用户名: 密码: 验证码:
DADS靶向诱导人胃癌细胞分化与周期阻滞及机理研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
胃癌是我国最常见的恶性肿瘤,其发病率和死亡率仍居首位,每年约有40余万人患胃癌,而死于胃癌者多达30余万,大大高于欧美发达国家。目前,传统的手术治疗、放射治疗、化疗治疗等疗效较差、易复发、副反应严重,5年生存率仅为30%左右。因此,提高5年生存率,探索治疗胃癌的新方法,寻找新一代更有效、低毒和价廉的抗肿瘤药物,一直是亟待解决的课题。现在,逆转人胃癌细胞的研究已成为生命科学研究前沿领域。二烯丙基二硫(diallyl disulfide, DADS)是大蒜中的一种脂溶性的有效成分,对乳腺癌、肝癌、肺癌、膀胱癌、结肠癌、前列腺癌和白血病等多种肿瘤均有明显的抑制作用,是一种很有开发潜力的抗肿瘤候选药物。
     【DADS诱导人胃癌细胞增殖抑制、分化与周期阻滞的研究】
     本研究采用MTT法、电镜观察、Con A凝集实验、碱性磷酸酶(ALP)比活性测定、骨架蛋白细胞化学、划痕标记荧光染料示踪技术(SLDT)、免疫组化等方法,观察DADS对体外培养的人胃癌MGC803细胞的生长抑制与诱导分化效应。MTT比色实验结果显示,DADS浓度从20、25、30到35mg·L~(-1)处理人胃癌MGC803细胞,其吸光值比对照组低,而且随浓度增加逐渐降低(P<0.05);相反,随着浓度增加,DADS对癌细胞增殖的抑制率从11.3%、35.2%、50.7%到60.6%逐渐增强(P<0.05),表明DADS对人胃癌细胞具有明显的生长抑制作用,且呈显著的剂量-效应依赖关系。电镜观察发现30mg·L~(-1)DADS引起瘤细胞异型性降低,核质比下降,细胞表面微绒毛数目减少,胞浆内细胞器丰富,胞核及部分细胞器呈退行性变,并可见细胞间连接结构;DADS处理后瘤细胞对Con A的凝集率从79.10%降为26.98%(P<0.05);ALP比活性由0.120 U/g下降至0.040 U/g(P<0.05),下降率为66.12%;骨架蛋白细胞化学研究表明,细胞骨架蛋白合成增加与重组,划痕标记染料示踪技术(SLDT)显示细胞间缝隙连接通讯功能得以恢复;免疫组化发现,DADS处理后的MGC803细胞p21WAF1表达增强,p21ras、c-Myc、突变型p53表达减弱而pRb的表达没有改变。上述结果提示,DADS可诱导人胃癌MGC803细胞增殖抑制与分化。DADS对人胃癌BGC823细胞也具有一样的生长抑制效应,且呈显著的剂量-效应依赖关系(P<0.05)。流式细胞分析表明,DADS可将MGC803和BGC823细胞阻滞在G_2/M期。DADS作用12 h时,G_2/M期细胞分别增加到35.7%和30.1%;24 h时,分别增加到51.7和58.1%;36h时,分别达58.7%和50.2%(P<0.05)。
     Western Blot结果显示,20、25、30、35mg·L~(-1)DADS处理,呈浓度依赖性抑制人胃癌细胞中ERK的活化(P<0.05);并且DADS +MEK抑制剂U0126能完全阻断ERK活化(P<0.05);时间效应上,30mg·L~(-1)DADS处理后15-30min抑制作用最强,2h左右恢复正常(P<0.05)。提示DADS抑制ERK通路可能是诱导人胃癌细胞增殖抑制与分化的重要分子机制。结论:DADS具有诱导人胃癌细胞分化及抑制增殖的作用。DADS诱导人胃癌细胞增殖抑制和分化与细胞周期G_2/M期阻滞有关。ERK活性抑制是DADS诱导人胃癌细胞分化与抑制增殖的重要分子机制之一。
     【DADS对人胃癌MGC803和BGC823细胞G_2/M期检查点激酶通路的影响】
     肿瘤是多基因变化导致细胞周期紊乱的一类疾病,周期调控机制失活,特别是G1/S和G_2/M期检测点(checkpoint)的失活在细胞癌变过程中起着至关重要的作用。采用药物激活细胞周期调控机制,从而诱导肿瘤细胞凋亡和分化已成为肿瘤治疗的一个可行的新途径。前期工作证实,DADS对人胃癌MGC803细胞周期的作用可能与调节Cdc25C、cyclinB1蛋白的表达有关。本实验发现,DADS呈时间依赖性抑制人胃癌BGC823细胞周期Cdc25C磷酸酶的表达,cyclinB1表达在DADS处理24 h后开始下降,其改变与MGC803细胞中的一致。Cdc25C作用于cyclinB1/ CDK1复合物,而其磷酸酶活性受细胞周期检查点Chk1和/或Chk2激酶调节。本研究显示,30mg·L~(-1)DADS分别刺激1h和2h后,MGC803和BGC823细胞磷酸化Chk1表达明显增加,并呈时间依赖性(P<0.05),提示DADS对两种细胞中的磷酸化Chk1的作用相仿,而人胃癌MGC803和BGC823细胞处理组与未处理组磷酸化Chk2表达无明显差异(P>0.05);总Chk1和Chk2蛋白表达在两种细胞处理前后均无改变(P>0.05)。RT-PCR检测发现,Chkl和Chk2 mRNA水平在处理组与未处理组之间无明显差异(P>0.05),表明DADS只对蛋白磷酸化有影响,而对其mRNA水平无影响。以上结果提示,DADS是通过激活Chk1而不是Chk2调节Cdc25C、cyclinB1的改变。Chk1是ATR的下游分子,我们进一步检测ATR的活性,结果显示,磷酸化ATR在MGC803和BGC823细胞中未处理组无表达,DADS处理15min后被激活,并呈时间依赖性(P<0.05),而ATR总蛋白表达均无改变,这进一步说明DADS可能是通过ATR/Chk1/Cdc25C/cyclin B1引起细胞周期G_2/M阻滞。免疫共沉淀分析结果表明,MGC803和BGC823细胞中DADS能促进Chk1与Cdc25C结合,而对Chk2与Cdc25C结合无影响,证明DADS作用的周期信号通路是Chk1/Cdc25C,而不是Chk2/Cdc25C。结论:DADS可能是通过靶向检查点激酶Chk1引起G_2/M期阻滞。DADS阻滞人胃癌细胞于G_2/M期与调节ATR/ Chk1/ Cdc25C/ cyclin B1信号通路有关,可能成为治疗胃癌的新靶点。
     【Chk1和Chk2基因沉默对DADS G_2/M期阻滞作用及检查点激酶通路的影响】
     RNAi(RNA interference)是转录后基因沉默的一种调控机制,是机体固有的对抗外源侵袭基因及维持自身基因稳定性的一种自我保护机制。本研究采用RNAi技术分别在MGC803和BGC823细胞中将Chk1、Chk2基因沉默,进一步确定DADS的作用靶点。将Chk1、Chk2 RNAi分别转染MGC803和BGC823细胞后,Western blot和实时荧光定量PCR验证结果显示,MGC803和BGC823细胞Chk1 mRNA表达分别被抑制76.0%和84.7%,蛋白表达分别下降85.0%和90.2%;chk2 mRNA表达分别降低84.1%和69.0%,蛋白表达分别下调83.4%和93.5%。
     MTT结果发现,Chk1基因沉默后,对MGC803和BGC823细胞增殖有明显抑制作用,其抑制率分别为38%和48.1%(P<0.05),而加以30mg·L~(-1)DADS后,其抑制率分别增加至56.3%和68.4%,与Chk1基因未沉默而用30mg·L~(-1)DADS处理组无差异(P>0.05)。相反,Chk2基因沉默后对细胞增殖无明显抑制作用(P>0.05)。流式细胞术分析Chk1和Chk2基因沉默后对BGC823细胞周期的影响,发现Chk1基因沉默后G_2期明显比未处理组减少12.5%(P<0.05),Chk2基因沉默后G_2期下降8.1%(P>0.05),Chk1和Chk2基因同时沉默G_2期下降13.6%(P<0.05),与Chk1基因单独沉默的影响接近(P>0.05)。Chk1基因沉默后加入DADS后,在24h、48h,G_2期分别为10.4%、17.1%,与未处理组18.6%、20.4%比较,对细胞周期G_2期影响不大(P>0.05),但较处理组58.1%、36.2%G_2期,分别下降47.7%、19.1%(P<0.05),这表明DADS的作用靶点可能是Chk1。Chk1和Chk2下游分子的改变显示,Chk1基因沉默后Cdc25C和cyclinB1表达较未处理组明显增加,而加入DADS后,Cdc25C和cyclinB1表达与未处理组相比无差异;但Chk2基因沉默后对Cdc25C和cyclinB1表达无影响,对DADS的作用无影响,证实DADS的主要作用靶点是Chk1而不是Chk2。结论:Chk1基因沉默可明显抑制人胃癌细胞增殖。DADS阻滞G_2/M期是通过Chk1/Cdc25C/cyclinB1信号通路起作用,其主要作用靶点是Chk1。Chk2不参与DADS对胃癌G_2/M期的阻滞作用。
Gastric cancer is the most common type of malignancy in China, and its morbidity and mortality rank first now. There are 400,000 people or so with gastric cancer, and 300,000 people succumb to it, which is much higher than that in the west developed countries. At present, therapeutic effect of orthodox surgical therapy, radiation therapy and chemotherapy is comparatively weak, easy to palindromia, furthermore , their side effect is serious and the 5-year survival rate of gastric cancer remains only about 30%. Therefore, it is an urgently solved topic to raise the 5-year survival rate, explore new method to treat gastric cancer, and look for new effective anticancer drugs with low toxicity and inexpensive price. Now human gastric cancer reversion research has become leading edge field in life science research. DADS inhibits the in vitro growth of colon, lung, breast and gastric cancer cell lines, and leukemia cell lines and it becomes a very potential candicate anticancer drug.
     【DADS inhibited growth of human gastric cancer MGC803 cells and induced them differentiation】
     MTT assay, electron microscope,Con A-mediated cell agglutination, ALP specific activity detection and optics microscope, cytochemical staining, scrape-loading and dye transfer technology were employed to confirm the DADS-induced cell growth inhibition and differentiation in MGC803 cell line. Exposure of human gastric cancer MGC803 cells to DADS from 25 to 35mg·L~(-1) for 96 h had less absorbance value than control,which exhibited a dose-dependent model (P<0.05),whereas DADS induced-growth inhibition rate enhanced from 11.3%,35.2%,50.7% to 60.6% as assessed by the MTT test(P<0.05).As exposed to electron microscope, MGC803 cells took on malignance declining as cellular heteromorphism diminished, nucleocytoplasmic proportion was reliable to reasonableness, the number of microvilli on cell surface decreased, cellular apparatus were abundant in plasm and intercellular conjunctional structure appeared after it being exposed to 30mg·L~(-1) DADS; Con A-mediated cell agglutination ratio was 26.98% versus controls 79.10% (p<0.05) ; cell ALP specific activity decreased by 66.12% from 0.120 U/g to 0.040 U/g (p<0.05). Cellular skeletin synthesized and reconstructured and recruitment of intercellular communication of gap junction were also observed with cytochemical technology; Gastric cancer cells showed negative of dye transfer,while positive transfer of LY dye from the load cells at the scraped-line to neighboring cells in gastric cancer cells induced by DADS. Immunihistichemical stain indicated that expression of p21WAF1 enhanced and that mutant p53,p21ras,c-myc downregulation expression and ther was no change in pRb. All these showed above suggested that human gastric cancer cells were induced differentiation into normal cells by DADS. Flow cytometry analysis revealed that treatment MGC803 cell with increasing quantities of DADS increased in the percentage of cells in the G_2/M phase. The proportion of cells in the G_2/M phase after treatment with 30mg·L~(-1) DADS for 36 h was comparable (58.7%), and more than four times that occurring in untreated cells (13.8%). Exposure of human gastric cancer BGC823 cells to DADS from 5 to 20 mg·L~(-1) for 96 h had less absorbance value than control, which exhibited a dose-dependent model (P<0.05), whereas DADS induced-growth inhibition rate enhanced from 34.2、39.2、55.7、69.6% as assessed by the MTT test (P<0.05).Which suggest that DADS inhibited growth of human gastric cancer BGC823 cells. After BGC823 cells were treated with 15mg·L~(-1) DADS, flow cytometry analysis displayed that cell percentage in S phase of the DADS treated MGC803 cells Was decreased,whereas cell percentage in G_2 phase Was significantly increased(P<0.05),and little change in Gl phase was found. Which is the same as that of MGC803 cells.
     Much attention has been focused on the ERK pathway as a possible target for newly designed anti-neoplastic drugs. Therefore, it is essential to establish the role that the ERK pathway plays in the process that leads to differentiation within a specific tissue or cell type. In this study, ERK activity is observed in the DADS-induced differentiation effect in human gastric cancer cell line MGC803. Western blot analysis revealed that although DADS did not influence the quantity of ERK1/2 protein expressed, it did decrease its phosphorylation in a concentration-dependent manner, compared with the controls (P<0.05). At 30 mg·L~(-1), DADS inhibited the activation of ERK1/2 in 15-30 min (P<0.05). These results suggested that the DADS-induced differentiation of MGC803 cells involved an alteration of the ERK1/2 signaling pathway .
     Conclusion: Differentiation and growth inhibition was induced by DADS. Cell Cycle G_2/M Arrest was involved in growth inhibition and differentiation induced by DADS in the Human Gastric Cancer Cells. ERK is involved in the differentiation and growth inhibition induced by diallyl disulfide in the human gastric cancer cells.
     【Checkpoint Kinase Signaling is Involved in Cell Cycle G_2/M Arrest Induced by Diallyl Disulfide in the Human Gastric Cancer Cell Line MGC803 and BGC823】
     Anticancer insights derived from cell cycle research has given birth to the idea of cell cycle G_2 checkpoint abrogation as a cancer cell specific therapy, based on the discovery that many cancer cells have a defective G1 checkpoint resulting in a dependence on the G_2 checkpoint during cell replication. The most promising target to date seems to be Chk1 and there will be a growing number of selective inhibitors of Chk1 available in the future with a variety of activities that promise to have potential G_2 checkpoint abrogation qualities. Cell cycle has recently become more appealing as a new target of anti-carcinogenic agent.It is a new mechanism that the role of Chk was reduced by anti-carcinogenic drug in tumor cells. Furthermore, G_2/M cell cycle arrest is mainly related with the Chk2-Cdc25C-Cdc2/cyclin B1 pathway in cancer cells.
     It was found that Cdc25C and CyclinB1 expression are critical events in G_2/M arrest of MGC803 cells by DADS. DADS increased the expression of cyclinB1 by 20% after 12 h. DADS inhibited the expression of the cell cycle-associated phosphatase Cdc25C by 80% after 48 h. Western blot analysis showed that DADS treatment of BGC823 cells decreased the level of Cdc25C in a time-dependent model (P<0.05). DADS increased the expression of cyclinB1 after 12h and then its expression decreased after 24h (P<0.05), which tendency is similar to MGC803 cells by DADS. Cdc25C was thought to be the major effector of the G_2/M DNA damage checkpoint response. Cdc25C which triggers cyclinB1/CDK1 complex is controlled by Chk1 or/and Chk2. So Chk1 and Chk2 activity was observed by DADS on human gastric cancer cells. Western blot analysis showed that phosphorylation of Chk1 was decreased following treatment of MGC803 and BGC823 cells with DADS after 1hrs in a time-dependent model. At the same time,the total Chk1 abundance did not change.Expression of Phospho-Chk2 was weak in these two untreated cell lines,furthermore, its expression wasn’t changed by DADS (P>0.05). Expression of Chk1 and Chk2 had no change after treated with DADS. The level of Chkl and Chk2 mRNA had no significant difference between the DADS treatment and contro1 by RT-PCR (P>0.05). Which showed that the level of Chkl and Chk2 mRNA had no relationship with cell cycle arrest.
     ATR is capable of specifically phosphorylating Chkl. Western blot analysis showed that Phospho-ATR was inactivated by DADS from 15 or 30 to 90 min in MGC803cells or BGC823 cells respectively(P<0.05). Expression of ATR had no change after treated with DADS. Coimmunoprecipitated Chk1 or Chk2 was detected by anti-Chk1 or anti-Chk2 immunoprecipitation (IP) followed by anti-Cdc25C immunoblotting (IB), DADS enhanced binding activity of Chk1 kinase with Cdc25C in MGC803 and BGC823 cells,but didn’t influence binding activity of Chk2 kinase with Cdc25C. Which verified that DADS induced G_2/M arrest by Chk1- Cdc25C on MGC803cells and BGC823 cells.
     Conclusion: Checkpoint kinase Chk1 may be the target to arrest cell cycle by DADS. ATR/ Chk1/Cdc25C/ cyclin B1 was regulated by DADS to arrest Human Gastric Cancer Cells at G_2/M.
     【Checkpoint Kinase Signaling is regulated in Cell Cycle G_2/M Arrest Induced by Chk1 and Chk2 RNAi】
     To investigate the effect of Chk1 or Chk2 gene silence on the cell viability of human gastric cancer cell line MGC803 and BGC823, and explore the role of Chk1 and Chk2 in cell cycle arrest. The siRNA targeting at Chk1 or Chk2 gene was transfected into MGC803 and BGC823 cells. The mRNA and protein expression of Chk1 was detected by RT-PCR and Western-blot respectively. The Chk1 or Chk2 expression at mRNA and protein levels in Chk1 or Chk2 siRNA-transfected group was reduced markedly respectively as compared with that in negative control and empty vector-transfected group (P<0.05).
     Cell viability was determined via MTT assay. Treated with DADS, cells-transfected viability by Chk1 SiRNA was decreased by 18.3% or 20.3% in MGC803 and BGC823 cells respectively (P<0.05). Inhibition of the Chk1 expression in Chk1 siRNA transfected group in BGC823 cells significantly abrogated G_2/M arrest induced by DADS, and the proportion of the cells in G_2/M phase was more than two double lower than that in other control groups (P<0.05).While inhibition of the Chk2 expression in Chk2 siRNA transfected group had little effect on G_2/M arrest.
     Chk1 gene silence increased expression of Cdc25C and cyclinB1,and inhibition of expression of Cdc25C and cyclinB1 by DADS was blocked (P<0.05). On the contrary,Chk2 gene silence can not do so.
     Conclusion: Chk1 gene silence can inhibited and growth of Human Gastric Cancer Cells, and take synergistic effect with DADS. DADS arrestted Human Gastric Cancer Cells at G_2/M by Chk1/Cdc25C /CyclinB1, and its main target was Chk1. Chk2 didn’t participate G_2/M arrest by DADS in Human Gastric Cancer Cells.
引文
1. 孙秀娣,牧人,周有尚,等.中国胃癌死亡率 20 年变化情况分析及其发展趋势预测. 中华肿瘤学杂志. 2004; 26(1): 4-9
    2. Fleischauer AT, Arab L. Garlic and cancer: a critical review of the epidemiologic literature. J Nutr. 2001;131(3s):1032S-40S
    3. Ryzhenkov VE, Makarov VG. Biologically active substances in garlic (Allium sativum L.) and their application in nutrition for humans. Vopr Pitan. 2003;72(4):42-46
    4. Milner JA. A historical perspective on garlic and cancer. J Nutr. 2001; 131(3s):1027S-1031S
    5. Nakagawa H, Tsuta K, Kiuchi K, Senzaki H, Tanaka K, Hioki K, Tsubura A.Growth inhibitory effects of diallyl disulfide on human breast cancer cell lines.Carcinogenesis. 2001; 22(6):891-7.
    6. Kalra N, Arora A, Shukla Y.Involvement of multiple signaling pathways in diallyl sulfide mediated apoptosis in mouse skin tumors.Asian Pac J Cancer Prev. 2006;7(4):556-62.
    7. De Martino A, Filomeni G, Aquilano K, Ciriolo MR, Rotilio G.Effects of water garlic extracts on cell cycle and viability of HepG2 hepatoma cells.J Nutr Biochem. 2006; 17(11):742-9.
    8. Wu XJ, Kassie F, Mersch-Sundermann V.The role of reactive oxygen species (ROS) production on diallyl disulfide (DADS) induced apoptosis and cell cycle arrest in human A549 lung carcinoma cells.Mutat Res. 2005; 579(1-2):115-24.
    9. Wargovich MJ, Woods C, Eng VW, Stephens LC, Gray K.Chemoprevention of N-nitrosomethylbenzylamine-induced esophageal cancer in rats by the naturally occurring thioether, diallyl sulfide.Cancer Res. 1988; 48(23):6872-5.
    10. Jakubikova J, Sedlak J. Garlic-derived organosulfides induce cytotoxicity, apoptosis, cell cycle arrest and oxidative stress in human colon carcinoma celllines.Neoplasma. 2006; 53(3):191-9.
    11. Yang JS, Kok LF, Lin YH, Kuo TC, Yang JL, Lin CC, Chen GW, Huang WW, Ho HC, Chung JG. Diallyl disulfide inhibits WEHI-3 leukemia cells in vivo.Anticancer Res. 2006; 26(1A):219-25.
    12. Xiao XL, Su Q, et al. Diallyl trisulfide induces apoptosis of human gastric cancer cell line MGC803 through caspase-3 pathway.Ai Zheng. 2006; 25(10):1247-51. Chinese.
    13. Robert V, Mouille B, Mayeur C, et al. Effects of the garlic compound diallyl disulfide on the metabolism, a dherence and cell cycle of HT-29colon carcinoma cells: evidence of sensitive and resistant sub-populations. Carcinogenesis. 2001; 22(8): 1155-61
    14. Knowles LM, Milner JA. Diallyl disulfide inhibits p34(cdc2) kinase activity through changes in complex formation and phosphorylation. Carcinogenesis. 2000; 21(6):1129- -34
    15. Kwon KB,Yoo SJ,Ryu DG,et al.Induction of apoptosis by diallyl disulfide through activation of caspase-3 in human leukemia HL-60 cells. Biochem Pharmacol, 2002,63(1):41-7
    16. Lea MA, Randolph VM, Patel M,et al.Increased acetylation of histone induced by diallyl disulfide and structurally related molecules. Int J Oncol, 1999, 15(2):347-52
    17. Nakagawa H, Tsuta K, Kiuchi K,et al.Growth inhibitory effects of diallyl disulfide on human breast cancer cell lines. Carcinogenesis. 2001;22(6): 891-7
    18. Lea MA, Randolph VM.Induction of histone acetylation in rat liver and organosulfur compounds including diallyl aisulfide. Anticancer Res, 2001,21(4A):2841-5
    19. Hong VS, Ham YA, Choi JH,et al.Effects of allyl sulfur compounds and garlic extract on the expression of Bcl-2,Bax,and p53 in non small cell lung cancer cell lines. Exp Mol Med. 2000;32(3):127-34
    20. 王代树, 梁云燕, 彭敬, 等. PKA-RII和PKC-α在人胃癌MGC803细胞c-myc和 c-H-ras 表达中的共调节作用. 实用生物学报, 1996,29(3):197-205
    21. Knowles LM, Milner JA. Depressed p34cdc2 kinase activity and G2/M phase arrest induced by diallyl disulfide in HCT-15 cells. Nutr Cancer. 1998;30(3):169-74
    22. Hiroyuki Niida and Makoto Nakanishi. DNA damage checkpoints in mammals Mutagenesis. 2006; 21(1): 3–9,
    23. Takumi K.checkpoint abrogators as anticancer drugs. Mol Cancer Ther. 2004;3:513-519
    24. Sedlacek HH. Mechanisms of action of flavopiridol.Crit Rev Oncol Hematol.2001; 38(2):139-170
    25. Schmidt M, et al. Protection against chemotherapy-induced cytotoxity by cyclin-dependent kinase inhibitors (CKI) in CKI-responsive cells compared with CKI-unresponsive cells. Oncogene, 2001; 20(43): 6164-6171
    26. Goodarzi AA, et al. The role of ATM and ATR in DNA damage-induced cell cycle control. Prog Cell Cycle Res, 2003; 5:393–411.
    27. Zhou BB, et al.. Drug discovery targeting Chk1 and Chk2 kinases. Prog Cell Cycle Res, 2003; 5:413–21.
    28. Sorensen CS, et al. Chk1 regulates the S phase checkpoint by coupling the physiological turnover and ionizing radiation-induced accelerated proteolysis of Cdc25A. Cancer Cell; 2003; 3:247-258.
    29. Zhao H et al. Piwnica-Worms. ATR-mediated checkpoint pathways regulate phosphorylation and activation of human Chk1. Mol. Cell. Biol. 2001; 21:4129-4139
    30. Guo, Z., A. et al. Requirement for Atr in phosphorylation of Chk1 and cell cycle regulation in response to DNA replication blocks and UV-damaged DNA in Xenopus egg extracts. 2000. Genes Dev. 14:2745-2756.
    31. Deep G, et al. Silymarin and silibinin cause G1 and G2-M cell cycle arrest viadistinct circuitries in human prostate cancer PC3 cells: a comparison of flavanone silibinin with flavanolignan mixture silymarin. Oncogene. 2006. 16; 25 (7):1053-69.
    32. 肖晓岚,苏琦,等.二烯丙基三硫诱导人胃癌细胞凋亡与钙稳态失衡相关. 中国药理学通报. 2005;21 (8): 962-966
    33. 张良运,苏琦,等.二烯丙基二硫对人胃癌 MGC803 细胞生长的影响.世界华人消化杂志, 2003;11(9): 1290-1293
    34. 谭晖, 苏琦,等.中国药理学通报.二烯丙基二硫诱导人白血病 HL-60 细胞凋亡及机制. 2004; 20(8):879-883
    35. 武明花, 苏琦, 等. 二烯丙基二硫对 HL-60 细胞的生长抑制和诱导分化作用.中华血液学杂志.2004; 25(5):300-302
    36. Chambard JC, Lefloch R, Pouyssegur J, Lenormand P. ERK implication in cell cycle regulation. Biochim Biophys Acta. 2006; 17; [Epub ahead of print]
    37. Beck SE, Jung BH,Del Rosario E, Gomez J, Carethers JM.BMP-induced growth suppression in colon cancer cells is mediated by p21(WAF1) stabilization and modulated by RAS/ERK.Cell Signal. 2007 Jan 24; [Epub ahead of print]
    38. Park YY, Nam HJ, Lee JH. Hepatocyte growth factor at S phase induces G2 delay through sustained ERK activation. Biochem Biophys Res Commun.2007 Mar 1; [Epub ahead of print]
    39. Yuan JP,Su Q,et al. Diallyl disulfide-induced G2/M arrest of human gastric cancer MGC803 cells involves activation of p38 MAP kinase pathways.World J Gastroenterol. 2004; 15; 10(18):2731-4.
    40. Xiao Z, Chen Z,Gunasekera AH,et al. Chk1 Mediates S and G2 Arrests through Cdc25A Degradation in Response to DNA-damaging Agents. J. Biol. Chem. 2003; 24(278): 21767-21773
    41. 李晓光, 谢锦玉, 吕有勇.大蒜油对人胃癌 BGC-823 细胞的生长抑制和诱导分化的作用. 华人消化杂志.1998;6(1):10-12
    42. Chen, Z.M., Chen, Y.Q.and Zhang, C. The Effects of Enzyme-digested Solution from Mensamaria intercedens (Lampart) on Gastric Cancer Cell MGc80-3 in Culture. J. Xiamen University (Natural Science) .1998;37: 594-599.
    43. Paulson AF,Lampe PD,Meyer RA,et al.Enhancement of gap junction assembly.Mol Cell Biol. 1994,5(suppl.):199-203
    44. Dillard CY,Narayanan SK,Dennis WF.The role of dibutyrl cyclic adenosine monophosphate and cytoskeletal elements in annular gap junction distribution in human adrenal cortical tumor(SW-13) cell.Mol Cell Biol. 1994;5(suppl.):197-199
    45. Krutovskikh VA, Troyanovsky SM,Piccoli C,et al.Differential effect of subcellular localization of communication impairing gap junction protein connexin 43 on tumor cell growth in vivo.Oncogene. 2000;19(4):505-513
    46. Verdoodt B, Decordier I, Geleyns K, et al.Induction of polyploidy and apoptosis after exposure to high concentration of the spindle poison nocodazole. Mutagenesis. 1999; 14 (5):513-520
    47. Yang SM. DMSO induced differentiation of human gastric adenocarcinoma cell line MGC803. Shi Yan Sheng Wu Xue Bao. 1994; 27:281-287.
    48. Chen RC, Su JH, Yang SM, Li J, Wang TJ and Zhou H. Effect of isoverbascoside, a phenylpropanoid glycoside antioxidant, on proliferation and differentiation of human gastric cancer cell. Acta. Pharmacol. Sin. 2002; 23: 997-1001.
    49. Ara C, Massimi M and Devirgiliis CL. Retinoic acid modulates gap junctional intercellular communication in hepatocytes and hepatoma cells.Cell. Mol. Life Sci. 2002; 59:1758-1765.
    50. Cho JH, Cho SD, Hu H, Kim SH, Lee SK and Lee YS. The role of ERK1/2 and p38 MAP kinase in the preventive mechanisms of mushroom phellinus linteus against the inhibition of gap junctional communication by hydrogen peroxide. Carcinogenesis. 2002; 23:1163-1169.
    51. Simon AM and Goodenough DA. Diverse functions of vertrbrate gap junction. Trends Cell Biol. 1998; 8: 477-483.
    52. Carruba G, Cocciadiferro L, Bellavia V, Rizzo S, Tsatsanis C, Spandidos D, Muti P, Smith C, Mehta P and Castagnetta L. Intercellular communication and human hepatocellular carcinoma. Ann.N.Y. Acad. Sci. 2004; 1028:202-212.
    53. Mills AA. p53: link to the past, bridge to the future.Genes & Dev., Sep 2005; 19: 2091 - 2099.
    54. Noda H, Maehara Y, Irie K, Kakeji Y, Yonemura T, Sugimachi K. Growth pattern and expressions of cell cycle regulator proteins p53 and p21WAF1/CIP1 in early gastric carcinoma.Cancer. 2001 ;92:1828-1835
    55. Okuyama T, Maehara Y, Kabashima A, Takahashi I, Kakeji Y, Sugimachi K.Combined evaluation of expressions of p53 and p21 proteins as prognostic factors for patients with gastric carcinom.Oncology 2002; 63:353-361
    56. Amannllah A, Liebermann DA, Hoffman B.P53-independent apoptosis associated with c-Myc-mediated block in myeloid cell differentiation. Oncogene, 2000, 19:2967-2977
    57. L?hr K, M?ritz C, Contente A, et al. p21/CDKN1A Mediates Negative Regulation of Transcription by p53.J Biol Chem. 2003; 278: 32507 - 32516.
    58. Grinstein E, Shan Y, Karawajew L, et al,Cell Cycle-controlled Interaction of Nucleolin with the Retinoblastoma Protein and Cancerous Cell Transformation.J. Biol. Chem., Aug 2006; 281: 22223 - 22235.
    59. Obaya AJ, Kotenko I, Cole MD, et al.The Proto-oncogene c-myc Acts through the Cyclin-dependent Kinase (Cdk) Inhibitor p27Kip1 to Facilitate the Activation of Cdk4/6 and Early G1 Phase Progression.J. Biol. Chem., Aug 2002; 277: 31263 - 31269.
    60. Prober DA,Edger BA.Ras1 promotes cellular growth in the Drosophilawing.Cell,2000,100(4):435-446
    61. Lewis TS,Shapiro PS,Ahn NG.Signal transduction through MAP kinase cascades.Adv Cancer Res,1998;74(1):49-53
    62. Segar R,Krebs EG.The MAPK signaling cascade.FASEB J,1995;9(3):726-36
    63. Mansour SJ, Matten WT, Hermann AS, et al.Transformation of mammalian cells by constitutively active MAP kinase kinase.Science,1994;265(4):966-75
    64. Brunet A, Pages G, Pouysseger J.Constitutively active mutants of MAP kinase kinase (MEK1) induce growth factor-relaxation and oncogenenicity when expressed in fibroblasts. Oncogene, 1994; 9 (15) :3379-87
    65. Kortenjann M,Thomae O,Shaw PE.Inhibition of v-raf- dependent c-fos expression and transformation by a kinase-defective mutant of the mitogen-activated protein kinase ERK2.Mol Cell Biol,1994; 14 (25) :4815-26
    66. Pages G,Lenormand P,L’Allemain G,et al. Mitogen- activated protein kinase p42mapk and p44mapk are required for fibroblast proliferation.Proc Natl Acad Sci U S A.1993;90(41):8319-30
    67. Minshull J,Sun H,Tonks NK,et al.A MAP-kinase dependent spindle assembly checkpoint in Xenopus egg extracts.Cell 1994;79(3):475-86
    68. Kuno Y,Kondo K,Iwata H,et al. Tumor-specific activation of mitogen-activated protein kinase in human colorectal and gastric carcinoma tissues. Jpn Cancer Res 1998;89(9):903-9
    69. Husain SS,Szabo IL , Pai R,et al. MAPK(ERK2) kinase—a key target for NSAIDs-induced inhibition of gastric cancer cell proliferation and growth.Life Sci 2001;69 (25 -26):3045-54
    70. Matsuzaki T, Aisaki KI, Yamamura Y, et al. Induction of erythroid differentiation by inhibition of Ras/ERK pathway in a Friend murine leukemia cell line. Oncogene 2000; 19 (12):1500-11
    71. Olsson AK, Vadhammar K, Nanberg E. Activation and Protein Kinase C- dependentnuclear accumulation of ERK in differentiating human neuroblastoma cells. Exp Cell Res 2000; 256(3):454-63
    72. He H,Wang X,Gorospe M,et al.Phorbol ester-induced mononuclear cell differen- tiation is blocked by the mitogen activated protein kinase kinase(MEK) inhibitor PD98059.Cell Growth Differ 1999;10(5):307-11
    73. Lessor T, Yoo JY, Davis M, et al.Regulation of heregulin betal-induced differentiation in a human breast carcinoma cell line by the extracellular- regulated kinase (ERK) pathway. J Cell Biochem 1998;70(4):587-92
    74. Yen A, Roberson MS, Varvayanis S, et al.Retinoic acid induced mitogen-activated protein(MAP) /extracellular signal-regulated kinase(ERK) kinase-dependent MAP kinase activation needed to elicit HL-60 cell differentiation and growth arrest.Cancer Res 1998;58(14):3136-44
    75. Sanuma S,Yan Z,Ohno S,et al.Protein kinase C beta 1 and protein kinase C beta 2 activate p57 mitogen-activated protein kinase and block differentiation in colon carcinoma cells.Cell Growth Differ 1996;7(5):587-95
    76. 向殊霖, 苏琦, 等.DADS 对人胃癌细胞裸鼠移植瘤的抗增殖作用.癌症,2005;24(8):940-944
    77. Hartwell LH, Kastan MB. Cell cycle control and cancer. Science, 1994;266(5192):1821-8
    78. McDonald ER 3rd,eEl-Deiry WS. Cell cycle control as a basis for cancer drug development.Int J Oncol 2000; 16 (5):871-886
    79. Blasina A, Weyer IV, Laus MC, Luyten WH, Parker AE and McGowan CH A human homologue of the checkpoint kinase Cds1 directly inhibits Cdc25 phosphatase. Curr. Biol. 1999;9: 1–10.
    80. Uto,K., Inoue,D., Shimuta,K., Nakajo,N. and Sagata,N. Chk1, but not Chk2, inhibits Cdc25 phosphatases by a novel common mechanism.EMBO J.2004; 23: 3386–3396.
    81. Lopez-Girona,A., Kanoh,J. and Russell,P. (2001) Nuclear exclusion of Cdc25 is not required for the DNA damage checkpoint in fission yeast.Curr. Biol., 11, 50–54.
    82. Peng,C.Y., Graves,P.R., Thoma,R.S., Wu,Z., Shaw,A.S. and Piwnica-Worms,H. (1997) Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216.Science, 277, 1501–1505.
    83. Furnari,B., Blasina,A., Boddy,M.N., McGowan,C.H. and Russell,P.(1999) Cdc25 inhibited in vivo and in vitro by checkpoint kinases Cds1 and Chk1. Mol. Biol. Cell, 10, 833–845.
    84. Graves,P.R., Yu,L., Schwarz,J.K., Gales,J., Sausville,E.A.,O’Connor,P.M. and Piwnica-Worms,H. (2000) The Chk1 protein kinase and the Cdc25C regulatory pathways are targets of the anticancer agent UCN-01. J. Biol. Chem., 275, 5600–5605.
    85. Falck,J., Mailand,N., Syljuasen,R.G., Bartek,J. and Lukas,J. (2001) The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature, 410, 842–847.
    86. Kastan,M.B. (2001) Cell cycle. Checking two steps. Nature, 410,766–767.
    87. Arata,Y., Fujita,M., Ohtani,K., Kijima,S. and Kato,J.Y. (2000) Cdk2-dependent and -independent pathways in E2F-mediated S phase induction.J. Biol. Chem., 275, 6337–6345.
    88. Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev, 2001;15:2177–96.
    89. King RW, Jackson PK, Kirschner MW. Mitosis in transition [J]. Cell , 1994, 79 (4): 563-71
    90. Frey RS, Li J, Singletary KW. Effects of genistein on cell proliferation and cell cycle arrest in nonneoplastic human mammary epithelial cells: involvement of Cdc2, p21(waf/cip1), p27(kip1), and Cdc25C expression. Biochem Pharmacol 2001 Apr 15;61(8):979-89
    91. Viallard JF, Lacombe F, Dupouy M, Ferry H, Belloc F, Reiffers J. Different expression profiles of human cyclin B1 in normal PHA-stimulated T lymphocytes and leukemic T cells. Cytometry 2000 Feb 1;39(2):117-25
    92. Lin H, Liu XY, Subramanian B, Nakeff A, Valeriote F, Chen BD. Mitotic arrest induced by XK469, a novel antitumor agent, is correlated with the inhibition of cyclin B1 ubiquitination. Int J Cancer 2002 Jan 1;97(1):121-8
    93. Hershko A. Mechanisms and regulation of the degradation of cyclin B. Philos Trans R Soc Lond B Biol Sci 1999 Sep 29;354(1389):1571-5; discussion 1575-6
    94. Hershko A, Ganoth D, Sudakin V, Dahan A, Cohen LH, Luca FC, Ruderman JV, Eytan E. Components of a system that ligates cyclin to ubiquitin and their regulation by the protein kinase cdc2. J Biol Chem 1994 Feb 18;269(7):4940-6
    95. Walworth,N., Davey,S. and Beach,D. (1993) Fission yeast chk1 protein kinase links the rad checkpoint pathway to cdc2. Nature, 363, 368–371.
    96. Murakami,H. and Okayama,H. (1995) A kinase from fission yeast responsible for blocking mitosis in S phase. Nature, 374, 817–819.
    97. Sanchez,Y., Wong,C., Thoma,R.S., Richman,R., Wu,Z., Piwnica-Worms,H. and Elledge,S.J. (1997) Conservation of the Chk1 checkpoint pathway in mammals: linkage of DNA damage to Cdk regulation throughCdc25. Science, 277, 1497–1501.
    98. Kaneko YS, Watanabe N, Morisaki H, Akita H, Fujimoto A, Tominaga K, Terasawa M, Tachibana A, Ikeda K and Nakanishi M.(1999). Cell-cycle-dependent and ATM-independent expression of human Chk1 kinase. Oncogene, 18, 3673–3681.
    99. Matsuoka,S., Huang,M. and Elledge,S.J. (1998) Linkage of ATM to cell cycle regulation by the Chk2 protein kinase. Science, 282, 1893–1897.
    100.Blasina A, de Weyer IV, Laus MC, Luyten WH, Parker AE and McGowan CH. A human homologue of the checkpoint kinase Cds1 directly inhibits Cdc25 phosphatase. Curr. Biol. 1999; 9: 1-10.
    101.Tominaga K, Morisaki H, Kaneko Y, Fujimoto A, Tanaka T, Ohtsubo M, Hirai M, Okayama H, Ikeda K and Nakanishi M. Role of human Cds1 (Chk2) kinase in DNA damage checkpoint and its regulation by p53. J. Biol. Chem. 1999; 274: 31463– 31467.
    102.Takai H, Tominaga K, Motoyama N, Minamishima YA, Nagahama H, Tsukiyama T, Ikeda K, Nakayama K and Nakanishi M. Aberrant cell cycle checkpoint function and early embryonic deathin Chk1(-/-) mice. Genes Dev. 2000; 14: 1439–1447.
    103.Liu Q, Guntuku S, Cui XS, et al. Chk1 is an essential kinase thatis regulated by Atr and required for the G(2)/M DNA damage checkpoint.Genes Dev. 2000; 14: 1448– 1459.
    104.Zhao H and Piwnica-Worms H. ATR-mediated checkpoint pathwaysregulate phosphorylation and activation of human Chk1. Mol Cell Biol. 2001; 21: 4129– 4139.
    105.Lopez-Girona A, Tanaka K, Chen XB, Baber BA, McGowan CH and Russell P. Serine-345 is required for Rad3-dependent phosphorylation and function of checkpoint kinase Chk1 in fission yeast. Proc.Natl Acad. Sci. USA, 2001; 98: 11289–11294.
    106.Zou L, Cortez D and Elledge SJ. Regulation of ATR substrate selection by Rad17-dependent loading of Rad9 complexes onto chromatin.Genes Dev. 2002: 16: 198–208.
    107.Weiss RS, Matsuoka S, Elledge SJ and Leder P. Hus1 acts upstream of chk1 in a mammalian DNA damage response pathway.Curr. Biol., 2002;12, 73–77.
    108.Chen MS, Ryan CE and Piwnica-Worms H Chk1 kinase negatively regulates mitotic function of Cdc25A phosphatase through 14-3-3 binding. Mol. Cell Biol. 2003; 23, 7488–7497
    109.Hirao A, Kong YY, Matsuoka S, Wakeham A, Ruland J, Yoshida H, Liu D, Elledge SJ and Mak TW. DNA damage-induced activationof p53 by the checkpoint kinaseChk2. Science, 2000; 287, 1824–1827.
    110.Takai H, Naka K, Okada Y, et al. Chk2-deficient mice exhibit radioresistance and defective p53-mediated transcription. EMBO J. 2002; 21:5195–5205.
    111.Melchionna R, Chen XB, Blasina A and McGowan CH. Threonine 68 is required for radiation-induced phosphorylation andactivation of Cds1. Nat. Cell Biol. 2000; 2: 762–765.
    112.Bartek,J. and Lukas,J. Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell, 2003;3:421–429.
    113.Falck J, Mailand N, Syljuasen RG., Bartek J and Lukas J. The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature, 2001; 410: 842–847.
    114.Cimprich KA, Shin TB, Keith CT and Schreiber SL. cDNA cloning and gene mapping of a candidate human cell cycle checkpoint protein. Proc. Natl Acad. Sci. USA, 1996; 93:2850–2855.
    115.Brown EJ and Baltimore D. ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 2000; 14:397–402.
    116.Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev. 2001; 15:2177–2196.
    117.Takai H, Tominaga K, Motoyama N, Minamishima YA, Nagahama H, Tsukiyama T, Ikeda K, Nakayama K and Nakanishi M. Aberrant cell cycle checkpoint function and early embryonic deathin Chk1(-/-) mice. Genes Dev. 2000, 14:1439–1447
    118.Liu Q, Guntuku S, Cui XS, et al. Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint.Genes Dev. 2000; 14: 1448– 1459.
    119.Yan Y, Black CP and Cowan KH. Irradiation-induced G2/M checkpoint response requires ERK1/2 activation. Oncogene. 2007; doi: 10.1038
    120.Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent andspecific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998;391:806-811
    121.Brummelkamp TR, Bernards R, Agami R. Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell .2002;2:243-247
    122.McCaffrey AP, Meuse L, Pham TT, Conklin DS, Hannon GJ, Kay MA. RNA interference in adult mice. Nature 2002;418: 38-39
    123.Hung L, Kumar V. Specific inhibition of gene expression and transactivation functions of hepatitis B virus X protein and c-myc by small interfering RNAs. FEBS Letters,2004; 560:210-214
    124.Yin JQ, Gao GS,Shao RG, et al.siRNAagents inhibit oncogene expression and attenuate human tumor cell growth. J Exp Ther Oncol, 2003;3(4):194-204
    125.Carpenter AE, Sabatini DM. Systematic genome-wide screens of gene function. Nat Rev Genet, 2004;5(1):11-22
    1. Gaudin D, Yielding KL. Response of a "resistant" plasmacytoma to alkylating agents and x-ray in combination with the "excision" repair inhibitors caffeine and chloroquine. Proc Soc Exp Biol, 1969; 131:1413-6.
    2. Shimada K, Takagi Y. The effect of caffeine on the repair of ultraviolet-damaged DNA in bacteria. Biochim Biophys Acta, 1967; 145:763-70.
    3. Weinert T, Hartwell L. Control of G2 delay by the rad9 gene of Saccharomyces cerevisiae. J Cell Sci Suppl, 1989; 12:145-8.
    4. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell, 1997; 88: 323–31.
    5. Suganuma M, Kawabe T, Hori H, Funabiki T, Okamoto T. Sensitization of cancer cells to DNA damage-induced cell death by specific cell cycle G2 checkpoint abrogation. Cancer Res, 1999;59:5887–91.
    6. Russell KJ, Wiens LW, Demers GW, Galloway DA, Plon SE, Groudine M. Abrogation of the G2 checkpoint results in differential radiosensitization of G1 checkpoint-deficient and G1 checkpoint-competent cells. Cancer Res, 1995; 55: 1639-42.
    7. Powell SN, DeFrank JS, Connell P, et al. Differential sensitivity of p53(–) and p53(+) cells to caffeine-induced radiosensitization and override of G2 delay. Cancer Res, 1995; 55:1643-8.
    8. Ribeiro JC, Barnetson AR, Jackson P, Ow K, Links M, Russell PJ. Caffeine-increased radiosensitivity is not dependent on a loss of G2/M arrest or apoptosis in bladder cancer cell lines. Int J Radiat Biol, 1999; 75:481-92.
    9. Asaad NA, Zeng ZC, Guan J, Thacker J, Iliakis G. Homologous recombination as a potential target for caffeine radiosensitization in mammalian cells: reduced caffeine radiosensitization in XRCC2 and XRCC3 mutants. Oncogene, 2000; 19: 5788-800.
    10. Levine AJ. The p53 tumor suppressor gene and gene product. Princess TakamatsuSymp, 1989; 20:221–30.
    11. Weston A, Harris CC. Chemical carcinogenesis. In: Bast RC Jr, Kufe DW, Pollock RE, Weichselbaum RR, Holland JF, Frei E III, editors. Cancer medicine e.5. Lewiston, NY:B.C. Decker Inc.; 2000. p. 185-94.
    12. Brose MS, Smyrk T, Weber B, Lynch HT. Genetic predisposition to cancer. In: Bast RC Jr, Kufe DW, Pollock RE, Weichselbaum RR, Holland JF, Frei E III, editors. Cancer medicine e.5. Lewiston, NY:B.C. Decker Inc.; 2000. p. 185–94.
    13. Sherr CJ. Cancer cell cycles. Science (Washington DC), 1996; 274:1672-7.
    14. Russell P. Checkpoints on the road to mitosis. Trends Biochem Sci, 1998; 23: 399-402.
    15. Volkmer E, Karnitz L. Human homologs of Schizosaccharomyces pombe Rad1, Hus1, and Rad9 form a DNA damage-responsive protein complex. J Biol Chem, 1999; 274:567-70.
    16. St Onge RP, Udell CM, Casselman R, Davey S. The human G2 checkpoint control protein hRAD9 is a nuclear phosphoprotein that forms complexes with hRAD1 and hHUA1. Mol Biol Cell, 1999; 10:1985-95.
    17. Bao S, Tibbetts RS, Brumbaugh KM, et al. ATR/ATM-mediated phosphorylation of human Rad17 is required for genotoxic stress responses. Nature (Lond), 2001; 411:969–74.
    18. Sanchez Y, Wong C, Thoma RS, et al. Conservation of the Chk1 checkpoint pathway in mammals: linkage of DNA damage to Cdk regulation through Cdc25. Science (Washington DC), 1997; 277:1497–501.
    19. Peng CY, Graves PR, Thoma RS, Wu Z, Shaw AS, Pownica-Worms H. Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216. Science (Washington DC), 1997;277:1501–5.
    20. Matsuoka S, Huang M, Elledge SJ. Linkage of ATM to cell cycle regulation by the Chk2 protein kinase. Science (Washington DC), 1998; 282:1893–7.
    21. Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev, 2001; 15:2177–96.
    22. Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, Qin J. BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA. Genes Dev, 2000; 14:927–39.
    23. Fernandez-Capetillo O, Chen HT, Celeste A, et al. DNA damage-induced G2-M checkpoint activation by histone H2AX and 53BP1. Nat Cell Biol, 2002;4:993–7.
    24. Peng A, Chen PL. NFBD1, like 53BP1, is an early and redundant transducer mediating Chk2 phosphorylation in response to DNA damage. J Biol Chem, 2003; 78:8873–6.
    25. Iwabuchi K, Bartel PL, Li B, Marraccino R, Fields S. Two cellular proteins that bind to wild-type but not mutant p53. Proc Natl Acad Sci USA, 1994; 91: 6098- 102.
    26. Hawn MT, Umar A, Carethers JM, et al. Evidence for a connection between the mismatch repair system and the G2 cell cycle checkpoint. Cancer Res, 1995; 55: 3721-5.
    27. Kobayashi J, Tauchi H, Sakamoto S, et al. NBS1 localizes to -H2AX foci through interaction with the FHA/BRCT domain. Curr Biol, 2002; 12:1846-51.
    28. Lopez-Girona A, Kanoh J, Russell P. Nuclear exclusion of Cdc25 is not required for the DNA damage checkpoint in fission yeast. Curr Biol, 2001; 11:50–4.
    29. Falck J, Mailand N, Syljuasen RG, Bartek J, Lukas J. The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature (Lond), 2001; 410:842-7.
    30. Bell DW, Varley JM, Szydlo TE, et al. Heterozygous germ line hCHK2 mutations in Li-Fraumeni syndrome. Science (Washington DC), 1999;286:2528–31.
    31. Hirao A, Kong YY, Matsuoka S, et al. DNA damage-induced activation of p53 by the checkpoint kinase Chk2. Science (Washington DC), 2000;287:1824–7.
    32. Takai H, Tominaga K, Motoyama N, et al. Aberrant cell cycle checkpoint function and early embryonic death in Chk1(–/–) mice. Genes Dev, 2000;14:1439–47.
    33. Liu Q, Guntuku S, Cui XS, et al. Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint. Genes Dev, 2000;14:1448–59.
    34. Chen Z, Xiao Z, Chen J, et al. Human Chk1 expression is dispensable for somatic cell death and critical for sustaining G2 DNA damage checkpoint. Mol Cancer Ther, 2003;2:543–8.
    35. Menoyo A, Alazzouzi H, Espin E, Armengol M, Yamamoto H, Schwartz S Jr. Somatic mutations in the DNA damage-response genes ATR and CHK1 in sporadic stomach tumors with microsatellite instability. Cancer Res, 2001;61:7727–30.
    36. Vassileva V, Millar A, Briollais L, Chapman W, Bapat B. Genes involved in DNA repair are mutational targets in endometrial cancers with microsatellite instability. Cancer Res, 2002;62:4095–9.
    37. Bulavin DV, Higashimoto Y, Popoff IJ, et al. Initiation of a G2/M checkpoint after ultraviolet radiation requires p38 kinase. Nature (Lond), 2001;411:102–7.
    38. Duckworth BC, Weaver JS, Ruderman JV. G2 arrest in Xenopus oocytes depends on phosphorylation of cdc25 by protein kinase A. Proc Natl Acad Sci USA, 2002;99:16794–9.
    39. Mailand N, Falck J, Lukas C, et al. Rapid destruction of human Cdc25A in response to DNA damage. Science (Washington DC), 2000;288:1425–9.
    40. Galaktionov K, Lee AK, Eckstein J, et al. CDC25 phosphatases as potential human oncogenes. Science (Washington DC), 1995;269:1575–7.
    41. van Vugt MA, Smits VA, Klompmaker R, Medema RH. Inhibition of Polo-like kinase-1 by DNA damage occurs in an ATM- or ATR-dependent fashion. J Biol Chem, 2001;276:41656–60.
    42. Murray A, Hunt T. Post-translational regulation of mitosis. The cell cycle. Newyork,NY:Oxford Univ. Press; 1993. p. 56–65.
    43. Ando T, Kawabe T, Ohara H, Ducommun B, Itoh M, Okamoto T. Involvement of the interaction between p21 and proliferating cell nuclear antigen for the maintenance of G2/M arrest after DNA damage. J Biol Chem, 2001;276:42971–7.
    44. Kawabe T, Suganuma M, Ando T, Kimura M, Hori H, Okamoto T. Cdc25C interacts with PCNA at G2/M transition. Oncogene, 2002;21:1717–26.
    45. Hermeking H, Lengauer C, Polyak K, et al. 14-3-3 sigma is a p53-regulated inhibitor of G2/M progression. Mol Cell, 1997;1:3–11.
    46. Umbricht CB, Evron E, Gabrielson E, Ferguson A, Marks J, Sukumar S. Hypermethylation of 14-3-3 sigma (stratifin) is an early event in breast cancer. Oncogene, 2001;26:3348–53.
    47. Qi W, Martinez JD. Reduction of 14-3-3 proteins correlates with increased sensitivity to killing of human lung cancer cells by ionizing radiation. Radiat Res, 2003;160:217–23.
    48. Qiu L, Burgess A, Fairlie DP, Leonard H, Parsons PG, Gabrielli BG. Histone deacetylase inhibitors trigger a G2 checkpoint in normal cells that is defective in tumor cells. Mol Biol Cell, 2000;11:2069–83.
    49. Cai RL, Yan-Neale Y, Cueto MA, Xu H, Cohen D. HDAC1, a histone deacetylase, forms a complex with Hus1 and Rad9, two G2/M checkpoint Rad proteins. J Biol Chem, 2000;275:27909–16.
    50. Taylor WR, Stark GR. Regulation of the G2/M transition by p53. Oncogene, 2001;20:1803–15.
    51. Williams M, Jarvis MF. Adenosine antagonists as potential therapeutic agents. Pharmacol Biochem Behav, 1988;29:433–41.
    52. Mante S, Minneman KP. Caffeine inhibits forskolin-stimulated cyclic AMP accumulation in rat brain. Eur J Pharmacol, 1990;175:203–5.
    53. Sarkaria JN, Busby EC, Tibbetts RS, et al. Inhibition of ATM and ATR kinaseactivities by the radiosensitizing agent, caffeine. Cancer Res, 1999;59:4375–82.
    54. Cortez D. Caffeine inhibits checkpoint responses without inhibiting the ataxia-telangiectasia-mutated (ATM) and ATM- and Rad3-related (ATR) protein kinases. J Biol Chem, 2003;39:37139–45.
    55. Samlaska CP, Winfield EA. Pentoxifylline. J Am Acad, 1994;30:603–21.
    56. Bohm L, Roos WP, Serafin AM. Inhibition of DNA repair by Pentoxifylline and related methylxanthine derivatives. Toxicology, 2003;193:153–60.
    57. Tam SW, Schlehel R. Staurosporine overrides checkpoints for mitotic onset in BHK cells. Cell Growth & Differ, 1992;3:811–7.
    58. Wang Q, Fan S, Eastman A, Worland PJ, Sausville EA, O'Connor PM. UCN-01: a potent abrogator of G2 checkpoint function in cancer cells with disrupted p53. J Natl Cancer Inst, 1996;88:956–65.
    59. Kohn EA, Yoo CJ, Eastman A. The protein kinase C inhibitor Go6976 is a potent inhibitor of DNA damage-induced S and G2 cell cycle checkpoints. Cancer Res, 2003;63:31–5.
    60. Jackson JR, Gilmartin A, Imburgia C, Winkler JD, Marshall LA, Roshak A. An indolocarbazole inhibitor of human checkpoint kinase (Chk1) abrogates cell cycle arrest caused by DNA damage. Cancer Res, 2000;60:566–72.
    61. Eastman A, Kohn EA, Brown MK, et al. A novel indolocarbazole, ICP-1, abrogates DNA damage-induced cell cycle arrest and enhances cytotoxicity: similarities and differences to the cell cycle checkpoint abrogator UCN-01. Mol Cancer Ther, 2002;1:1067–78.
    62. Senderowicz AM. Novel small molecule cyclin-dependent kinases modulators in human clinical trials. Cancer Biol Ther, 2003;2:S84–95.
    63. Fuse E, Tanii H, Kurata N, et al. Unpredicted clinical pharmacology of UCN-01 caused by specific binding to human 1-acid glycoprotein. Cancer Res, 1998;58:3248–53.
    64. Sorensen CS, Syljuasen RG, Falck J, et al. Chk1 regulates the S phase checkpoint by coupling the physiological turnover and ionizing radiation-induced accelerated proteolysis of Cdc25A. Cancer Cell, 2003;3:247–58.
    65. Curman D, Cinel B, Williams DE, et al. Inhibition of the G2 DNA damage checkpoint and of protein kinases Chk1 and Chk2 by the marine sponge alkaloid debromohymenialdisine. J Biol Chem, 2001;276:17914–9.
    66. Yu Q, La Rose J, Zhang H, Takemura H, Kohn KW, Pommier Y. UCN-01 inhibits p53 up-regulation and abrogates -radiation-induced G(2)-M checkpoint independently of p53 by targeting both of the checkpoint kinases, Chk2 and Chk1. Cancer Res, 2002;62:5743–8.
    67. Roberge M, Berlinck RG, Xu L, et al. High-throughput assay for G2 checkpoint inhibitors and identification of the structurally novel compound isogranulatimide. Cancer Res, 1998;58:5701–6.
    68. Wang Y, Li J, Booher RN, et al. Radiosensitization of p53 mutant cells by PD0166285, a novel G(2) checkpoint abrogator. Cancer Res, 2001;61:8211–7.
    69. Ghosh S, Schroeter D, Paweletz N. Okadaic acid overrides the S-phase check point and accelerates progression of G2-phase to induce premature mitosis in HeLa cells. Exp Cell Res, 1996;227:165–9.
    70. Roberge M, Tudan C, Hung SM, Harder KW, Jirik FR, Anderson H. Antitumor drug fostriecin inhibits the mitotic entry checkpoint and protein phosphatases 1 and 2A. Cancer Res, 1994;54:6115–21.
    71. Britton R, Roberge M, Brown C, van Soest R, Andersen RJ. New okadaic acid analogues from the marine sponge Merriamum oxeato and their effect on mitosis. J Nat Prod, 2003;66:838–43.
    72. Rundle NT, Xu L, Andersen RJ, Roberge M. G2 DNA damage checkpoint inhibition and antimitotic activity of 13-hydroxy-15-oxozoapatlin. J Biol Chem, 2001;276:48231–6.
    73. Russell KJ, Wiens LW, Demers GW, et al. Preferential radiosensitization of G1 checkpoint-deficient cells by methylxanthines. Int J Radiat Oncol Biol, 1996;36:1099–106
    74. Graves PR, Yu L, Schwarz JK, et al. The Chk1 protein kinase and the Cdc25C regulatory pathways are targets of the anticancer agent UCN-01. J Biol Chem, 2000;275:5600–5.
    75. Sato S, Fujita N, Tsuruo T. Interference with PDK1-Akt survival signaling pathway by UCN-01 (7-hydroxystaurosporine). Oncogene, 2002;21:1717–38.
    76. Ishihara H, Martin BL, Brautigan DL, et al. Calyculin A and okadaic acid: inhibitors of protein phosphatase activity. Biochem Biophys Res Commun, 1989;159:871–7.
    77. Walsh AH, Cheng A, Honkanen RE. Fostriecin, an antitumor antibiotic with inhibitory activity against serine/threonine protein phosphatases types 1 (PP1) and 2A (PP2A), is highly selective for PP2A. FEBS Lett, 1997;416:230–40.
    [1] Hurley, L. H. DNA and its associated processes as targets for cancer therapy. Nat. Rev. Cancer 2002, 2(3), 188-200.
    [2] Thuong, N., Hélène, C. Angew. Chem. Int. Ed. Engl. 1993, 32, 666.
    [3] Dickinson, L. A., Burnett, R., Melander, C., Edelson, B. S., Arora,P. S., Dervan, P. B., Gottesfeld, J. M. Arresting cancer proliferation by small-molecule gene regulation. Chem. Biol. 2004, 11(11), 1583-94.
    [4] Shinohara, K.-I., Narita, A., Oyoshi, T., Bando, T., Teraoka, H.,Sugiyama, H. Sequence-specific gene silencing in mammalian cells by alkylating pyrrole-imidazole polyamides J. Am. Chem. Soc. 2004,126(16):5113-8
    [5] Han, H., Hurley, L. H. G-quadruplex DNA: a potential target for anti-cancer drug design Trends Pharmacol. Sci. 2000, 21(4), 136-42.Review
    [6] Maloney, D. J., Deng, J.-Z., Starck, S. R., Gao, Z., Hecht, S. M. J.Am. Chem. Soc. (+)-Myristinin A, a naturally occurring DNA polymerase beta inhibitor and potent DNA-damaging agent . 2005,127(12):4140-1.
    [7] Sancar, A., Lindsey-Boltz, L. A., Unsal-Kacmaz, K., Linn, S.Annu. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints Annu Rev Biochem. 2004,73:39-85.
    [8] Vassilev, L. T. J. p53 Activation by small molecules: application in oncology Med. Chem. 2005, 48(14):4491-9.
    [9] Zhou, B. –B., Bartek, J. Targeting the checkpoint kinases: chemosensitization versus chemoprotection. Nature Rev. Cancer.2004, 4(3): 216-25.
    [10] Prudhomme, M. Curr. Med. Chem. –Anti-Cancer Agents Combining DNA damaging agents and checkpoint 1 inhibitors 2004, 4(5):435-8.
    [11] Kong, N., Fotouhi, N., Wovkulich, P. M., Roberts, J. Drugs Future 2003, 28, 881.
    [12] Kastan, M. B., Bartek, J. Cell-cycle checkpoints and cancer .Nature 2004,432(7015): 316-23.
    [13] Li, Q., Zhu, G. Curr. T argeting serine/threonine protein kinase B/Akt and cell-cycle checkpoint kinases for treating cancer. Top. Med. Chem. 2002,2(9), 939-71.
    [14] Kawabe, T. Mol. G2 checkpoint abrogators as anticancer drugs.Mol Cancer Ther. 2004, 3(4):513-9.
    [15] Tentori, L., Portarena, I., Graziani, G. Pharmaco. Potential clinical applications of poly (ADP-ribose) polymerase (PARP) inhibitors Res. 2002, 45(2), 73-85.
    [16] Walwroth N, Davery S, Beach D. Fission yeast chk1 protein kinase links the rad checkpoint pathway to cdc2. Nature.1993,363(6427):368-71.
    [17] Chen Y, Sanchez Y. Chk1 in DNA damage response:conserved roles from yeast to mammals. DNA Repair, 2004, 3:1 025-32.
    [18] Yolanda S, Calvin W, Richard ST,et al. Conservation of chk1 checkpoint pathway in mammals:linkage of DNA damage to cdk regulation through cdc25. Science, 1997, 277:1497-501.
    [19] Chen P, Luo C,Deng Y,et al. The1.7 crystal structure of human cell cycle checkpoint kinase chk1:implications for chk1 regulation. Cell, 2000, 100:681-92.
    [20] Galvan V, Kurakin AV,Bredese DE. Interaction of checkpoint kinase 1 and the X-linked inhibitor of apoptosis during mitosis. FEBS Lett, 2004, 558(1-3):57-62.
    [21] Zhao H, Piwnica-Worms H. ATR-mediated checkpoint pathways regulate phosphorylation and activation of human chk1. Mol Cell Biol, 2001, 21:4 129-39.
    [22] Gatei M,Sloper K,Sorensen C,et al. ATM and DBS1 dependent phosphorylation of chk1 on S317 in response to IR. J Biol Chem, 2003, 278(17):14806-11.
    [23] Shiloh Y. ATM and related protein kinases:safeguarding genome integrity. NatRev Cancer, 2003, 3:155-68.
    [24] Shann YJ, Hsu MT. Cloning and characterization of liverspecific isoform of chk1 gene from rat. J Biol Chem, 2001, 276(52):48863-70.
    [25] Katsuragi Y, Sagata N. Regulation of chk1 kinase by autoinhibition and ATR-mediated phosphorylation. Mol Biol Cell, 2004, 15:1680-89.
    [26] Sancar, A., Lindsey-Boltz, L. A., Unsal-Kacmaz, K., Linn, S.Annu. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Rev. Biochem, 2004, 73, 39-85.
    [27] Nasmyth, K. Viewpoint: putting the cell cycle in order. Science, 1996, 274(5293):1643-5.
    [28] Kastan, M. B., Bartek, J. Cell-cycle checkpoints and cancer. Nature, 2004, 432(7015):316-23.
    [29] Massague, J. G1 cell-cycle control and cancer. Nature 2004,432 (7015):298-306.
    [30] Nelson, W.G., Lim, D. –S. DNA strand breaks: the DNA template alterations that trigger p53-dependent DNA damage response pathways.Mol Cell Biol. 1994, 14(3):1815-23.
    [31] Sherr, C. J., McCormick, F. The RB and p53 pathways in cancer. Cancer Cell. 2002, 2(2):103-12.
    [32] Wahl GM, Carr AM. The evolution of diverse biological responses to DNA damage: insights from yeast and p53.Nat Cell Biol. 2001 Dec;3(12):E277-86. Erratum in: Nat Cell Biol 2002 Apr; 4(4):328.
    [33] Molinari, M, Mercurio, C., Dominguez, J., Goubin, F., Draetta, G.F. Human Cdc25 A inactivation in response to S phase inhibition and its role in preventing premature mitosis. EMBO Rep. 2000, 1(1):71-9.
    [34] Falck J, Mailand N, Syljuasen RG, Bartek J, Lukas J. The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis.Nature. 2001,410(6830):842-7.
    [35] Donzelli, M., Draetta, G. F. Regulating mammalian checkpoints through Cdc25 inactivation. EMBO Rep. 2003, 4(7), 671-7
    [36] Kumagai A, Guo Z, Emami KH,et al. The Xenopus chk1protein kinase mediates a caffeine-sensitive pathway of checkpoint control in cell-free extracts. J Cell Biol, 1998, 142:1 559-69.
    [37] Okubo E, Lehman JM,Friedrich TD. Negative regulation of mitotic promoting factor by the checkpoint kinase chk1 in simian virus 40 lytic infection. J Virol, 2003, 77:1 257-67.
    [38] den Elzen N,Kosoy A,Christopoulos H,et al. Resisting Arrest:recovery from checkpoint arrest through dephosphorylation of chk1 by PP1. Cell Cycle, 2004, 3:5 29-33.
    [39] Guo Z, Dunphy WG. Response of Xenopus Cds1 in cellfree extracts to DNA templates with double-stranded ends. Mol Biol Cell, 2000,11:1 535-46.
    [40] Bertoni F,Codegoni AM,Furlan D,et al. Chk1 frameshift mutations in genetically unstable colorectal and endometrial cancers. Genes Chromosomes Cancer, 1999, 26:176-80.
    [41] Menoyo A,Alazzouzi H,Espin E,et al. Somatic mutations in the DNA damage-response genes ATR and CHK1 in sporadic stomach tumors with microsatellite instability.Cancer Res,2001,61:7 727-30.
    [42] Haruki N,Saito H,Tatematsu Y,et al. Histological typeselective, tumor- predominant expression of novel chk1 isoform and infrequent in vivo somatic chk2 mutation in small cell lung cancer. Cancer Res, 2000, 60:4 689-92.
    [43] George Z, Michael DR,David AFG. Chk1-deficient tumor cells are viable but exhibit multiple checkpoint and survival defect. EMBO J, 2003, 22:713-23.
    [44] Wang Y, Decker SJ,Sebolt-Leopold J. Knockdown of chk1,Wee1 and Myt1 by RNA interference abrogates G(2)checkpoint and induces apoptosis. CancerBiol Ther, 2004, 3:305-13.
    [45] Chen Z,Xiao Zh,Chen J,et al. Human chk1 expression is dispensable for somatic cell death and critical for sustaining G2 DNA damage checkpoint. Mol Cancer Ther, 2003, 2:543-48.
    [46] Carrassa L, Broggini M,Erba E,et al. Chk1,but not Chk2,is involved in the cellular response to DNA damaging agents:differential activity in cells expressing,or not p53. Cell Cycle, 2004, 3(9):1 177-81.
    [47] Lam MH, Liu Q,Elledge SJ,Chk1 is haploinsufficient for multiple functions critical to tumor suppression. Cancer Cell, 2004, 6:45-58.
    [48] Liu Q,Guntuku S,Cui XS,et al. Chk1 is an essential kinase that is regulated by Atr and required for the G2/M DNA damage checkpoint. Genes Dev, 2000, 14:1 448-59.
    [49] Yin MB,Hapke G,Wu J,et al. Chk1 signaling pathways that mediated G2 / M check- point in relation to cellular resistance to the novel topoisomerase I poison BNP1350. Biochem Biophys Res Commun, 2002, 295:435-44.
    [50] Hattori H, Kuroda M,Ishida T,et al. Human DNA damage checkpoints and their relevance to soft tissue sarcoma. Int Pathol, 2004, 54:26-31.
    [51] Bartek, J., Lukas, J. Chk1 and Chk2 kinases in checkpoint control and cancer.Cancer Cell. 2003, 3(5):421-9.
    [52] Liu, Q., Guntuku, S., Cui, X., Matsuoka, S, Cortez, D., Tamai, K.,Luo, G., Carattini-Rivera, S., DeMayo, F., Bradley, A., Donehower, L.A., Elledge, S. J. Genes Dev. 2000, 14, 1448.
    [53] Takai, H., Tominaga, K., Motoyama, N., Minamishima, Y.A., Nagahama, H., Tsukiyama, T., Ikeda, K., Nakayama, K., Nakanishi, M., Nakayama, K. Aberrant cell cycle checkpoint function and early embryonic death in Chk1(-/-) mice. Genes Dev. 2000, 14(12), 1439-47.
    [54] Chen, Z., Xiao, Z., Chen, J., Ng, S. C., Sowin, T, J., Sham, H., Rosenberg, S.,Fesik, S., Zhang, H. Mol. Cancer. Ther. 2003, 2,543.
    [55] Arienti, K.L., Brunmark, A., Axe, F.U., McClure, K., Lee, A.,Blevitt, J., Neff, D.K., Huang, L., Crawford, S., Pandit, C.R.,Karlsson, L., Breitenbucher, J. G. Checkpoint kinase inhibitors: SAR and radioprotective properties of a series of 2-arylbenzimidazoles.J Med Chem. 2005 Mar 24,48(6):1873-85. J. Med. Chem. 2005, 48, 1873.
    [56] Pommier, Y., Sordet, O., Rao, A.V., Zhang, H., Kohn, K.W. Targeting chk2 kinase: molecular interaction maps and therapeutic rationale. Curr.Pharm. Des. 2005, 11(22), 2855-72.

© 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