用户名: 密码: 验证码:
RhoC基因对肝癌细胞生物学行为影响及机制的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
信号蛋白分子RhoC在肿瘤细胞中的异常表达近年来引起了人们的广泛重视,但现有的研究多集中在RhoC调控肌动蛋白细胞骨架影响肿瘤转移方面,还有许多关键性的问题如RhoC在细胞增殖、凋亡、分化和耐药性等方面的作用知之不多。本课题以人肝癌细胞Bel7402为对象,研究RhoC基因对肝癌细胞生物学行为的影响及其相关的分子机制,为靶向RhoC的肝癌基因治疗奠定理论和实验基础。
     免疫细胞化学法检测RhoC基因在肝癌细胞和正常肝细胞中的表达;RNAi基因沉默肝癌细胞RhoC,RT-PCR和Western blot检测基因沉默效果;MTT法、平板克隆和软琼脂克隆形成实验以及AgNOs染色检测细胞增殖;划痕实验和细胞铺展实验检测细胞迁移侵袭能力;流式细胞术检测细胞周期、细胞凋亡、细胞膜通透性和线粒体膜电位变化;RT-PCR检测细胞周期蛋白、凋亡相关蛋白、MMPs表达;明胶酶谱分析检测MMPs活性;测量IC50值评价肝癌细胞化疗药物敏感性;As2O3诱导实验检测肝癌细胞分化水平;基因转染实验观测RhoC高表达所致的正常肝细胞恶性转化倾向。
     实验结果显示:RhoC在肝癌细胞中表达高于正常肝细胞,基因沉默RhoC可以显著抑制肝癌细胞增殖和转移,促进肝癌细胞大量凋亡,增加肝癌细胞对化疗药物环磷酰胺和长春新碱的敏感性,促进肿瘤细胞分化和诱导分化;RhoC转化正常肝细胞可使之出现恶性变倾向。进一步机理研究揭示:RhoC基因沉默对肝癌细胞增殖的作用可能与其调控Cyclin D1、CDK4表达上调及P16和P21表达下调有关;对肝癌细胞凋亡的作用可能与其调控Bcl-2/Bax基因、增加细胞膜通透性和降低线粒体膜电位有关;对迁移和侵袭的作用与其调控MMP-2和MMP-9基因表达有关。
     本研究表明,RhoC在肝癌细胞增殖、分化、凋亡、迁移及对化疗药物敏感性等方面起一定的作用,这种作用可能与其调控相关的分子有关。
Rho protein is an important member of Ras superfamily, which plays a diverse role in cell biology, including cell growth, differentiation and metastasis. RhoC is an important member of Rho GTPases. Recent researches showed the overexpression of RhoC in a great number of malignant tumors, and its activation could lead to enhanced tumor metastasis through specific signal transduction pathway. The high level expression of RhoC is closely correlated to tumor metastasis. Many studies suggested RhoC as a promising index in assessing tumor malignancy, with a possibility of acting as a therapeutic target for cancer gene therapy. However, research proofs on the role of RhoC in tumors are not strong enough to support RhoC as a therapeutic target, because there are still a lot of evidences should be provided and a lot of important mechanisms should be elucidated. This research systemically studied the effects of RhoC gene silencing in hepatocellular carcinoma (HCC) develpoment and cell differentiation. Also the role that RhoC plays in biological features and drug sensitivity in HCC cells were investigated. The importance of this research was to investigate the possibility of RhoC as a therapeutic target in HCC gene therapy.
     Immunohistochemistry was applied to detect RhoC expression in HCC cells. Eukaryotic expression plasmid pU6mRFP was employed as RNA interference vector; specific DNA sequence was synthesized to construct pU6mRFP siRNA-RhoC and pU6mRFP siRNA-scramble as negative control. Human HCC cell line Bel7402 was transfected to detect RhoC inhibiting efficiency. Lipotransfect was conducted to transfect Bel7402 cells, and its efficiency was assessed through laser confocal microscope. Semi-quantitative RT-PCR was used to detect mRNA level, Western blot was applied to detect RhoC protein expression. G418 was used to select stable transfection clone. MTT, cell nuclear AgNORs stain, plate clone formation and soft agar clone formation test were conducted to determine cell proliferation and growth feature. FACS was applied to determine cell cycle, cell apoptosis, cell membrane and mitochondrial membrane potential. As2O3 induction test, alkaline phosphate stain and biochemical analysis were employed to determine cell differenciation level. Cell migration capacity was determined through injury healing and cell spreading test. Zymography was used to detect MMPs activity. Semi-quantitative RT-PCR was performed to determine the expression level of cell cycle proteins, apoptosis associating proteins, MMPs and VEGF. IC50 values were measured to determine the sensitivities of anticancer drugs to HCC cells. Normal hepatocellular cell line HL7702 was transfected with RhoC to test RhoC transformation capacity. The results showed as followings:
     1. RNA interfering vector targetting RhoC was successfully constructed, with inhibiting effeciency in transfected Bel7402 cells as high as 85%.
     2. The effects of RhoC gene silencing in the proliferation of HCC and its mechanism. (1)The cultured cell proliferation capacity was compared between RNAi group (Bel7402 cell transfected with pU6mRFP siRNA-RhoC), negative control group (Bel7402 cells transfected with pU6mRFP siRNA-scramble) and parental Bel7402 cells. From the fourth day on, the proliferation speed in RNAi group was notably lower than in parental Bel7402 and negative control groups (0.41±0.10 vs. 0.73±0.11 and 0.71±0.07,p<0.05). These phenomena lasted to the end of the 7 day experiment. There was not remarkable difference between the two control groups. (2)Cell cycle analysis showed that G0-G1 cell percentage in RNAi group was significantly higher than in parental Bel7402 group (73.14±5.93% vs.57.05±5.97% and 52.99±4.80%,p<0.05). G2-S cell percentage was notably lower in RNAi group than in parental Bel7402 cells (24.86±3.66% vs.38.81±3.77% and 47.00±4.80%,P<0.01).There was not significant difference between two control groups.(3)Plate clone formation test showed that there was rare clones in RNAi group under naked eye observation, and more clones were formed in control groups. Statistical analysis showed that both clone number and clone formation efficiency in RNAi group were notably lower than in control groups (20.33±5.42% vs. 70.58±10.10% and 69.83±14.77%,p<0.01), suggesting RhoC gene silencing could notably inhibit the potential of cell growth. (4)AgNORs stain test showed that average cell stain particle in RNAi group was significantly lower than in parental Bel7402 and negative control(1.23±0.35 vs. 3.47±0.93 and 3.17±0.78,p<0.01), demonstrating the reduced cell proliferation capacity. (5)Soft agar clone formation test showed that clone formation number and percentage in RNAi group were significantly higher than in the control groups(0.133±0.115% vs. 2.867±0.987% and 2.533±1.007%,p<0.01), while the differences between two control groups were not remarkable (p>0.05), showing that RhoC silencing changed the non-anchor growth capacity of tumor cells. (6)Compared with Bel7402 and negative control groups, the following growth associated gene expressions were significantly decreased: CyclinD1(0.45±0.21 vs. 1.25±0.24 and 1.12±0.15,p<0.05)and CDK4(0.55±0.08 vs. 1.18±0.32 and 1.10±0.29,p<0.05). The following genes were notably increased: P16 ( 1.07±0.23 vs. 0.36±0.12 and 0.35±0.13,p<0.01)and P21(0.42±0.12 vs. 0.17±0.06 and 0.19±0.08,p<0.05). No remarkable changes were found in the following genes: CyclinA, CyclinG1, P19 and P27.
     3.The effects of RhoC gene silencing on Bel7402 cell apoptosis and its mechanism.(1)The results of cell apoptosis detected through FACS showed that the apoptosis ratio in RNAi group was significantly higher than in parental Bel7402 and negative control groups (21.00±2.23% vs. 6.47±1.64% and 4.63±0.47%,p<0.01). (2) Typical DNA fragment“ladder”for cell apoptosis could be found in RNAi group in gel electrophoresis analysis, suggesting there was great amount of cell apoptosis in RNAi group. There was not such DNA fragment“ladder”be found in parental Bel7402 and negative control groups, suggesting that there was not great amount cell apoptosis in control groups. (3)Wright stain test showed that there was lot of apoptosis cells in RNAi group, showing RhoC silencing could induce tumor cell death. (4)The membrane permeability in RNAi group was remarkably higher than in parental 7402 and negative control ones(30.70±5.63% vs.13.73±1.30% and 14.97±1.74%,p<0.01). (5)In RNAi group, the mitochondrial membrane potential was notably lower than in control ones ( 35.30±2.26% vs.65.60±5.54% and 65.37±5.61% , p<0.01 ) . (6)Compared with Bel7402 and negative control groups, Bcl-2 gene expression in RNAi group was significantly lower(0.28±0.15 vs. 0.96±0.21 and 1.03±0.24,p<0.05),and Bax gene expression was notably higher(1.09±0.21 vs. 0.26±0.10 and 0.25±0.07,p<0.01).
     4.Effects of RhoC gene silencing on Bel7402 cell differenciation and induced-differenciation. (1) Alkaline phosphates (ALP) stain showed that the stain color in RNAi group was more than in control ones. ALP activity in RNAi group was significantly higher than in parental Bel7402 and negative control groups (65.33±11.93u/L vs. 35.67±6.03u/L and 37.33±6.11u/L,p<0.05) .γ-GT activity was notably higher than in control groups ( 12.67±3.06u/L vs. 23.33±5.03u/L and 24.33±5.13u/L,p<0.05).(2)Under As2O3 induction, ALP activity in RNAi group was significantly higher than in parental 7402 and negative controls(123.67±24.58u/L vs. 56.67±11.93u/L and 54.67±10.26u/L,p<0.05).γ-GT activity was notably lower than in control ones(6.33±1.53u/L vs. 11.67±2.52u/L and 10.67±2.08u/L,p<0.05).
     5.Influence of RhoC gene silencing on Bel7402 invasion and metastasis. (1)Twenty four hours after scratch, the relative cell migration in RNAi group was significantly lower than in parental Bel7402 and negative control ones(32%±6% vs. 64%±13% and 67%±11%,p<0.01). (2)After 2 hours of cell spreading, the spreading ratio in RNAi group was notably lower than in parental 7402 and negative control ones(32.67±12.01% vs. 65.67±11.93% and 63.33±13.87%,p<0.05). (3)Zymography analysis showed that gelatinase activities could be found in parental Bel7402 and negative control ones, but not in RNAi group, suggesting the less production of MMPs with the silencing of RhoC gene. (4)MMP2 expression in RNAi group was significantly lower than in parental 7402 and negative control ones(0.26±0.10 vs. 1.01±0.23 and 1.03±0.27,p<0.01). MMP9 expression could not be detected in RNAi group, but with considerable level expression in control ones.
     6.Influence of RhoC on Bel7402 cell drug sensitivity. The value of IC50 for cyclophosphamide in RNAi group was significantly lower than in parental 7402 and negative control ones ( 838.54±69.22μg/ml vs. 1178.72±116.73μg/ml and 1168.36±168.68μg/ml ,p<0.01).Significant decrement of IC50 for vincristine could be found in RNAi group than in control ones(9.57±2.42μg/ml vs. 47.51±12.62μg/ml and 56.96±7.56μg/ml,p<0.01).
     7.The transformation effects of RhoC on normal liver cell HL7702. (1) The RhoC expression level in PcDNA3-RhoCgroup(HL7702 cells transfected with PcDNA3-RhoC) was significantly higher than in HL7702 and PcDNA3(HL7702 cells transfected with PcDNA3) groups(1.11±0.23 vs. 0.35±0.07 and 0.34±0.09,p<0.01), and Western blot detection proofed this difference. (2)From the third day on of cell culture, cell growth in PcDNA3-RhoC group was remarkably higher than in HL7702 and PcDNA3 groups(0.83±0.10 vs. 0.54±0.11 and 0.58±0.55,p<0.05). (3)The clone formation efficiency in PcDNA3-RhoC group was significantly higher than in HL7702 and PcDNA3 group(64.27±8.57% vs. 42.33±6.70% and 40.93±5.78%,p<0.01).(4) AgNORs stain test showed that the average stain particles in PcDNA3-RhoC group was notably higher than in HL7702 and PcDNA3 groups(2.77±0.40 vs. 1.07±0.23 and 1.13±0.25, P<0.01). (5)Soft agar clone formation in PcDNA3-RhoC group could be detected, but could not in the HL7702 and PcDNA3 groups. (6)The ALP stain was more intensive in PcDNA3-RhoC group than in HL7702 and PcDNA3 groups.
     In summary, the overexpression of RhoC in hepatocellular carcinoma cells can control tumor cell proliferation and metastasis through regulating the gene expressions involving cell growth and migration. RhoC gene silencing can inhibit proliferation and metastasis capacity of tumor cells, as well as enhancing the sensitivity of chemical therapy drugs such as cyclophosphamide and vincristine. This research lays a foundation to selecting RhoC as target molecule in tumor gene therapy both theoretically and experimentally.
引文
1. Jaffe, A.B. and A. Hall, Rho GTPases: biochemistry and biology. Annu Rev Cell Dev Biol, 2005. 21: p. 247-69.
    2. Aznar, S. and J.C. Lacal, Rho signals to cell growth and apoptosis. Cancer Lett, 2001. 165(1): p. 1-10.
    3. Fromm, C., et al., The small GTP-binding protein Rho links G protein-coupled receptors and Galpha12 to the serum response element and to cellular transformation. Proc Natl Acad Sci U S A, 1997. 94(19): p. 10098-103.
    4. Etienne-Manneville, S. and A. Hall, Rho GTPases in cell biology. Nature, 2002. 420(6916): p. 629-35.
    5. Vega, F.M. and A.J. Ridley, Rho GTPases in cancer cell biology. FEBS Lett, 2008. 582(14): p. 2093-101.
    6. Shi, J. and L. Wei, Rho kinase in the regulation of cell death and survival. Arch Immunol Ther Exp (Warsz), 2007. 55(2): p. 61-75.
    7. Fritz, G. and B. Kaina, Rho GTPases: promising cellular targets for novel anticancer drugs. Curr Cancer Drug Targets, 2006. 6(1): p. 1-14.
    8. Riento, K. and A.J. Ridley, Rocks: multifunctional kinases in cell behaviour. Nat Rev Mol Cell Biol, 2003. 4(6): p. 446-56.
    9. Loirand, G., P. Guerin, and P. Pacaud, Rho kinases in cardiovascular physiology and pathophysiology. Circ Res, 2006. 98(3): p. 322-34.
    10. Amano, M., et al., The COOH terminus of Rho-kinase negatively regulates rho-kinase activity. J Biol Chem, 1999. 274(45): p. 32418-24.
    11. Amano, M., Y. Fukata, and K. Kaibuchi, Regulation and functions of Rho-associated kinase. Exp Cell Res, 2000. 261(1): p. 44-51.
    12. Matsui, T., et al., Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. Embo J, 1996. 15(9): p. 2208-16.
    13. Hanks, S.K., A.M. Quinn, and T. Hunter, The protein kinase family: conserved features and deduced phylogeny of the catalytic domains. Science, 1988. 241(4861): p. 42-52.
    14. Nakagawa, O., et al., ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett, 1996. 392(2): p. 189-93.
    15. Hiroki, J., et al., Inflammatory stimuli upregulate Rho-kinase in human coronary vascular smooth muscle cells. J Mol Cell Cardiol, 2004. 37(2): p. 537-46.
    16. Leung, T., et al., A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to peripheral membranes. J Biol Chem, 1995. 270(49): p. 29051-4.
    17. Breitenlechner, C., et al., Protein kinase A in complex with Rho-kinase inhibitors Y-27632, Fasudil, and H-1152P: structural basis of selectivity. Structure, 2003. 11(12): p. 1595-607.
    18. Ishizaki, T., et al., Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol Pharmacol, 2000. 57(5): p. 976-83.
    19. Uehata, M., et al., Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature, 1997. 389(6654): p. 990-4.
    20. Amano, M., et al., Phosphorylation and activation of myosin by Rho-associated kinase (Rho-kinase). J Biol Chem, 1996. 271(34): p. 20246-9.
    21. Mills, J.C., et al., Apoptotic membrane blebbing is regulated by myosin light chain phosphorylation. J Cell Biol, 1998. 140(3): p. 627-36.
    22. Coleman, M.L., et al., Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat Cell Biol, 2001. 3(4): p. 339-45.
    23. Sebbagh, M., et al., Caspase-3-mediated cleavage of ROCK I induces MLC phosphorylation and apoptotic membrane blebbing. Nat Cell Biol, 2001. 3(4): p. 346-52.
    24. Di Cristofano, A. and P.P. Pandolfi, The multiple roles of PTEN in tumor suppression. Cell, 2000. 100(4): p. 387-90.
    25. Li, Z., et al., Regulation of PTEN by Rho small GTPases. Nat Cell Biol, 2005. 7(4): p. 399-404.
    26. Leung, T., et al., The p160 RhoA-binding kinase ROK alpha is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol Cell Biol, 1996. 16(10): p. 5313-27.
    27. Yoneda, A., H.A. Multhaupt, and J.R. Couchman, The Rho kinases I and II regulate different aspects of myosin II activity. J Cell Biol, 2005. 170(3): p. 443-53.
    28. Papakonstanti, E.A., A.J. Ridley, and B. Vanhaesebroeck, The p110delta isoform of PI
    3-kinase negatively controls RhoA and PTEN. Embo J, 2007. 26(13): p. 3050-61.
    29. Doran, J.D., et al., New insights into the structure-function relationships of Rho-associated kinase: a thermodynamic and hydrodynamic study of the dimer-to-monomer transition and its kinetic implications. Biochem J, 2004. 384(Pt 2): p. 255-62.
    30. Shirao, S., et al., Sphingosylphosphorylcholine is a novel messenger for Rho-kinase-mediated Ca2+ sensitization in the bovine cerebral artery: unimportant role for protein kinase C. Circ Res, 2002. 91(2): p. 112-9.
    31. Riento, K., et al., RhoE binds to ROCK I and inhibits downstream signaling. Mol Cell Biol, 2003. 23(12): p. 4219-29.
    32. Manapov, F., P. Muller, and J. Rychly, Translocation of p21(Cip1/WAF1) from the nucleus to the cytoplasm correlates with pancreatic myofibroblast to fibroblast cell conversion. Gut, 2005. 54(6): p. 814-22.
    33. Benitah, S.A., et al., Rho GTPases in human cancer: an unresolved link to upstream and downstream transcriptional regulation. Biochim Biophys Acta, 2004. 1705(2): p. 121-32.
    34. Perona, R., et al., Activation of the nuclear factor-kappaB by Rho, CDC42, and Rac-1 proteins. Genes Dev, 1997. 11(4): p. 463-75.
    35. Montaner, S., et al., Activation of serum response factor by RhoA is mediated by the nuclear factor-kappaB and C/EBP transcription factors. J Biol Chem, 1999. 274(13): p. 8506-15.
    36. Sotiropoulos, A., et al., Signal-regulated activation of serum response factor is mediated by changes in actin dynamics. Cell, 1999. 98(2): p. 159-69.
    37. Cen, B., et al., Megakaryoblastic leukemia 1, a potent transcriptional coactivator for serum response factor (SRF), is required for serum induction of SRF target genes. Mol Cell Biol, 2003. 23(18): p. 6597-608.
    38. Robertson, L.M., et al., Regulation of c-fos expression in transgenic mice requires multiple interdependent transcription control elements. Neuron, 1995. 14(2): p. 241-52.
    39. Sadowski, H.B., et al., A common nuclear signal transduction pathway activated by growthfactor and cytokine receptors. Science, 1993. 261(5129): p. 1739-44.
    40. Simon, A.R., et al., Regulation of STAT3 by direct binding to the Rac1 GTPase. Science, 2000. 290(5489): p. 144-7.
    41. Friedland, J.C., et al., alpha6beta4 integrin activates Rac-dependent p21-activated kinase 1 to drive NF-kappaB-dependent resistance to apoptosis in 3D mammary acini. J Cell Sci, 2007. 120(Pt 20): p. 3700-12.
    42. Zhu, S., et al., RhoA prevents apoptosis during zebrafish embryogenesis through activation of Mek/Erk pathway. Oncogene, 2008. 27(11): p. 1580-9.
    43. Matos, P. and P. Jordan, Increased Rac1b expression sustains colorectal tumor cell survival. Mol Cancer Res, 2008. 6(7): p. 1178-84.
    44. Yamaki, N., M. Negishi, and H. Katoh, RhoG regulates anoikis through a phosphatidylinositol 3-kinase-dependent mechanism. Exp Cell Res, 2007. 313(13): p. 2821-32.
    45. Li, K., et al., RhoE enhances multidrug resistance of gastric cancer cells by suppressing Bax. Biochem Biophys Res Commun, 2009. 379(2): p. 212-6.
    46. Moore, M., et al., Rho kinase inhibition initiates apoptosis in human airway epithelial cells. Am J Respir Cell Mol Biol, 2004. 30(3): p. 379-87.
    47. Rattan, R., et al., Rho/ROCK pathway as a target of tumor therapy. J Neurosci Res, 2006. 83(2): p. 243-55.
    48. Li, X., et al., Inhibition of protein geranylgeranylation and RhoA/RhoA kinase pathway induces apoptosis in human endothelial cells. J Biol Chem, 2002. 277(18): p. 15309-16.
    49. Ikeda, H., et al., Involvement of Rho/Rho kinase pathway in regulation of apoptosis in rat hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol, 2003. 285(5): p. G880-6.
    50. Fukushima, M., et al., Fasudil hydrochloride hydrate, a Rho-kinase (ROCK) inhibitor, suppresses collagen production and enhances collagenase activity in hepatic stellate cells. Liver Int, 2005. 25(4): p. 829-38.
    51. Han, S., N. Sidell, and J. Roman, Fibronectin stimulates human lung carcinoma cell proliferation by suppressing p21 gene expression via signals involving Erk and Rho kinase. Cancer Lett, 2005. 219(1): p. 71-81.
    52. De Sarno, P., et al., Anti-apoptotic effects of muscarinic receptor activation are mediated by Rho kinase. Brain Res, 2005. 1041(1): p. 112-5.
    53. Olson, M.F., A. Ashworth, and A. Hall, An essential role for Rho, Rac, and Cdc42 GTPases in cell cycle progression through G1. Science, 1995. 269(5228): p. 1270-2.
    54. Klein, E.A., et al., NFkappaB-independent signaling to the cyclin D1 gene by Rac. Cell Cycle, 2007. 6(9): p. 1115-21.
    55. Villalonga, P. and A.J. Ridley, Rho GTPases and cell cycle control. Growth Factors, 2006. 24(3): p. 159-64.
    56. Clark, E.A., et al., Genomic analysis of metastasis reveals an essential role for RhoC. Nature, 2000. 406(6795): p. 532-5.
    57. van Golen, K.L., et al., RhoC GTPase, a novel transforming oncogene for human mammary epithelial cells that partially recapitulates the inflammatory breast cancer phenotype. Cancer Res, 2000. 60(20): p. 5832-8.
    58. Nur, E.K.M.S., et al., The CDC42-specific inhibitor derived from ACK-1 blocks v-Ha-Ras-induced transformation. Oncogene, 1999. 18(54): p. 7787-93.
    59. Mira, J.P., et al., Endogenous, hyperactive Rac3 controls proliferation of breast cancer cells by a p21-activated kinase-dependent pathway. Proc Natl Acad Sci U S A, 2000. 97(1): p. 185-9.
    60. Osada, S., et al., A domain containing the Cdc42/Rac interactive binding (CRIB) region of p65PAK inhibits transcriptional activation and cell transformation mediated by the Ras-Rac pathway. FEBS Lett, 1997. 404(2-3): p. 227-33.
    61. Westwick, J.K., et al., Rac regulation of transformation, gene expression, and actin organization by multiple, PAK-independent pathways. Mol Cell Biol, 1997. 17(3): p. 1324-35.
    62. Tapon, N., et al., A new rac target POSH is an SH3-containing scaffold protein involved in the JNK and NF-kappaB signalling pathways. Embo J, 1998. 17(5): p. 1395-404.
    63. Teramoto, H., et al., The small GTP-binding protein rho activates c-Jun N-terminal kinases/stress-activated protein kinases in human kidney 293T cells. Evidence for a Pak-independent signaling pathway. J Biol Chem, 1996. 271(42): p. 25731-4.
    64. Muller, S.M., E. Okan, and P. Jones, Regulation of urokinase receptor transcription by Ras- and Rho-family GTPases. Biochem Biophys Res Commun, 2000. 270(3): p. 892-8.
    65. Huang, S., et al., Urokinase plasminogen activator/urokinase-specific surface receptor expression and matrix invasion by breast cancer cells requires constitutive p38alpha mitogen-activated protein kinase activity. J Biol Chem, 2000. 275(16): p. 12266-72.
    66. Dang, J., et al., A region between -141 and -61 bp containing a proximal AP-1 is essential for constitutive expression of urokinase-type plasminogen activator receptor. Eur J Biochem, 1999. 264(1): p. 92-9.
    67. Itoh, K., et al., An essential part for Rho-associated kinase in the transcellular invasion of tumor cells. Nat Med, 1999. 5(2): p. 221-5.
    68. Aznar, S., et al., Simultaneous tyrosine and serine phosphorylation of STAT3 transcription factor is involved in Rho A GTPase oncogenic transformation. Mol Biol Cell, 2001. 12(10): p. 3282-94.
    69. Frankel, P., et al., Ral and Rho-dependent activation of phospholipase D in v-Raf-transformed cells. Biochem Biophys Res Commun, 1999. 255(2): p. 502-7.
    70. Song, Y., B.Q. Hoang, and D.D. Chang, ROCK-II-induced membrane blebbing and chromatin condensation require actin cytoskeleton. Exp Cell Res, 2002. 278(1): p. 45-52.
    71. Orlando, K.A. and R.N. Pittman, Rho kinase regulates phagocytosis, surface expression of GlcNAc, and Golgi fragmentation of apoptotic PC12 cells. Exp Cell Res, 2006. 312(17): p. 3298-311.
    72. Shiratsuchi, A., T. Mori, and Y. Nakanishi, Independence of plasma membrane blebbing from other biochemical and biological characteristics of apoptotic cells. J Biochem, 2002. 132(3): p. 381-6.
    73. Orlando, K.A., N.L. Stone, and R.N. Pittman, Rho kinase regulates fragmentation and phagocytosis of apoptotic cells. Exp Cell Res, 2006. 312(1): p. 5-15.
    74. Erwig, L.P., et al., Differential regulation of phagosome maturation in macrophages and dendritic cells mediated by Rho GTPases and ezrin-radixin-moesin (ERM) proteins. Proc Natl Acad Sci U S A, 2006. 103(34): p. 12825-30.
    75. Tosello-Trampont, A.C., K. Nakada-Tsukui, and K.S. Ravichandran, Engulfment of apoptotic cells is negatively regulated by Rho-mediated signaling. J Biol Chem, 2003. 278(50): p.49911-9.
    76. Fritz, G., I. Just, and B. Kaina, Rho GTPases are over-expressed in human tumors. Int J Cancer, 1999. 81(5): p. 682-7.
    77. Abraham, M.T., et al., Motility-related proteins as markers for head and neck squamous cell cancer. Laryngoscope, 2001. 111(7): p. 1285-9.
    78. Zhao, X., et al., Overexpression of RhoA induces preneoplastic transformation of primary mammary epithelial cells. Cancer Res, 2009. 69(2): p. 483-91.
    79. Horiuchi, A., et al., Overexpression of RhoA enhances peritoneal dissemination: RhoA suppression with Lovastatin may be useful for ovarian cancer. Cancer Sci, 2008. 99(12): p. 2532-9.
    80. Kamai, T., et al., Significant association of Rho/ROCK pathway with invasion and metastasis of bladder cancer. Clin Cancer Res, 2003. 9(7): p. 2632-41.
    81. Jiang, K., et al., Akt mediates Ras downregulation of RhoB, a suppressor of transformation, invasion, and metastasis. Mol Cell Biol, 2004. 24(12): p. 5565-76.
    82. Sato, N., et al., RhoB is frequently downregulated in non-small-cell lung cancer and resides in the 2p24 homozygous deletion region of a lung cancer cell line. Int J Cancer, 2007. 120(3): p. 543-51.
    83. Couderc, B., et al., In vivo restoration of RhoB expression leads to ovarian tumor regression. Cancer Gene Ther, 2008. 15(7): p. 456-64.
    84. Kleer, C.G., et al., RhoC GTPase expression as a potential marker of lymph node metastasis in squamous cell carcinomas of the head and neck. Clin Cancer Res, 2006. 12(15): p. 4485-90.
    85. Wang, W., et al., Expression and significance of RhoC gene in hepatocellular carcinoma. World J Gastroenterol, 2003. 9(9): p. 1950-3.
    86. Lin, M., et al., Regulation of pancreatic cancer cell migration and invasion by RhoC GTPase and caveolin-1. Mol Cancer, 2005. 4(1): p. 21.
    87. Kondo, T., et al., Expression of RHOC is associated with metastasis of gastric carcinomas. Pathobiology, 2004. 71(1): p. 19-25.
    88. van Golen, K.L., et al., A novel putative low-affinity insulin-like growth factor-binding protein, LIBC (lost in inflammatory breast cancer), and RhoC GTPase correlate with the inflammatory breast cancer phenotype. Clin Cancer Res, 1999. 5(9): p. 2511-9.
    89. Kleer, C.G., et al., Characterization of RhoC expression in benign and malignant breast disease: a potential new marker for small breast carcinomas with metastatic ability. Am J Pathol, 2002. 160(2): p. 579-84.
    90. van Golen, K.L., et al., RhoC GTPase overexpression modulates induction of angiogenic factors in breast cells. Neoplasia, 2000. 2(5): p. 418-25.
    91. Price, L.S. and J.G. Collard, Regulation of the cytoskeleton by Rho-family GTPases: implications for tumour cell invasion. Semin Cancer Biol, 2001. 11(2): p. 167-73.
    92. Watts, K.L., et al., RhoA signaling modulates cyclin D1 expression in human lung fibroblasts; implications for idiopathic pulmonary fibrosis. Respir Res, 2006. 7: p. 88.
    93. Lee, S. and D.M. Helfman, Cytoplasmic p21Cip1 is involved in Ras-induced inhibition of the ROCK/LIMK/cofilin pathway. J Biol Chem, 2004. 279(3): p. 1885-91.
    94. Narumiya, S., M. Tanji, and T. Ishizaki, Rho signaling, ROCK and mDia1, in transformation, metastasis and invasion. Cancer Metastasis Rev, 2009.
    95. Kleer, C.G., et al., RhoC-GTPase is a novel tissue biomarker associated with biologically aggressive carcinomas of the breast. Breast Cancer Res Treat, 2005. 93(2): p. 101-10.
    96. Espina, C., et al., A critical role for Rac1 in tumor progression of human colorectal adenocarcinoma cells. Am J Pathol, 2008. 172(1): p. 156-66.
    97. Rajnicek, A.M., L.E. Foubister, and C.D. McCaig, Prioritising guidance cues: directional migration induced by substratum contours and electrical gradients is controlled by a rho/cdc42 switch. Dev Biol, 2007. 312(1): p. 448-60.
    98. Ridley, A.J., Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol, 2006. 16(10): p. 522-9.
    99. Yamazaki, D., S. Kurisu, and T. Takenawa, Regulation of cancer cell motility through actin reorganization. Cancer Sci, 2005. 96(7): p. 379-86.
    100. Hall, A., Rho GTPases and the control of cell behaviour. Biochem Soc Trans, 2005. 33(Pt 5): p. 891-5.
    101. Malliri, A. and J.G. Collard, Role of Rho-family proteins in cell adhesion and cancer. Curr Opin Cell Biol, 2003. 15(5): p. 583-9.
    102. Yu, W., et al., Involvement of RhoA, ROCK I and myosin II in inverted orientation of epithelial polarity. EMBO Rep, 2008. 9(9): p. 923-9.
    103. Abouzahr-Rifai, S., et al., Resistance of tumor cells to cytolytic T lymphocytes involves Rho-GTPases and focal adhesion kinase activation. J Biol Chem, 2008. 283(46): p. 31665-72.
    104. Merajver, S.D. and S.Z. Usmani, Multifaceted role of Rho proteins in angiogenesis. J Mammary Gland Biol Neoplasia, 2005. 10(4): p. 291-8.
    105. Takenawa, T. and H. Miki, WASP and WAVE family proteins: key molecules for rapid rearrangement of cortical actin filaments and cell movement. J Cell Sci, 2001. 114(Pt 10): p. 1801-9.
    106. Miki, H., et al., IRSp53 is an essential intermediate between Rac and WAVE in the regulation of membrane ruffling. Nature, 2000. 408(6813): p. 732-5.
    107. Govind, S., et al., Cdc42Hs facilitates cytoskeletal reorganization and neurite outgrowth by localizing the 58-kD insulin receptor substrate to filamentous actin. J Cell Biol, 2001. 152(3): p. 579-94.
    108. Tolias, K.F., et al., Type Ialpha phosphatidylinositol-4-phosphate 5-kinase mediates Rac-dependent actin assembly. Curr Biol, 2000. 10(3): p. 153-6.
    109. Zhang, H.S., A.A. Postigo, and D.C. Dean, Active transcriptional repression by the Rb-E2F complex mediates G1 arrest triggered by p16INK4a, TGFbeta, and contact inhibition. Cell, 1999. 97(1): p. 53-61.
    110. Ogawa, T., et al., Rho-associated kinase inhibitor reduces tumor recurrence after liver transplantation in a rat hepatoma model. Am J Transplant, 2007. 7(2): p. 347-55.
    111. Fromigue, O., Z. Hamidouche, and P.J. Marie, Blockade of the RhoA-JNK-c-Jun-MMP2 cascade by atorvastatin reduces osteosarcoma cell invasion. J Biol Chem, 2008. 283(45): p. 30549-56.
    112. Ikoma, T., et al., A definitive role of RhoC in metastasis of orthotopic lung cancer in mice. Clin Cancer Res, 2004. 10(3): p. 1192-200.
    113. Turner, N.A., et al., Simvastatin inhibits MMP-9 secretion from human saphenous vein smooth muscle cells by inhibiting the RhoA/ROCK pathway and reducing MMP-9 mRNAlevels. Faseb J, 2005. 19(7): p. 804-6.
    114. Pille, J.Y., et al., Anti-RhoA and anti-RhoC siRNAs inhibit the proliferation and invasiveness of MDA-MB-231 breast cancer cells in vitro and in vivo. Mol Ther, 2005. 11(2): p. 267-74.
    115. Croft, D.R., et al., Conditional ROCK activation in vivo induces tumor cell dissemination and angiogenesis. Cancer Res, 2004. 64(24): p. 8994-9001.
    116. Stoletov, K., et al., High-resolution imaging of the dynamic tumor cell vascular interface in transparent zebrafish. Proc Natl Acad Sci U S A, 2007. 104(44): p. 17406-11.
    117. Mammoto, A., T. Mammoto, and D.E. Ingber, Rho signaling and mechanical control of vascular development. Curr Opin Hematol, 2008. 15(3): p. 228-34.
    118. Bayless, K.J., R. Salazar, and G.E. Davis, RGD-dependent vacuolation and lumen formation observed during endothelial cell morphogenesis in three-dimensional fibrin matrices involves the alpha(v)beta(3) and alpha(5)beta(1) integrins. Am J Pathol, 2000. 156(5): p. 1673-83.
    119. Garrett, W.S., et al., Developmental control of endocytosis in dendritic cells by Cdc42. Cell, 2000. 102(3): p. 325-34.
    120. Tzima, E., Role of small GTPases in endothelial cytoskeletal dynamics and the shear stress response. Circ Res, 2006. 98(2): p. 176-85.
    121. Wheeler, A.P. and A.J. Ridley, Why three Rho proteins? RhoA, RhoB, RhoC, and cell motility. Exp Cell Res, 2004. 301(1): p. 43-9.
    122. Fritz, G., B. Kaina, and K. Aktories, The ras-related small GTP-binding protein RhoB is immediate-early inducible by DNA damaging treatments. J Biol Chem, 1995. 270(42): p. 25172-7.
    123. Steeg, P.S., Cancer: micromanagement of metastasis. Nature, 2007. 449(7163): p. 671-3.
    124. Dittert, D.D., et al., Prognostic significance of immunohistochemical RhoA expression on survival in pancreatic ductal adenocarcinoma: a high-throughput analysis. Hum Pathol, 2008. 39(7): p. 1002-10.
    125. Faried, A., et al., Clinical and prognostic significance of RhoA and RhoC gene expression in esophageal squamous cell carcinoma. Ann Surg Oncol, 2007. 14(12): p. 3593-601.
    126. Ullmannova-Benson, V., et al., DLC1 tumor suppressor gene inhibits migration and invasion of multiple myeloma cells through RhoA GTPase pathway. Leukemia, 2009. 23(2): p. 383-90.
    127. Robert, V.J., N.L. Vastenhouw, and R.H. Plasterk, RNA interference, transposon silencing, and cosuppression in the Caenorhabditis elegans germ line: similarities and differences. Cold Spring Harb Symp Quant Biol, 2004. 69: p. 397-402.
    128. Hannon, G.J., RNA interference. Nature, 2002. 418(6894): p. 244-51.
    129. Brummelkamp, T.R., R. Bernards, and R. Agami, A system for stable expression of short interfering RNAs in mammalian cells. Science, 2002. 296(5567): p. 550-3.
    130. Yoon, S.Y., et al., Construction of a vector generating both siRNA and a fluorescent reporter: a siRNA study in cultured neurons. Mol Cells, 2004. 18(1): p. 127-30.
    131. Quinones-Hinojosa, A., et al., Techniques to assess the proliferative potential of brain tumors. J Neurooncol, 2005. 74(1): p. 19-30.
    132. Malumbres, M. and M. Barbacid, Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer, 2009. 9(3): p. 153-66.
    133. Alao, J.P., The regulation of cyclin D1 degradation: roles in cancer development and thepotential for therapeutic invention. Mol Cancer, 2007. 6: p. 24.
    134. Larrea, M.D., et al., Phosphorylation of p27Kip1 regulates assembly and activation of cyclin D1-Cdk4. Mol Cell Biol, 2008. 28(20): p. 6462-72.
    135. Matsuda, Y. and T. Ichida, p16 and p27 are functionally correlated during the progress of hepatocarcinogenesis. Med Mol Morphol, 2006. 39(4): p. 169-75.
    136. Liggett, W.H., Jr. and D. Sidransky, Role of the p16 tumor suppressor gene in cancer. J Clin Oncol, 1998. 16(3): p. 1197-206.
    137. Jung, P. and H. Hermeking, The c-MYC-AP4-p21 cascade. Cell Cycle, 2009. 8(7): p. 982-9.
    138. Ioachim, E., Expression patterns of cyclins D1, E and cyclin-dependent kinase inhibitors p21waf1/cip1, p27kip1 in colorectal carcinoma: correlation with other cell cycle regulators (pRb, p53 and Ki-67 and PCNA) and clinicopathological features. Int J Clin Pract, 2008. 62(11): p. 1736-43.
    139. McKee, A.E. and C.J. Thiele, Targeting caspase 8 to reduce the formation of metastases in neuroblastoma. Expert Opin Ther Targets, 2006. 10(5): p. 703-8.
    140. Kuwana, T. and D.D. Newmeyer, Bcl-2-family proteins and the role of mitochondria in apoptosis. Curr Opin Cell Biol, 2003. 15(6): p. 691-9.
    141. Adams, J.M. and S. Cory, The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene, 2007. 26(9): p. 1324-37.
    142. Spira, A.I. and M.A. Carducci, Differentiation therapy. Curr Opin Pharmacol, 2003. 3(4): p. 338-43.
    143. Clark, I.M., et al., The regulation of matrix metalloproteinases and their inhibitors. Int J Biochem Cell Biol, 2008. 40(6-7): p. 1362-78.
    144. Marty, M. and X. Pivot, The potential of anti-vascular endothelial growth factor therapy in metastatic breast cancer: clinical experience with anti-angiogenic agents, focusing on bevacizumab. Eur J Cancer, 2008. 44(7): p. 912-20.
    145. Scotto, K.W. and D.A. Egan, Transcriptional regulation of MDR genes. Cytotechnology, 1998. 27(1-3): p. 257-69.

© 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