspase-3,XIAP,Grp78) and the latter is downstream target proteins of the former. Wnt/β-catenin isn’t related to the HSP27 protein expression. In short, the Wnt/β-catenin signaling pathway contributes to HCC apoptosis, proliferation, and differentiation through regulation of the expression of angiogenesis factors.
     (5) The influence of silencingβ-catenin on HCC angiogenesis
     In our studies, after the siRNA againstβ-catenin was transfected into the HepG2 cells, the protein expression of angiogenesis factors (MMP-2, MMP-9, VEGF-A, VEGF-C and bFGF) was detected by western blot and the mRNA expression of VEGF-A and VEGF-C was detected by RT-PCR. The protein expression of MMP-2, MMP-9, VEGF-A, VEGF-C and bFGF decreased at 72h and increased at 96 h. The mRNA expression of VEGF-A and VEGF-C is similar to the protein expression. These indicate that Wnt/β-catenin signaling pathway can regulate the expression of these proteins and the latter is downstream target proteins of the former. In short, the Wnt/β-catenin signaling pathway contributes to HCC angiogenesis, infiltration and metastasis through regulation of the expression of angiogenesis factors.
     (6) The influence of silencingβ-catenin on HCC telomerase and other signaling pathway
     In our studies, after the siRNA againstβ-catenin was transfected into the HepG2 cells, the protein expression of telomerase (TERT) and other signaling pathways (MAPK family, Akt1, Smad3, STAT3 and GSK-3β) was detected by western blot. ERK1/2 (p44/42MAPK), JNK/SAPK, p38MAPK, and Akt1 protein levels showed no change following transfection, while their phosphorylated protein levels showed changes. After the transfection, p-ERK1/2 (p-p42MAPK) protein expression increased gradually at 72 and 96h; expression of p-JNK/SAPK protein 54kD form decreased while levels of the 46kD form gradually increased at 72 and 96h; no p-p38MAPK protein was observed in HepG2 cells; p-Akt1 protein expression decreased at 72h and increased again slightly at 96h. The protein expression of TERT decreased at 72h and increased at 96 h. The protein expression of Smad3 increased at 72h and decreased at 96h. The STAT3 protein expression showed no change following transfection. GSK-3βand p-GSK-3βprotein expression increased gradually at 72 and 96 h. Thus,β-catenin signaling pathway may affect Ras/MAPK and PI3K/Akt signaling pathways through regulating the phosphorylation state of ERK1/2, JNK/SAPK and Akt1 protein in HepG2 cells. In addition, the Wnt/β-catenin signaling pathway may participate in HCC genesie and development through regulating the protein expression of GSK-3β, p-GSK-3β, Smad3, TERT. These signaling pathways activate each other, thereby form an intricate network.
     In summary, siRNA may silence the mRNA and protein expression ofβ-catenin and block the cell cycle in G0/G1 phase. Only one time transient transfection may temporarily inhibit HCC cell growth, which palys a certain role in the therapy of HCC, but may not cure HCC.β-catenin signaling pathway is related to the protein expression of cyclin A, cyclin E, caspase-3, XIAP, Grp78, MMP-2, MMP-9, VEGF-A, VEGF-C, bFGF, TERT and Smad3, while it is not related to HSP27 and STAT3 protein expression in HCC.β-catenin signaling pathway may affect Ras/MAPK and PI3K/Akt signaling pathways through regulating the phosphorylation state of ERK1/2, JNK/SAPK and Akt1 protein in HepG2 cells. These pathways andβ-catenin pathway might regulate some downstream factors in common, thereby compensating for the inhibitory effect ofβ-catenin and affecting tumor cell growth and others factors.
     In short,β-catenin signaling pathway may contribute to affect HCC cell growth, cell cycle, apoptosis, angiogenesis, telomerase and other signaling pathway through regulating these factors, so silencingβ-catenin may play a certain role on the therapy of HCC. This pathway also might be involved in HCC genesis and development through regulating these factors. Only make the processes and interrelations of the signaling pathway clear, we can understand tumor genesis, development, prognosis and prevention and control cell growth, differentiation and apoptosis to achieve to cure the tumor through interfering the signaling pathway.
引文
[1] FRAZIER ME, JOHNSON GM, THOMASSEN DG, et al. Realizing the potential of the genome revolution: the genomes to life program. Science, 2003, 300 (5617): 290-293.
    [2] BREMBECK FH, ROSARIO M, BIRCHMEIER W. Balancing cell adhesion and Wnt signaling, the key role of beta-catenin. Curr Opin Genet Dev, 2006 Feb, 16(1): 51-59.
    [3] LEE HC, KIM M, WANDS JR. Wnt/Frizzled signaling in hepatocellular carcinoma. Front Biosci, 2006, 11: 1901-1915.
    [4] PENNISI E. How a growth control path takes a wrong turn to cancer. Science, 1998, 281: 1438-1439.
    [5] LEE HS, PARK MH, YANG SJ, et al. Novel candidate targets of Wnt/beta-catenin signaling in hepatoma cells. Life Sci, 2007, 80: 690-698.
    [6] MARTENSSON A, OBERG A, JUNG A,et al. Beta-catenin expression in relation to genetic instability and prognosis in colorectal cancer. Oncol Rep, 2007, 17: 447-452.
    [7] FUCHS SY, OUGOLKOV AV, SPIEGELMAN VS, et al. Oncogenic beta-catenin signaling networks in colorectal cancer. Cell Cycle, 2005, 4: 1522-1539.
    [8] KOBAYASHI M, HONMA T, MATSUDA Y, et al. Nuclear translocation of beta-catenin in colorectal cancer. Br J Cancer, 2000, 82: 1689-1693.
    [9] OZAKI S, IKEDA S, ISHIZAKI Y, et al. Alterations and correlations of the components in the Wnt signaling pathway and its target genes in breast cancer. Oncol Rep, 2005, 14: 1437-1443.
    [10] OGASAWARA N, TSUKAMOTO T, MIZOSHITA T, et al. Mutations and nuclear accumulation of beta-catenin correlate with intestinal phenotypic expression in human gastric cancer. Histopathology, 2006, 49: 612-621.
    [11] SETO ES, BELLEN HJ. The ins and outs of Wingless signaling. Trends Cell Biol, 2004, 14: 45-53.
    [12] KARIM R, TSE G, PUTTI T, et al. The significance of the Wnt pathway in the pathology of human cancers. Pathology, 2004, 36: 120-128.
    [13] TERAI S, OKITA K. Dose cytoplasmic cyclin D1 regulate the cell cycle directly? J Gastroenterol, 1999 Aug, 34(4): 547-548.
    [14] MOLENAAR JJ, EBUS ME, KOSTER J, et al. Cyclin D1 and CDK4 activity contribute to the undifferentiated phenotype in neuroblastoma. Cancer Res, 2008 Apr15, 68(8): 2599-2609.
    [15] AALTONEN K, AMINI RM, LANDBERG G, et al. Cyclin D1 expression is associated with poor prognostic features in estrogen receptor positive breast cancer. Breast Cancer Res Treat, 2008 Feb 1.[Epub ahead of print]
    [16] HARA A, SAEGUSA M, ICHINOE M, et al. Diagnostic and prognostic significance of cyclin A expression in low-grade astrocytomas: comparison with astrogliosis and high-grade tumours. J Clin Pathol, 2008 Mar, 61(3): 287-292.
    [17] MELCK A, MASOUDI H, GRIFFITH OL, et al. Cell cycle regulators show diagnostic and prognostic utility for differentiated thyroid cancer. Ann Surg Oncol, 2007 Dec, 14(12): 3403-3411.
    [18] VAN DE WETERING M, SANCHO E, VERWEIJ C, et al. Theβ-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell, 2002, 111(2): 241-250.
    [19] SANGKHATHAT S, KUSAFUKA T, MIAO J, et al. In vitro RNA interference against beta-catenin inhibits the proliferation of pediatric hepatic tumors. Int J Oncol, 2006, 28(3):715-722.
    [20] BISSONNETTE M, KHARE S, VON LINTIG FC, et al. Mutation and nonmutational activation of p21 ras in rat colonic azoxymethane-induced tumors: effects on mitogen-activated protein kinase, cyclooxygenase-2, and cyclinD1. Cancer Res, 2000, 60: 4602-4609.
    [21] TETSU O, MCCORMICK F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature, 1999, 398: 422-426.
    [22] PIERCE M, WANG C, STUMP M, et al. Overexpression of the beta-catenin binding domain of cadherin selectively kills colorectal cancer cells. Int. J. Cancer, 2003, 107: 229-237.
    [23] MOSSER DD, MORIMOTO RI. Molecular chaperones and the stress of oncogenesis. Oncogene, 2004, 23(16): 2907-2918.
    [24] SCHEPERS H, GEUGIEN M, VAN DER TOORN M, et al. HSP27 protects AML cells against VP-16-induced apoptosis through modulation of p38 and c-Jun. Exp Hematol, 2005, 33(6): 660-670.
    [25] LUSTIG B, BEHRENS J. The Wnt signaling pathway and its role in tumor development. J Cancer Res Clin Oncol, 2003, 129: 199-221.
    [26] NAKAMOTO K, KURATSU J, OZAWA M. Beta-catenin cleavage in non-apoptoticcells with reduced cell adhesion activity. Int J Mol Med, 2005 Jun, 15(6): 973-979.
    [27] CERVELLO M, GIANNITRAPANI L, LA ROSA M, et al. Induction of apoptosis by the proteasome inhibitor MG132 in human HCC cells: Possible correlation with specific caspase-dependent cleavage of beta-catenin and inhibition of beta-catenin- mediated transactivation. Int J Mol Med, 2004 May, 13(5): 741-748.
    [28] POMA P, NOTARBARTOLO M, LABBOZZETTA M, et al. Antitumor effects of the novel NF-kappaB inhibitor dehydroxymethyl-epoxyquinomicin on human hepatic cancer cells: analysis of synergy with cisplatin and of possible correlation with inhibition of pro-survival genes and IL-6 production. Int J Oncol, 2006 Apr, 28(4): 923-930.
    [29] WADA A, YOKOO H, YANGITA T, et al. Lithium: potential therapeutics against acute brain injuries and chronic neurodegenerative diseases. J Pharmacol Sci, 2005 Dec, 99(4): 307-321.
    [30] FANELLI MA, MONTT-GUEVARA M, DIBLASI AM, et al. P-Cadherin and beta-catenin are useful prognostic markers in breast cancer patients; beta-catenin interacts with heat shock protein Hsp27. Cell Stress Chaperones, 2008 Summer, 13(2): 207-220.
    [31] TAKAHASHI H, SHIBUYA M. The vascular endothelial growth factor (VEGF) /VEGF receptor system and its role under physiological and pathological conditions. Clin Sci (Lond), 2005, 109(3): 227-241.
    [32] CARMELIET P. VEGF as a key mediator of angiogenesis in cancer. Oncology, 2005, 69(Suppl 3): 4-10.
    [33] HE Y, KARPANEN T, ALITALO K. Role of lymphangiogenic factors in tumor metastasis. Biochim Biophys Acta, 2004, 1654(1): 3-12.
    [34] ROEB E, MATERN S. Matrix metalloproteinases and colorectal cancer. Med Klin (Munich), 2003, 98: 763-770.
    [35] LIU HL, LI XH, WANG DY, et al. Matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-1 expression in fibrotic rat liver. World J Gastroenterol, 2000, 6: 881-884.
    [36] CURRAN S, MURRAY GI. Matrix metalloproteinase: molecular aspects of their roles in tumor invasion and metastasis. Eur J Cancer, 2000, 36: 1621-1630.
    [37] WAAS ET, LOMME RM, DEGROOT J, et al. Tissue levels of active matrix metalloproteinase-2 and -9 in colorectal cancer. Br J Cancer, 2002, 86: 1876-1883.
    [38] KLEINER DE, STETLER-STEVENSON WG. Matrix metalloproteinases and metastasis. Cancer Chemother Pharmacol, 1999, 43(Suppl): S42-51.
    [39] JI F, WANG WL, YANG ZL, et al. Study on the expression of matrix metallo proteinase-2 mRNA in human gastric cancer. World J Gastroenterol, 1999, 5: 455-457.
    [40] SAKAMOTO Y, MAFUNE K, MORI M, et al. Overexpression of MMP-9 correlates with growth of small hepatocellular carcinoma. Int J Oncol, 2000, 17: 237-243.
    [41] MAATTA M, SOINI Y, LIAKKA A, et al. Differential expression of matrix metalloproteinase (MMP)-2, MMP-9, and membrane type 1-MMP in hepatocellular and pancreatic adenocarcinoma: implications for tumor progression and clinical prognosis. Clin Cancer Res, 2000, 6: 2726-2734.
    [42] KUYVENHOVEN JP, VAN HOEK B, BLOM E, et al. Assessment of the clinical significance of serum matrix metalloproteinases MMP-2 and MMP-9 in patients with various chronic liver diseases and hepatocellular carcinoma. Thromb Haemost, 2003, 89: 718-725.
    [43] YAMAMOTO H, ITOH F, ADACHI Y, et al. Relation of enhanced secretion of active matrix metalloproteinases with tumor spread in human hepatocellular carcinoma. Gastroenterology, 1997, 112: 1290-1296.
    [44] ZHANG SC, WERNIG M, DUNCAN ID, et al. In vitro differentiation of transplatable neural precursors from human embryonic stem cells. Nat Biotechnology, 2001, 19(12): 1129-1133.
    [45] LIU J, OPOPARI AW, TAN L, et al. Dysfunctional apoptosome activation in ovarian cancer: implications for chemoresistance. Cancer Res, 2002, 2(62): 924-931.
    [46] OKADA BM, THIERY JP, JOUANNEAU J. Fibroblast growth factor-2. Int J Biochem, 2000, 32(3): 263-267.
    [47] SONG S, WIENTJES M, GAN Y, et al. Fibroblast growth factors: an epigenetic mechanism of broad spectrum resistance to anticancer drugs. Proc Natl Acad Sci USA, 2000, 97(15): 8658-8663.
    [48] CALVIELLO G, RESCI F, SERINI S, et al. Docosahexaenoic acid induces proteasome-dependent degradation of beta-catenin, down-regulation of survivin and apoptosis in human colorectal cancer cells not expressing COX-2. Carcinogenesis, 2007 Jun, 28(6): 1202-1209.
    [49] HAHN JY, CHO HJ, BAE JW, et al. Beta-catenin overexpression reduces myocardial infarct size through differential effects on cardiomyocytes and cardiac fibroblasts. JBiol Chem, 2006 Oct 13, 281(41): 30979-30989.
    [50] EASWARAN V, LEE SH, INGE L, et al. Beta-Catenin regulates vascular endothelial growth factor expression in colon cancer. Cancer Res, 2003, 63(12): 3145-3153.
    [51] EGEBALD M, WERB Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer, 2002, 2: 161-174.
    [52] WU B, CRAMPTON SP, HUGHES CC. Wnt signaling induces MMP expression and regulates T cell transmigration. Immunity, 2007 Feb, 26(2): 227-239.
    [53] SAEKI H, TANAKA S, SUGIMACHI K, et al. Interrelation between expression of matrix metalloproteinase 7 and beta-catenin in esophageal cancer. Dig Dis Sci, 2002, 47(12): 2738-2742.
    [54] OUGOLKOV AV, YAMASHITA K, MAI M, et al. Oncogenicβ-catenin and MMP-7 (matrilysin) cosegregate in late-stage clinical colon cancer. Gastroenterology, 2002, 122(1): 60-71.
    [55] THIRUNAVUKKARASU M, HAN Z, ZHAN L, et al. Adeno-sh-beta-catenin abolishes ischemic preconditioning-mediated cardioprotection by downregulation of its target genes VEGF, Bcl-2, and survivin in ischemic rat myocardium. Antioxid Redox Signal, 2008 Aug, 10(8): 1475-1484.
    [56] KAGA S, ZHAN L, ALTAF E, et al. Glycogen synthase kinase-3 beta/ beta-catenin promotes angiogenic and anti-apoptotic signaling through the induction of expression in rat ischemic preconditioned myocardium. J Mol Cell Cardiol, 2006 Jan, 40(1): 138-147.
    [57] SUYAMA K, SHAPIRO I, GUTTMAN M, et al. A signaling pathway leading to metastasis is controlled by N-cadherin and the FGF receptor. Cancer Cell, 2002, 2(4): 301-314.
    [58] HAJIME K, CAO RH, BRAKENHIELM E, et al. Blockade of vascular endothelial growth factor receptor-3 signaling inhibits fibroblasts growth factor-2-induced lymphangiogenesis in mouse cornea. Proc Natl Acad Sci USA, 2002, 99(13): 8868-8873.
    [59] DEMIRKESEN C, BUYUKPINARBSSILI N, RAMAZANOGLU R, et al. The correlation of angiogenesis with metastasis in primary cutaneous melanoma: a comparative analysis of microvessel density, expression of vascular endothelial growth factor and basic fibroblastic growth factor. Pathology, 2006, 38(2): 132-137.
    [60] BLASCO MA, LEE HW, HAMDE MP, et al. Telomere shortening and tumorformation by mouse cells lacking telomerase RNA. Cell, 1997, 91: 25-34.
    [61] PERRY PJ, ARNOLD JR, JENKINS TC. Telomerase inhibitors for the treatment of cancer: the current perspective. Expert Opin Investig Drugs, 2001, 10(12): 2141-2156.
    [62] DING A, TONG SX, FENG P, et al. The expression and clinical significance of hTERT andβ-catenin in gallbladder carcinoma. Fudan Univ J Med Sci, 2006, 33(1): 44-48.
    [63] CHENG YW, WU TC, CHEN CY, et al. Human telomerase reverse transcriptase activated by E6 oncoprotein is required for human papillomavirus-16/18-infected lung tumorigenesis. Clin Cancer Res, 2008 Nov 15, 14(22): 7173-7179.
    [64] WADA T, PENNINGER JM. Mitogen-activated protein kinases in apoptosis regulation. Oncogene, 2004, 23(16): 2838-2849.
    [65] SEGER R, KREBS EG. The MAPK signaling cascade. FASEB J, 1995, 9(9): 726-735.
    [66] KOBEL M, POHL G, SCHMITT WD, et al. Activation of mitogen-activated protein kinase is required for migration and invasion of placental site trophoblastic tumor. Am J Pathol, 2005, 167(3): 879-885.
    [67] ATSUMI T, KATO K, UNO K, et al. Pathophysiological role of the activation of p38 mitogen-activated protein kinases in poorly differentiated gastric cancer. Pathol Int, 2007 Oct, 57(10): 635-644.
    [68] LIANG B, WANG S, ZHU XG, et al. Increased expression of mitogen-activated protein kinase and its upstream regulating signal in human gastric cancer. World J Gastroenterol, 2005 Feb 7, 11(5): 623-628.
    [69] BESSARD A, FREMIN C, EZAN F, et al. RNAi-mediated ERK2 knockdown inhibits growth of tumor cells in vitro and in vivo. Oncogene, 2008 Sep 11, 27(40): 5315-5325.
    [70] SHAMA J, GARCIA-MEDINA R, POUYSSEGUR J, et al. Major contribution of MEK1 to the activation of ERK1/ERK2 and to the growth of LS174T colon carcinoma cells. Biochem Biophys Res Commun, 2008 Aug 8, 372(4): 845-849.
    [71] MOUNTZIOS G, PLANCHARD D, BESSE B, et al. Mitogen-activated protein kinase activation in lung adenocarcinoma: a comparative study between ever smokers and never smokers. Clin Cancer Res, 2008 Jul 1, 14(13): 4096-4102.
    [72] ANDERSON LM, FORNWALD LW. Cell cycle activation in lung adenocarcinoma cells by the ErbB3/phosphatidylinositol 3-kinase/Akt pathway. Carcinogenesis, 2003, 24: 1581-1592.
    [73] NAKANISHI K, SAKAMOTO M, YAMASAKI S, et al. Akt phosphorylation is a riskfactor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer, 2005 Jan 15, 103(2): 307-312.
    [74] MICHL P, DOWNWARD J. Mechanisms of disease: PI3K/AKT signaling in gastrointestinal cancers. Z Gastroenterol, 2005 Oct, 43(10): 1133-1139.
    [75] BLEEKER FE, FELICIONI L, BUTTITTA F, et al. AKT1(E17K) in human solid tumours. Oncogene, 2008 May 26. [Epub ahead of print]
    [76] SOUNG YH, LEE JW, NAM SW, et al. Mutational analysis of AKT1, AKT2 and AKT3 genes in common human carcinomas. Oncology, 2006, 70(4): 285-289.
    [77] ZHU L, HU C, LI J, et al. Real-time imaging nuclear translocation of Akt1 in HCC cells. Biochem Biophys Res Commun, 2007 May 18, 356(4): 1038-1043.
    [78] KIM SG, KIM HA, JONG SH, et al. The Endogenous Ratio of Smad2 and Smad3 Influences the Cytostatic Function of Smad3. Molecular Biology of the Cell, 2005, 16(10): 4672-4683.
    [79] NAKASHIMA K, YANAGISAWA M, ARAKAWA H, et al. Synergistic signaling in fetal brain by STAT3-smad1 complex bridged by p300. Science, 1999, 284: 479-482.
    [80] LLABBE E, LETAMENDIA A, ATTISANO L. Association of Smads with lymphoid enhancer binding factor 1/T cell specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathway. Proc Natl Acad Sci (USA), 2000, 97: 8358-8363.
    [81] BUETTNER R, MORA L B, JOVE R. Activated STAT signaling in human tumors provides novel molecular targets for therapeutic Intervention. Clin Cancer Res, 2002, 8(4): 945-954.
    [82] CHUNG H, SEO S, MOON M, et al. Phosphatidylinositol-3-kinase/Akt/glycogen synthase kinase-3 beta and ERK1/2 pathways mediate protective effects of acylated and unacylated ghrelin against oxygen-glucose deprivation-induced apoptosis in primary rat cortical neuronal cells. J Endocrinol, 2008 Sep, 198(3): 511-521.
    [83] SUMI T, TSUNEYOSHI N, NAKATSUJI N, et al. Defining early lineage specification of human embryonic stem cells by the orchestrated balance of canonical Wnt/beta-catenin, Activin/Nodal and BMP signaling. Development, 2008 Sep, 135(17): 2969-2979.
    [84] LABBE E, LOCK L, LETAMENDIA A, et al. Transcriptional cooperation between the transforming growth factor-beta and Wnt pathways in mammary and intestinal tumorigenesis. Cancer Res, 2007 Jan 1, 67(1): 75-84.
    [85] DENNIS EA. Diversity of group types, regulation, and function of phospholipase A2. J Biol Chem, 1994, 269(18): 13057-13060.
    [86] LUU HH, ZHANG R, HAYDON RC, et al. Wnt/beta-catenin signaling pathway as a novel cancer drug target. Curr Cancer Drug Targets, 2004, 4: 653-671.
    [87] DIHLMANN S, VON KNEBEL DOEBERITZ M. Wnt/β-catenin-pathway as a molecular target for future anti-cancer therapeutics. Int J Cancer, 2005, 113: 515-524.
    [88] KUNDU JK, CHOI KY, SURH YJ. Beta-Catenin mediated signaling: a novel molecular target for chemoprevention with anti-inflammatory substances. Biochim Biophys Acta, 2006, 1765: 14-24.
    [89] DIHLMANN S, KLEIN S, DOEBERITZ MV MK. Reduction of beta-catenin/T-cell transcription factor signaling by aspirin and indomethacin is caused by an increased stabilization of phosphorylated beta-catenin. Mol Cancer Ther, 2003, 2: 509-516.
    [90] AGRAWAL N, DASARADHI PV, MOHMMED A, et al. RNA interference: biology, mechanism, and applications. Microbiol Mol Biol Rev, 2003, 67: 657-685.
    [91] LIPARDI C, WEI Q, PATERSON BM. RNAi as random degradative PCR: siRNA primers convert mRNA into dsRNAs that are degraded to generate new siRNAs. Cell, 2001, 107: 297-307.
    [92] HUTVAGNER G, MCLACHLAN J, PASQUINELLI AE, et al. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science, 2001, 293: 834-838.
    [93] BASS BL. Double-stranded RNA as a template for gene silencing. Cell, 2000, 101: 235-238.
    [94] ZAMORE PD, TUSCHL T, SHARP PA, et al. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 2000, 101: 25-33.
    [95] ELBASHIR SM, LENDECKEL W, TUSCHL T. RNA interference is mediated by 21-and 22-nucleotide RNAs. Genes Dev, 2001, 15(2): 188-200.
    [96] BERSTEIN E, CAUDY AA, HAMMORD SM, et al. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature, 2001, 409: 363-366.
    [97] CERUTTI H. RNA interference: traveling in the cell and gaining functions. Trends Genet, 2003, 19: 39-46.
    [98] MOURRAIN P, BECLIN C, ELMAYAN T, et al. Arabidopsis SGS2 and SGS3 genes are required for posttranscriptional gene silencing and natural virus resistance. Cell,2000, 101: 533-542.
    [99] BRANTL S. Antisense-RNA regulation and RNA interference. Biochim Biophys Acta, 2002, 1575: 15-25.
    [100] BERNSTEIN E, KIM SY, CARMELL MA, et al. Dicer is essential for mouse development. Nat Genet, 2003, 35: 215-217.
    [101] HALL IM, NOMA K, GREWAL SI. RNA interference machinery regulates chromosome dynamics during mitosis and meiosis in fission yeast. Proc Natl Acad Sci USA, 2003, 100: 193-198.
    [102] CARRINGTON JC, AMBROS V. Role of microRNAs in plant and animal development. Science, 2003, 301: 336-338.
    [103] BORKHARDT A. Blocking oncogenes in malignant cells by RNA interference-new hope for a highly specific cancer treatment. Cancer Cell, 2002, 2: 167-168.
    [104] ELBASHIR SM, HARBORTH J, WEBER K, et al. Analysis of gene function in somatic mammalian cells using small interfering RNAs. Methods, 2002, 26: 199-213.
    [105] DAVENPORT RJ. Gene silencing: A faster way to shut down genes. Science, 2001, 292: 1469-1471.
    [106] DUXBURY MS, WHANG EE. RNA interference: A Practical Approach. J Surg Res, 2004, 117: 339-344.
    [107] HREBORTH J, ELBASHIR SM, VANDENBURGH K, et al. Sequence, chemical, and structural variation of small interfering RNAs and short hairpin RNAs and the effect on mammalian gene silencing. Antisense Nucleic Acid Drug Dev, 2003, 13: 83-105.
    [108] SHIRATSUCHI H, NAKASHIMA T, HIRAKAWA N, et al. beta-Catenin nuclear accumulation in head and neck mucoepidermoid carcinoma: Its role in cyclin D1 overexpression and tumor progression. Head Neck, 2007 Jun, 29(6): 577-584.
    [109] VERMA UN, SURABHI RM, SCHMALTIEG A, et al. Small interfering RNAs directed against beta-Catenin inhibit the in vitro and in vivo growth of colon cancer cells. Clin Cancer Res, 2003, 9: 1291-1300.
    [110] HU L, SHI J, WANG L. Down-regulation of beta-catenin nuclear localization by aspirin correlates with growth inhibition of Jurkat cell line. J Huazhong Univ Sci Technolog Med Sci, 2006, 26(6): 731-734.
    [111] WANG L, SHAO ZM. Cyclin E expression and prognosis in breast cancer patients: a meta-analysis of published studies. Cancer Invest, 2006 Oct, 24(6): 581-587.
    [112] YUE H, JIANG HY. Expression of cell cycle regulator p57kip2, cyclin E protein andproliferating cell nuclear antigen in human pancreatic cancer: an immunohistochemical study. World J Gastroenterol, 2005 Aug 28, 11(32): 5057-5060.
    [113] MUSSMAN JG, HORN HF, CARROLL DE, et al. Synergistic induction of centrosome hyperamplification by loss of p53 and cyclin E overexpression. Oncogene, 2000 Mar 23, 19(13): 1635-1646.
    [114] COVERLEY D, LAMAN H, LASKEY RA. Distinct roles for cyclins E and A during DNA replication complex assembly and activation. Nat Cell Biol, 2002, 4(7): 523-528.
    [115] RSSIG L, JADIDI AS, URBICH C, et al. Akt-dependent phosphorylation of p21Cip1 regulates PCNA binding and proliferation of endothelial cells. Mol Cell Biol, 2001, 21(16): 5644-5657.
    [116] MORAN DM, MATTOCKS MA, CAHILL PA, et al. Interleukin-6 Mediates G(0)/G(1) Growth Arrest in Hepatocellular Carcinoma Through a STAT 3-Dependent Pathway. J Surg Res, 2007 Jun 13. [Epub ahead of print].
    [117] DAN HC, SUN M, KANEKO S, et al. Akt phosphorylation and stabilization of X-linked inhibitor of apoptosis protein (XIAP). J Biol Chem, 2004 Feb 13, 279(7): 5405-5412.
    [118] CARTER BZ, MILELLA M, TSAO T, et al. Regulation and targeting of antiapoptotic XIAP in acute myeloid leukemia. Leukemia, 2003 Nov, 17(11): 2081-2089.
    [119] LIN H, CHEN C, LI X, et al. Activation of the MEK/MAPK pathway is involved in bryostatin1-induced monocytic differenciation and up-regulation of X-linked inhibitor of apoptosis protein. Exp Cell Res, 2002 Jan 15, 272(2): 192-198.
    [120] HUANG S, ZHANG F, MIAO L, et al. Lentiviral-mediated Smad4 RNAi induced anti-proliferation by p16 up-regulation and apoptosis by caspase 3 down-regulation in hepatoma SMMC-7721 cells. Oncol Rep, 2008 Nov, 20(5): 1053-1059.
    [121] NGUYEN TH, MUSTAFA FB, PERVAIZ S, et al. ERK1/2 activation is required for resveratrol-induced apoptosis in MDA-MB-231 cells. Int J Oncol, 2008 Jul, 33(1): 81-92.
    [122] TSUCHIYA T, TSUNO NH, ASAKAGE M, et al. Apoptosis induction by p38 MAPK inhibitor in human colon cancer cells. Hepatogastroenterology, 2008 May-Jun, 55(84): 930-935.
    [123] RAMSAY RG, CIZNADIJA D, MANTAMADIOTIS T, et al. Expression of stress response protein glucose regulated protein-78 mediated by c-Myb. Int J Biochem Cell Biol, 2005 Jun, 37(6): 1254-1268.
    [124] SKANDRANI D, GAUBIN Y, BEAU B, et al. Effect of selected insecticides on growth rate and stress protein expression in cultured human A549 and SH-SY5Y cells. Toxicol In Vitro, 2006 Dec, 20(8): 1378-1386.
    [125] WEI D, LE X, ZHENG L, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene, 2003 Jan 23, 22(3): 319-329.
    [126] QIU Z, HUANG C, SUN J, et al. RNA interference-mediated signal transducers and activators of transcription 3 gene silencing inhibits invasion and metastasis of human pancreatic cancer cells. Cancer Sci, 2007 Jul, 98(7): 1099-1106.
    [127] TSAREVA SA, MORIGGL R, CORVINUS FM, et al. Signal Transducer and Activator of Transcription 3 Activation Promotes Invasive Growth of Colon Carcinomas through Matrix Metalloproteinase Induction. Neoplasia, 2007 April, 9(4): 279-291.
    [128] XIE TX, HUANG FJ, ALDAPE KD, et al. Activation of stat3 in human melanoma promotes brain metastasis. Cancer Res, 2006 Mar 15, 66(6): 3188-3196.
    [129] DING Y, BOGUSLAWSKI EA, BERGHUIS BD, et al. Mitogen-activated protein kinase kinase signaling promotes growth and vascularization of fibrosarcoma. Mol Cancer Ther, 2008 Mar, 7(3): 648-658.
    [130] ZHANG YH, WEI W, XU H, et al. Inducing effects of hepatocyte growth factor on the expression of vascular endothelial growth factor in human colorectal carcinoma cells through MEK and PI3K signaling pathways. Chin Med J (Engl), 2007 May 5, 120(9): 743-748.
    [131] ADYA R, TAN BK, PUNN A, et al. Visfatin induces human endothelial VEGF and MMP-2/9 production via MAPK and PI3K/Akt signalling pathways: novel insights into visfatin-induced angiogenesis. Cardiovasc Res, 2008, 78(2): 356-365.
    [132] CHE G, CHEN J, LIU L, et al. Transfection of nm23-H1 increased expression of beta-Catenin, E-Cadherin and TIMP-1 and decreased the expression of MMP-2, CD44 v 6 and VEGF and inhibited the metastatic potential of human non-small cell lung cancer cell line L9981. Neoplasma, 2006, 53(6): 530-537.
    [133] WANG M, ZHANG W, CRISOSTOMO P, et al. STAT3 mediates bone marrow mesenchymal stem cell VEGF production. J Mol Cell Cardiol, 2007 Jun, 42(6): 1009-1015.
    [134] YU P, PASSAM FH, YU DM, et al. beta2-Glycoprotein I inhibits vascular endothelialgrowth factor and basic fibroblast growth factor induced angiogenesis through its amino terminal domain. J Thromb Haemost, 2008 Apr 30. [Epub ahead of print]
    [135] HARA T, MIYAZAKI H, LEE A, et al. Androgen receptor and invasion in prostate cancer. Cancer Res, 2008 Feb 15, 68(4): 1128-1135.
    [136] FANG JW, HUANG SL, LIU HS, et al. Role of FGF-2 /FGFR signaling pathway in cancer and its signification in breast cancer. Chin Sci Bulletin, 2003, 48(15): 1539-1547.
    [137] SCHRAMEK H. MAP kinases: from intracellular signals to physiology and disease. News Physiol Sci, 2002, 17(14):62-67.
    [138] KOYAMA T, MIKAMI T, KOYAMA T, et al. Apoptosis induced by chemo- therapeutic agents involves c-Jun N-terminal kinase activation in sarcoma cell lines. J Orthop Res, 2006 Jun, 24(6): 1153-1162.
    [139] KYRIAKS JM, AVRUCH J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev, 2001, 81: 807-869.
    [140] ADAMS JL, BADGER AM, KUMAR S, et al. p38MAPkinase: molecular target for the inhibition of proinflammatory cytokines. Prog Med Chem, 2001, 38: 1-60.
    [141] EL TOUNY LH, BANERJEE PP. Akt GSK-3 pathway as a target in genistein- induced inhibition of TRAMP prostate cancer progression toward a poorly differentiated phenotype. Carcinogenesis, 2007, 28(8): 1710-1707.
    [142] DEEB D, GAO X, DULCHAVSKY SA, et al. CDDO-Me inhibits proliferation, induces apoptosis, down-regulates Akt, mTOR, NF-kappaB and NF-kappaB-regulated antiapoptotic and proangiogenic proteins in TRAMP prostate cancer cells. J Exp Ther Oncol, 2008, 7(1): 31-39.
    [143] LU J, RHO O, WILKER E, et al. Activation of epidermal akt by diverse mouse skin tumor promoters. Mol Cancer Res, 2007 Dec, 5(12): 1342-1352.
    [144] ASSELIN E, MILLS GB, TSANG BK. XIAP regulates Akt activity and caspase-3 -dependent cleavage during cisplatin-induced apoptosis in human ovarian epithelial cancer cells. Cancer Res, 2001 Mar 1, 61(5): 1862-1868.
    [145] PARK S, KIM D, KANEKO S, et al. Molecular cloning and characterization of the human AKT1 promoter uncovers its up-regulation by the Src/Stat3 pathway. J Biol Chem, 2005 Nov 25, 280(47): 38932-38941.
    [146] AN WL, BJORKDAHL C, LIU R, et al. Mechanism of zinc-induced phosphorylationof p70 S6 kinase and glycogen synthase kinase 3beta in SH-SY5Y neuroblastoma cells. J Neurochem, 2005 Mar, 92(5): 1104-1115.
    [147] CASPI M, ZILBERBERG A, ELDAR-FINKELMAN H, et al. Nuclear GSK-3beta inhibits the canonical Wnt signalling pathway in a beta-catenin phosphorylation- independent manner. Oncogene, 2008 Jun 5, 27(25): 3546-3555.
    [148] CLIFFORD RL, DEACON K, KNOX AJ. Novel Regulation of Vascular Endothelial Growth Factor-A (VEGF-A) by Transforming Growth Factor {beta} 1: requirement for Smads, {beta}-CATENIN, AND GSK3{beta}. J Biol Chem, 2008 Dec 19, 283(51): 35337-35353.
    [149] LIU Y, BORCHERT GL, SURAZYNSKI A, et al. Proline oxidase, a p53-induced gene, targets COX-2/PGE2 signaling to induce apoptosis and inhibit tumor growth in colorectal cancers. Oncogene, 2008 Dec 4, 27(53): 6729-6737.
    [150] LAMARRE M, DESROSIERS RR. Up-regulation of protein L-isoaspartyl methyltransferase expression by lithium is mediated by glycogen synthase kinase-3 inactivation and beta-catenin stabilization. Neuropharmacology, 2008 Oct, 55(5): 669-676.
    [151] HOLMES T, OBRIEN TA, KNIGHT R, et al. Glycogen synthase kinase-3beta inhibition preserves hematopoietic stem cell activity and inhibits leukemic cell growth. Stem Cells, 2008 May, 26(5): 1288-12897.
    [152] HATAGUCHI K, OHSUGI M, ABE Y, et al. Ajuba negatively regulates the Wnt signaling pathway by promoting GSK-3beta-mediated phosphorylation of beta-catenin. Oncogene, 2008 Jan 10, 27(3): 274-284.
    [153] PATEL S, WOODGETT J. Glycogen synthase kinase-3 and cancer: good cop, bad cop? Cancer Cell, 2008 Nov 4, 14(5): 351-353.
    [154] KAWADA M, SENO H, UENOYAMA Y, et al. Signal transducers and activators of transcription 3 activation is involved in nuclear accumulation of beta-catenin in colorectal cancer. Cancer Res, 2006 Mar 15, 66(6): 2913-2917.
    [155] YAN S, ZHOU C, ZHANG W, et al. Beta-Catenin/TCF pathway upregulates STAT3 expression in human esophageal squamous cell carcinoma. Cancer Lett, 2008 Nov 18, 271(1): 85-97.
    [156] HAO J, LI TG, QI X, et al. WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Dev Biol, 2006 Feb 1, 290(1): 81-91.
    [157] ZHANG M, LEE CH, LUO DD, et al. Polarity of response to transforming growth factor-beta1 in proximal tubular epithelial cells is regulated by beta-catenin. J Biol Chem, 2007 Sep 28, 282(39): 28639-28647.
    [158] ROSS KR, COREY DA, DUNN JM, et al. SMAD3 expression is regulated by mitogen-activated protein kinase kinase-1 in epithelial and smooth muscle cells. Cell Signal, 2007 May, 19(5): 923-931.
    [159] LI H, XU D, LI J, et al. Transforming growth factor beta suppresses human telomerase reverse transcriptase (hTERT) by Smad3 interactions with c-Myc and the hTERT gene.J Biol Chem, 2006 Sep 1, 281(35): 25588-25600.
    [160] LI H, LIU JP. Mechanisms of action of TGF-beta in cancer: evidence for Smad3 as a repressor of the hTERT gene. Ann N Y Acad Sci, 2007 Oct, 1114: 56-68.
    [161] HU B, TACK DC, LIU T, et al. Role of Smad3 in the regulation of rat telomerase reverse transcriptase by TGFbeta. Oncogene, 2006 Feb 16, 25(7): 1030-1041.
NGLC 2004-2010.National Geological Library of China All Rights Reserved.
Add:29 Xueyuan Rd,Haidian District,Beijing,PRC. Mail Add: 8324 mailbox 100083
For exchange or info please contact us via email.