Tautomycetin对乳腺癌细胞增殖的抑制作用及机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
Tautomycetin (TMC)最初是从土壤链霉菌中分离得到的具有抗真菌活性的聚酮化合物。近几年又发现,TMC是蛋白磷酸酶PP1特异性抑制剂,具有免疫抑制活性,并且可以特异性抑制结肠癌细胞生长。但是,TMC抑制肿瘤细胞生长是否与其抑制PP1活性相关未得到证明,抑制肿瘤细胞生长的机制还未详细阐明。我们的前期实验发现TMC并不是特异性抑制结肠癌细胞生长,它对乳腺癌等多种细胞系具有显著生长抑制作用。
     本研究阐明了TMC的肿瘤细胞生长抑制活性与PP1抑制活性之间是否相关,并重点研究了TMC抑制乳腺癌细胞生长的信号通路,具体研究结果及结论如下:
     (1)采用MTT和磷酸酶PP1活性测定等方法,测定TMC及其不同衍生物的磷酸酶PP1抑制、肿瘤细胞生长抑制和免疫抑制活性。结果显示:多个衍生物具有较强的PP1抑制活性,却没有显著的肿瘤细胞生长抑制和免疫抑制活性;而部分衍生物具有较强的肿瘤细胞生长抑制活性,PP1抑制活性却不显著;TMC1μmol/L和衍生物TMC-F150μmol/L都能抑制MCF-7细胞内PP160%的活性,而对MCF-7细胞增殖的抑制率分别为90%和24%。表明TMC的肿瘤细胞生长抑制与免疫抑制活性不是通过其蛋白磷酸酶PP1抑制活性实现的。
     (2)采用MTT、Transwell细胞迁移和侵袭实验和流式细胞术等方法检测TMC对MCF-7细胞增殖、侵袭和凋亡的影响。结果显示:TMC抑制MCF-7细胞增殖的ICso值为0.36μmol/L; TMC0.4μmol/L抑制MCF-7细胞迁移率和侵袭率分别为76%和79%;TMC0.5μmol/L诱导MCF-7细胞早期凋亡和晚期凋亡的比率分别为21%和34%;Western blotting检测TMC对凋亡相关蛋白表达。结果显示:TMC可以显著降低Bcl-2表达量和Bad磷酸化水平,提高Bax表达量和cytochrome-c线粒体释放量,caspase-9和caspase-7被活化。以上结果表明,TMC通过调控Bcl-2家族,依赖线粒体/细胞色素c介导的凋亡信号激活下游caspase级联通路,最终诱导MCF-7细胞凋亡。
     (3)采用MTT、流式细胞术和Western blotting等方法,检测TMC对耐阿霉素乳腺癌细胞MCF-7/ADR细胞增殖等影响。结果显示:TMC抑制MCF-7/ADR细胞生长的ICso值为1.26μmol/L; TMC1μmol/L诱导MCF-7细胞早期凋亡和晚期凋亡的比率分别为17.2%和28.4%;TMC同样可以显著降低Bcl-2蛋白表达量,激活caspase-9和caspase-7o以上结果表明,TMC对MCF-7/ADR细胞同样敏感,并通过依赖线粒体/细胞色素c介导凋亡通路诱导MCF-7/ADR细胞凋亡。
     (4)采用Western blotting、双荧光素酶报告基因等实验检测TMC对MAPK (ERK、p-38和JNK)、Wnt、p53和Akt信号通路相关蛋白磷酸化和转录因子活性的影响。发现TMC可以显著降低Akt及其下游调控蛋白FKHR的磷酸化,并且0.4μmol/L TMC使Akt下游调控的转录因子CREB转录活性降低70%;TMC同时可以显著抑制上游直接调控Akt的PDK1激酶磷酸化,但是对EGFR和HER2酪氨酸激酶受体磷酸化无显著影响。进一步观察转染Akt持续激活质粒myr-Akt对TMC抑制MCF-7增殖的影响。结果显示:0.5μmol/L TMC对转染空载体和myr-Akt质粒的MCF-7细胞的增殖抑制率分别为64%和31%。以上结果表明,TMC通过阻断MCF-7细胞中Akt信号通路,从而调控下游细胞生存和凋亡相关蛋白及转录因子活性。
     (5)通过siRNA技术将PP1基因沉默,检测对Akt磷酸化的影响。结果显示:PP1基因沉默并没有显著影响Akt激酶的磷酸化水平。以上结果进一步表明TMC抑制MCF-7细胞增殖与其抑制PP1活性无主要关联。
     (6)采用MTT、Western blotting和细胞转染等方法,检测TMC与Leptomycin B (LMB)联合用药是否具有协同效应及其机制。结果显示:LMB先加24h再加TMC共同作用协同效应较好,协同效应指数CI值为0.38-0.49;联合用药达到半数抑制时,TMC和LMB的用量分别为单独用药时的23.8%和17.7%;TMC和LMB联用可以增强TMC对Akt和FKHR等蛋白磷酸化的下调作用;LMB可以增强TMC诱导的FKHR细胞核聚集。以上结果表明,TMC与LMB联用对抑制MCF-7细胞增殖具有显著协同效应,协同效应与LMB促进TMC诱导的FKHR细胞核聚集有关联。
     综上,论文研究发现TMC的抗肿瘤活性与PP1抑制活性之间的相关性较低;阐明了TMC诱导MCF-7细胞凋亡的机制;证明Akt和FKHR抑制剂类药物与核输出抑制剂联用具有协同效应。研究工作为TMC药物机制提供了新的例证,为进一步确定TMC抗肿瘤的直接作用靶点奠定了基础;同时,为TMC联合用药提供了一个新的设计思路。
Tautomycetin (TMC) is polyketide natural product originally isolated as antifungal antibiotics from Streptomyces griseochromogens. TMC was later found to be able to specifically inhibit protein phosphatase type1(PP1). TMC has been also identified as a potent immunosuppressor and a potential drug for colorectal cancer. The anticancer activities of TMC are both thought to be related to its ability to inhibit protein phosphatases. Unfortunately, this hypothesis has not yet been fully clarified. Our studies showed that TMC could inhibit proliferation of different kinds of cancer cells in vitro.
     In this study, we presented the correlation between the anti-cancer activity of TMC and the inhibitory activity of PP1, with the focus on the signaling pathway of the anticancer activity of TMC. The main results and conclusions were listed as follows:
     (1) Phosphatase inhibition, immunosuppressive and anticancer activities of TMC and its analogues were determinded. The results showed that some analogues of TMC exhibited strong PP1-inhibiting activity rather than anti-tumor activity and immunosuppressive activity; some analogues exhibited cytotoxic activity rather than PP1-inhibiting activity. TMC (1μmol/L) and TMC-F1(50μmol/L) could both inhibited PPl activity in MCF-7cell lines by60%, but with90%and24%inbibiting percentages of MCF-7cell proliferation, respectively. Therefore, the anti-tumor activity and immunosuppressive activity were not correlated with the inhibitory activity of protein phosphatase PP1.
     (2) The effects of TMC on the MCF-7cell proliferation, invasion and apoptosis were examined by MTT, cell invasion assays and flow cytometry analysis, respectively. The estimated IC50value was0.36umol/L in MCF-7cell lines; the percentages of cell migration and invasion of MCF-7cell lines inhibiting by0.4μmol/L TMC were76%and79%, respectively; the percentages of MCF-7cell early and advanced apoptosis induced by0.5μmol/L TMC were21%and34%, respectively. Furthermore, we examined the effects of TMC on the apoptosis relative protein expression by western blotting. The results indicated that the expression level of Bcl-2and phosphorylation of Bad were both decreased, expression of Bax and mitochondria release of cytochrome-c were enhanced, caspase-9and caspase-7were activated by TMC treatment. Therefore, it was argued that TMC could activate caspase cascade pathway through the regulations of Bcl-2family and mitochondrial/cytochrome-c mediated apoptosis pathway, which could induce the apoptosis of MCF-7cells.
     (3) Detecting the influence of TMC on Adriamycin resistant human breast adenocarcinoma MCF-7/ADR cells by MTT and Western blotting. The results showed that TMC inhibited the proliferation of MCF-7/ADR cells with the IC50values being1.26μmol/L. The early and late apoptosis rate of MCF-7/ADR cells induced by1μmol/L TMC increased to17.2%and28.4%, respectively. Similarly. TMC could significantly decrease the phosphorylation of Akt in MCF-7/ADR cells and the expression of Bcl-2. while activate caspase-9and caspase-7. It indicated that TMC is sensitive to MCF-7/ADR cells and could activate caspase cascade pathway through the regulations of Bcl-2family and mitochondrial/cytochrome-c mediated apoptosis pathway, which could induce the apoptosis of MCF-7/ADR cells.
     (4) The effects of TMC on the relative protein phosphorylations and transcription factor activities of MAPK (ERK. p-38and JNK), Wnt. p53and Akt signal pathway were examined by Western blotting and dual luciferase assay system. The results showed that the phosphorylation of Akt and its downstream target FKHR were decreased significantly, and the transcription activity of CREB were also decreased by70%. Meanwhile, the phosphorylation of kinase PDK1was inhibited by TMC, but those of tyrosine kinase EGFR and HER2were not. Next, we examined the role of Akt in TMC-mediated inhibition. We found that TMC inhibited proliferation of MCF-7cells transfected with blank vector and myr-Akt plasmid by64%and31%, respectively. We demonstrated that TMC inhibited Akt and its downstream signaling, subsequently resulting in apoptosis of MCF-7cells.
     (5) The gene PP1was silenced by siRNA technology, then the influence on phosphorylation of Akt was detected and no significant change was observed. It was further indicated that the inhibitory of TMC on MCF-7was not clearly correlated with the inhibition of PP1.
     (6) To investigate whether there is synergistic effect between TMC and Leptomycin B (LMB), MTT, Western blotting and Cell transfection assays were conducted. The results showed that the synergistic effect was better by adding LMB first and TMC was added24h later, with a CI value0.38-0.49. In combination treatment, the required dosage of TMC and LMB for half-inhibition is reduced to23.8%and17.7%compared to single treatment. LMB could enhance the down-regulating effect of TMC on phosphorylation of Akt and FKHR. LMB could also enhance the gathering of nuclear induced by TMC. It was implied that combination treatment of TMC and LMB has notable synergistic effect on the inhibitory of MCF-7cell growth. The effect may be related to the function of LMB to promote the nuclear gathering which is induced by TMC.
     In summary, our results indicated that cytotoxicity of TMC was not associated with its PP1inhibition activity. Moreover, we provided a plausible mechanism of TMC inducing the MCF-7cell apoptosis and proved that there was a synergistic antitumor activity of Akt inhibitors combined with nuclear export inhibitors. The results of this study proposed a new paradigm for investigating the mechanism of the anti-tumor drugs which inhibit protein phosphatase, laid the foundation for further identifying the direct target of its anti-tumor activity. Lastly, our study provided a new design strategy for rational drug combination.
引文
[1]DELONG A. Switching the flip:protein phosphatase roles in signaling pathways [J]. Current opinion in plant biology,2006,9(5):470-7.
    [2]FUJIKI H, SUGANUMA M. Carcinogenic aspects of protein phosphatase 1 and 2A inhibitors [J]. Marine Toxins as Research Tools,2009,221-54.
    [3]CHENG X C, KIHARA T, YING X, et al. A new antibiotic, tautomycetin [J]. Journal of Antibiotics, 1989,42(1):141-4.
    [4]CHENG X C, UBUKATA M. ISONO K. The structure of tautomycetin, a dialkylmaleic anhydride antibiotic [J]. The Journal of antibiotics,1990,43(7):890.
    [5]UBUKATA M, CHENG X C, UZAWA J, et al. Biosynthesis of the dialkylmaleic anhydride-containing antibiotics, tautomycin and tautomycetin [J]. J Chem Soc, Perkin Trans 1,1995,19):2399-404.
    [6]CHOI S S, HUR Y A, SHERMAN D H, et al. Isolation of the biosynthetic gene cluster for tautomycetin, a linear polyketide T cell-specific immunomodulator from Streptomyces sp. CK4412 [J]. Microbiology, 2007,153(4):1095-102.
    [7]LI W, LUO Y, JU J, et al. Characterization of the Tautomycetin Biosynthetic Gene Cluster from Streptomyces griseochromogenes Provides New Insight into Dialkylmaleic Anhydride Biosynthesis [J]. Journal of natural products,2009,72(3):450-9.
    [8]LUO Y, LI W, JU J, et al. Functional characterization of TtnD and TtnF, unveiling new insights into tautomycetin biosynthesis [J]. Journal of the American Chemical Society,2010,132(19):6663-71.
    [9]YANG D, LI W, HUANG S X, et al. Functional Characterization of ttnl Completing the Tailoring Steps for Tautomycetin Biosynthesis in Streptomyces griseochromogenes [J]. Organic Letters,2012, 14(5):1302-5.
    [10]MITSUHASHI S, MATSUURA N, UBUKATA M, et al. Tautomycetin is a novel and specific inhibitor of serine/threonine protein phosphatase type 1, PP1 [J]. Biochemical and biophysical research communications,2001,287(2):328-31.
    [11]SHIM J H, LEE H K, CHANG E J, et al. Immunosuppressive effects of tautomycetin in vivo and in vitro via T cell-specific apoptosis induction [J]. Proceedings of the National Academy of Sciences, 2002,99(16):10617.
    [12]WEE Y M, CHOI M Y, KANG C H, et al. The synergistic effect of tautomycetin on cyclosporine A-mediated immunosuppression in a rodent islet allograft model [J]. Molecular Medicine,2010, 16(7-8):298.
    [13]LIU S. YU Z, YU X, et al. SHP2 is a target of the immunosuppressant tautomycetin [J]. Chemistry & biology,2011,18(1):101-10.
    [14]LEE J H, LEE J S, KIM S E, et al. Tautomycetin inhibits growth of colorectal cancer cells through p21cip/WAF1 induction via the extracellular signal-regulated kinase pathway [J]. Molecular cancer therapeutics,2006,5(12):3222.
    [15]ADLER J T, COOK M, LUO Y. et al. Tautomycetin and tautomycin suppress the growth of medullary thyroid cancer cells via inhibition of glycogen synthase kinase-3β [J]. Molecular cancer therapeutics, 2009,8(4):914.
    [16]孙岩.变构菌素诱导乳腺癌细胞MCF-7凋亡机制的研究[D].大连:大连理工大学生命学院,2011.
    [17]BARFORD D. Molecular mechanisms of theprotein serine/threonine phosphatases [J]. Trends in biochemical sciences,1996,21(11):407-12.
    [18]MCCLUSKEY A, SIM A T R, SAKOFF J A. Serine-threonine protein phosphatase inhibitors: development of potential therapeutic strategies [J]. Journal of medicinal chemistry,2002,45(6): 1151-75.
    [19]SHENOLIKAR S, NAIRN A. Protein phosphatases:recent progress [J]. Advances in second messenger and phosphoprotein research,1991,23(1):121.
    [20]GOLDBERG J, HUANG H, KWON Y, et al. Three-dimensional structure of the catalytic subunit of protein serine/threonine phosphatase-1 [J]. Nature,2002,376:745-753.
    [21]ZHANG Z, ZHAO S, LONG F, et al. A mutant of protein phosphatase-1 that exhibits altered toxin sensitivity [J]. Journal of Biological Chemistry,1994,269(25):16997-7000.
    [22]MUNRO S, CEULEMANS H, BOLLEN M, et al. A novel glycogen-targeting subunit of protein phosphatase 1 that is regulated by insulin and shows differential tissue distribution in humans and rodents [J]. FEBS Journal,2005,272(6):1478-89.
    [23]BERNDT N, DOHADWALA M, LIU C W Y. Constitutively active protein phosphatase 1 [alpha] causes Rb-dependent G1 arrest in human cancer cells [J]. Current Biology,1997,7(6):375-86.
    [24]AYLL N V, MART NEZ-A C, GARC A A, et al. Protein phosphatase la is a Ras-activated Bad phosphatase that regulates interleukin-2 deprivation-induced apoptosis [J]. The EMBO journal,2000, 19(10):2237-46.
    [25]AMMOSOVA T, JEREBTSOVA M, BEULLENS M, et al. Nuclear targeting of protein phosphatase-1 by HIV-1 Tat protein [J]. Journal of Biological Chemistry,2005,280(43):36364.
    [26]SCHONTHAL A. Role of PP2A in intracellular signal transduction pathways [J]. Front Biosci,1998, 3:D1262-D73.
    [27]JANSSENS V, GORIS J. Protein phosphatase 2A:a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling [J]. Biochemical Journal,2001,353(3):417.
    [28]FELLNER T, LACKNER D H, HOMBAUER H, et al. A novel and essential mechanism determining specificity and activity of protein phosphatase 2A (PP2A) in vivo [J]. Genes & development,2003, 17(17):2138-50.
    [29]STONE S R, HOFSTEENGE J, HEMMINGS B A. Molecular cloning of cDNAs encoding two isoforms of the catalytic subunit of protein phosphatase 2A [J]. Biochemistry,1987,26(23):7215-20.
    [30]CHO U S, XU W. Crystal structure of a protein phosphatase 2A heterotrimeric holoenzyme [J]. Nature,2006,445(7123):53-7.
    [31]EICHHORN P J A, CREYGHTON M P, BERNARDS R. Protein phosphatase 2A regulatory subunits and cancer [J]. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer,2009,1795(1):1-15.
    [32]BECKER W, KENTRUP H, KLUMPP S, et al. Molecular cloning of a protein serine/threonine phosphatase containing a putative regulatory tetratricopeptide repeat domain [J]. Journal of Biological Chemistry,1994,269(36):22586-92.
    [33]CHEN M X, MCPARTLIN A E, BROWN L, et al. A novel human protein serine/threonine phosphatase, which possesses four tetratricopeptide repeat motifs and localizes to the nucleus [J]. The EMBO journal,1994,13(18):4278.
    [34]CHINKERS M. Targeting of a distinctive protein-serine phosphatase to the protein kinase-like domain of the atrial natriuretic peptide receptor [J]. Proceedings of the National Academy of Sciences,1994, 91(23):11075.
    [35]CHEN M X, COHEN P T W. Activation of protein phosphatase 5 by limited proteolysis or the binding of polyunsaturated fatty acids to the TPR domain [J]. FEBS letters,1997,400(1):136-40.
    [36]GOLDEN T, ARAGON 1 V, ZHOU G, et al. Constitutive over expression of serine/threonine protein phosphatase 5 (PP5) augments estrogen-dependent tumor growth in mice [J]. Cancer letters,2004, 215(1):95-100.
    [37]ZUO Z, DEAN N M, HONKANEN R E. Serine/threonine protein phosphatase type 5 acts upstream of p53 to regulate the induction of p21 WAF1/Cipl and mediate growth arrest [J]. Journal of Biological Chemistry,1998,273(20):12250-8.
    [38]GOLDEN T, SWINGLE M. HONKANEN R E. The role of serine/threonine protein phosphatase type 5 (PP5) in the regulation of stress-induced signaling networks and cancer [J]. Cancer and Metastasis Reviews,2008.27(2):169-78.
    [39]VAN DUUREN B. Tumor-promoting agents in two-stage carcinogenesis [J]. Progress in experimental tumor research,1969,11:31-68.
    [40]FUJIKI H, TANAKA Y, MIYAKE R, et al. Activation of calcium-activated, phospholipid-dependent protein kinase (protein kinase C) by new classes of tumor promoters:teleocidin and debromoaplysiatoxin [J]. Biochemical and biophysical research communications,1984,120(2):339.
    [41]SUGANUMA M, FUJIKI H, FURUYA-SUGURI H. et al. Calyculin A. an inhibitor of protein phosphatases, a potent tumor promoter on CD-1 mouse skin [J]. Cancer research,1990,50(12):3521.
    [42]FUJIKI H, SUGANUMA M, SUGURI H, et al. Induction of ornithine decarboxylase activity in mouse skin by a possible tumor promoter, okadaic acid [J]. Proceedings of the Japan Academy Ser B: Physical and Biological Sciences,1987,63(2):51-3.
    [43]SUGANUMA M, FUJIKI H, SUGURI H, et al. Okadaic acid:an additional non-phorbol-12-tetradecanoate-13-acetate-type tumor promoter [J]. Proceedings of the National Academy of Sciences,1988,85(6):1768.
    [44]LEWY D, SOENEN D, BOGER D. Fostriecin:chemistry and biology [J]. Current medicinal chemistry,2002,9(22):2005-32.
    [45]BUCK S B, HARDOUIN C. ICHIKAWA S, et al. Fundamental role of the fostriecin unsaturated lactone and implications for selective protein phosphatase inhibition [J]. Journal of the American Chemical Society,2003,125(51):15694-5.
    [46]STAMPWALA S, BUNGE R, HURLEY T, et al. Novel antitumor agents CI-920, PD 113,270 and PD 113,271. Ⅱ. Isolation and characterization [J]. The Journal of antibiotics,1983,36(12):1601.
    [47]DE JONG R, DE VRIES E, MULDER N. Fostriecin:a review of the preclinical data [J]. Anti-cancer drugs,1997,8(5):413.
    [48]ROBERGE M, TUDAN C, HUNG S M F, et al. Antitumor drug fostriecin inhibits the mitotic entry checkpoint and protein phosphatases 1 and 2A [J]. Cancer research,1994,54(23):6115.
    [49]WALL J G, BURRIS 3RD H, VON HOFF D D, et al. A phase I clinical and pharmacokinetic study of the topoisomerase 1 inhibitor topotecan (SK&F 104864) given as an intravenous bolus every 21 days [J]. Anti-cancer drugs,1992,3(4):337.
    [50]KALEV P, A SABLINA A. Protein phosphatase 2A as a potential target for anticancer therapy [J]. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry,2011,11(1): 38-46.
    [51]CARDOSO M C, LEONHARDT H, NADAL-GINARD B. Reversal of terminal differentiation and control of DNA replication:cyclin A and Cdk2 specifically localize at subnuclear sites of DNA replication [J]. Cell,1993,74(6):979-92.
    [52]DE FALCO M, DE LUCA A. Cell cycle as a target of antineoplastic drugs [J]. Current pharmaceutical design,2010,16(12):1417-26.
    [53]VERMEULEN K, VAN BOCKSTAELE D R, BERNEMAN Z N. The cell cycle:a review of regulation, deregulation and therapeutic targets in cancer [J]. Cell proliferation,2003,36(3):131-49.
    [54]SCHWARTZ G K, SHAH M A. Targeting the cell cycle:a new approach to cancer therapy [J]. Journal of clinical oncology,2005,23(36):9408-21.
    [55]SENDEROWICZ A M. Flavopiridol:the first cyclin-dependent kinase inhibitor in human clinical trials [J]. Investigational new drugs,1999,17(3):313-20.
    [56]LUKAS J, PARRY D, AAGAARD L, et al. Retinoblastoma-protein-dependent cell-cycle inhibition by the tumour suppressor p16 [J]. Nature,1995,375:503-6.
    [57]MALUMBRES M. Cyclins and related kinases in cancer cells [J]. Journal of BU ON:official journal of the Balkan Union of Oncology,2007,12:S45.
    [58]PAGANO M, DRAETTA G, JANSEN-DURR P. Association of cdk2 kinase with the transcription factor E2F during S phase [J]. Science,1992,255(5048):1144-7.
    [59]PAGANO M, PEPPERKOK R, VERDE F, et al. Cyclin A is required at two points in the human cell cycle [J]. The EMBO journal,1992,11(3):961.
    [60]GIRARD F, STRAUSFELD U, FERNANDEZ A, et al. Cyclin A is required for the onset of DNA replication in mammalian fibroblasts [J]. Cell,1991,67(6):1169.
    [61]PAGANO M, PEPPERKOK R, LUKAS J, et al. Regulation of the cell cycle by the cdk2 protein kinase in cultured human fibroblasts [J]. The Journal of cell biology,1993,121(1):101-11.
    [62]TOYOSHIMA H, HUNTER T. p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21 [J]. Cell,1994,78(1):67.
    [63]MOTOKURA T, BLOOM T, KIM H G, et al. A novel cyclin encoded by a bell-linked candidate oncogene [J]. Nature,1991,350(6318):512-5.
    [64]WEI P, GARBER M E, FANG S M. et al. A novel CDK9-associated C-type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA [J]. Cell,1998, 92(4):451-62.
    [65]SERRANO M, HANNON G J, BEACH D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK.4 [J]. Nature,1993.366(6456):704-7.
    [66]VAN DEN HEUVEL S, HARLOW E. Distinct roles for cyclin-dependent kinases in cell cycle control [J]. Science,1993,262(5142):2050-4.
    [67]MACLACHLAN T K. SANG N, GIORDANO A. Cyclins, cyclin-dependent kinases and cdk inhibitors:implications in cell cycle control and cancer [J]. Critical reviews in eukaryotic gene expression,1995,5(2):127.
    [68]MALUMBRES M, BARBACID M. Mammalian cyclin-dependent kinases [J]. Trends in biochemical sciences,2005,30(11):630-41.
    [69]GRA A X, REDDY E P. Cell cycle control in mammalian cells:role of cyclins, cyclin dependent kinases (CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors (CKIs) [J]. Oncogene,1995.11(2):211.
    [70]MORGAN D O. Principles of CDK regulation [J]. Nature,1995,374(6518):131.
    [71]JEFFREY P D, RUSSO A A, POLYAK K, et al. Mechanism of CDK activation revealed by the structure of a cyclinA-CDK2 complex [J]. Nature,1995,376(6538):313-20.
    [72]MEYERSON M, HARLOW E. Identification of G1 kinase activity for cdk6, a novel cyclin D partner [J]. Molecular and cellular biology,1994,14(3):2077-86.
    [73]KATO J, MATSUSHIME H, HIEBERT S W. et al. Direct binding of cyclin D to the retinoblastoma gene product (pRb) and pRb phosphorylation by the cyclin D-dependent kinase CDK4 [J]. Genes & development,1993,7(3):331.
    [74]UBERSAX J A, WOODBURY E L, QUANG P N, et al. Targets of the cyclin-dependent kinase Cdkl [J]. Nature,2003,425(6960):859-64.
    [75]DE BONDT H L, ROSENBLATT J, JANCARIK J, et al. Crystal structure of cyclin-dependent kinase 2 [J]. Nature,1993,363:595-602.
    [76]SHAPIRO G I. Cyclin-dependent kinase pathways as targets for cancer treatment [J]. Journal of clinical oncology,2006,24(11):1770-83.
    [77]KOFF A, GIORDANO A, DESAI D, et al. Formation and activation of a cyclin E-cdk2 complex during the G1 phase of the human cell cycle [J]. Science,1992,257(5077):1689-94.
    [78]GYURIS J, GOLEMIS E, CHERTKOV H, et al. Cdil, a human G1 and S phase protein phosphatase that associates with Cdk2 [J]. Cell,1993,75(4):791.
    [79]DE AZEVEDO W F, MUELLER-DIECKMANN H J. SCHULZE-GAHMEN U, et al. Structural basis for specificity and potency of a flavonoid inhibitor of human CDK2, a cell cycle kinase [J]. Proceedings of the National Academy of Sciences,1996,93(7):2735.
    [80]TIMOFEEV O, CIZMECIOGLU O, SETTELE F, et al. Cdc25 phosphatases are required for timely assembly of CDK1-cyclin B at the G2/M transition [J]. Journal of Biological Chemistry,2010, 285(22):16978.
    [81]OH J S, HAN S J, CONT1 M. Wee1B, Myt1, and Cdc25 function in distinct compartments of the mouse oocyte to control meiotic resumption [J]. The Journal of cell biology,2010,188(2):199-207.
    [82]CONTOUR-GALCERA M O, SIDHU A, PR VOST G, et al. What's new on CDC25 phosphatase inhibitors [J]. Pharmacology & therapeutics,2007,115(1):1-12.
    [83]MART N A, ODAJIMA J, HUNT S L, et al. Cdk.2 is dispensable for cell cycle inhibition and tumor suppression mediated by p27 Kip1 and p21 Cipl [J]. Cancer Cell,2005,7(6):591-8.
    [84]NAKAYAMA K, ISHIDA N, SHIRANE M, et al. Mice Lacking p27 Kip1 Display Increased Body Size, Multiple Organ Hyperplasia, Retinal Dysplasia, and Pituitary Tumors [J]. Cell,1996,85(5): 707-20.
    [85]KIYOKA WA H, KINEMAN R D, MANOVA-TODOROVA K O, et al. Enhanced Growth of Mice Lacking the Cyclin-Dependent Kinase Inhibitor Function of p27 Kip1 [J]. Cell,1996,85(5):721-32.
    [86]WADE HARPER J. ELLEDGE S J. Cdk inhibitors in development and cancer [J]. Current opinion in genetics & development,1996,6(1):56-64.
    [87]SHERR C J, ROBERTS J M. CDK inhibitors:positive and negative regulators of Gl-phase progression [J]. Genes & development,1999,13(12):1501.
    [88]LI F, AMBROSINI G, CHU E Y, et al. Control of apoptosis and mitotic spindle checkpoint by survivin [J]. Nature,1998,396(6711):580-3.
    [89]KOPS G J P L, KIM Y, WEAVER B A A, et al. ZW10 links mitotic checkpoint signaling to the structural kinetochore [J]. The Journal of cell biology,2005,169(1):49-60.
    [90]DE CARCER G, PEREZ DE CASTRO I, MALUMBRES M. Targeting cell cycle kinases for cancer therapy [J]. Current medicinal chemistry,2007,14(9):969-85.
    [91]MALUMBRES M, BARBACID M. Is Cyclin D1-CDK4 kinase a bona fide cancer target? [J]. Cancer Cell,2006,9(1):2-4.
    [92]LAPENNA S, GIORDANO A. Cell cycle kinases as therapeutic targets for cancer [J]. Nature Reviews Drug Discovery,2009,8(7):547-66.
    [93]VASSILEV L T, TOVAR C, CHEN S, et al. Selective small-molecule inhibitor reveals critical mitotic functions of human CDK1 [J]. Proceedings of the National Academy of Sciences,2006,103(28): 10660-5.
    [94]KERRJFR W A H, CURRIE A. Apoptosis:a basic biological phenomenon with wide-ranging implications in tissue kinetics [J]. Br J Cancer,1972,26:239-57.
    [95]ORRENIUS S. Mitochondrial regulation of apoptotic cell death [J]. Toxicology letters,2004,149(1): 19-23.
    [96]MEYN R E, ROBERTSON L. Implications of apoptotic cell death regulation in cancer therapy [J]. Seminars in Cancer Biology,1995,6(1):53-60.
    [97]FADEEL B, ORRENIUS S. Apoptosis:a basic biological phenomenon with wide-ranging implications in human disease [J]. Journal of internal medicine,2005,258(6):479-517.
    [98]POP C, SALVESEN G S. Human caspases:activation, specificity, and regulation [J]. Journal of Biological Chemistry,2009,284(33):21777-81.
    [99]STENNICKE H R, RENATUS M, MELDAL M, et al. Internally quenched fluorescent peptide substrates disclose the subsite preferences of human caspases 1,3,6,7 and 8 [J]. Biochemical Journal, 2000,350(2):563.
    [100]EARNSHAW W C, MARTINS L M, KAUFMANN S H. Mammalian caspases:structure, activation, substrates, and functions during apoptosis [J]. Annual review of biochemistry,1999,68(1):383-424.
    [101]CREAGH E M, CONROY H, MARTIN S J. Caspase-activation pathways in apoptosis and immunity [J]. Immunological reviews,2003,193(1):10-21.
    [102]SCHNEIDER P, TSCHOPP J. Apoptosis induced by death receptors [J]. Pharmacochemistry Library, 2000,31:281-6.
    [103]PEREIRA W, AMARANTE-MENDES G. Apoptosis:A Programme of Cell Death or Cell Disposal? [J]. Scandinavian Journal of Immunology,2011,73(5):401-7.
    [104]SHERIDAN J P, MARSTERS S A. PITTI R M, et al. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors [J]. Science,1997,277(5327):818-21.
    [105]LI H, ZHU H, XU C. et al. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis [J]. Cell.1998.94(4):491-501.
    [106]GROSS A, MCDONNELL J M. KORSMEYER S J. BCL-2 family members and the mitochondria in apoptosis [J]. Genes & development,1999,13(15):1899-911.
    [107]YOSHIDA H, KONG Y Y, YOSHIDA R, et al. Apafl is required for mitochondrial pathways of apoptosis and brain development [J]. Cell,1998,94(6):739.
    [108]RAFFO A J, PERLMAN H, CHEN M W, et al. Overexpression of bcl-2 protects prostate cancer cells from apoptosis in vitro and confers resistance to androgen depletion in vivo [J]. Cancel research, 1995,55(19):4438.
    [109]WEI M C, ZONG W X, CHENG E H Y, et al. Proapoptotic BAX and BAK:a requisite gateway to mitochondrial dysfunction and death [J]. Science's STKE,2001.292(5517):727.
    [110]LUO X, BUDIHARDJO I, ZOU H, et al. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors [J]. Cell,1998, 94(4):481-90.
    [111]RAMEH L E, CANTLEY L C. The role of phosphoinositide 3-kinase lipid products in cell function [J]. Journal of Biological Chemistry,1999,274(13):8347-50.
    [112]HILES 1 D. OTSU M, VOLINIA S, et al. Phosphatidylinositol 3-kinase:structure and expression of the 110 kd catalytic subunit [J]. Cell,1992,70(3):419-29.
    [113]CANTLEY L C. The phosphoinositide 3-kinase pathway [J]. Science's STKE,2002,296(5573): 1655.
    [114]ALESSI D R, ANDJELKOVIC M, CAUDWELL B, et al. Mechanism of activation of protein kinase B by insulin and IGF-1 [J]. The EMBO journal.1996,15(23):6541.
    [115]LYNCH D K, ELLIS C A. EDWARDS P, et al. Integrin-linked kinase regulates phosphorylation of serine 473 of protein kinase B by an indirect mechanism [J]. Oncogene,1999,18(56):8024.
    [116]HENNESSY B T, SMITH D L, RAM P T, et al. Exploiting the PI3K/Akt pathway for cancer drug discovery [J]. Nature Reviews Drug Discovery,2005,4(12):988-1004.
    [117]LUO J, MANNING B D, CANTLEY L C. Targeting the PI3K-Akt pathway in human cancer: rationale and promise [J]. Cancer Cell,2003,4(4):257-62.
    [118]WANG Q, WANG X, HERNANDEZ A, et al. Regulation of TRAIL expression by the phosphatidylinositol 3-kinase/Akt/GSK-3 pathway in human colon cancer cells [J]. Journal of Biological Chemistry,2002,277(39):36602.
    [119]SEGER R, KREBS E. The MAPK signaling cascade [J]. The FASEB Journal,1995,9(9):726-35.
    [120]JOHNSON G L. LAPADAT R. Mitogen-activated protein kinase pathways mediated by ERK,INK, and p38 protein kinases [J]. Science's STKE,2002,298(5600):1911.
    [121]LIU Z, HSU H, GOEDDEL D V, et al. Dissection of TNF receptor 1 effector functions:JNK activation is not linked to apoptosis while NF-kappaB activation prevents cell death [J]. Cell,1996, 87(3):565.
    [122]KUMAR S, BOEHM J, LEE J C. p38 MAP kinases:key signalling molecules as therapeutic targets for inflammatory diseases [J]. Nature Reviews Drug Discovery,2003,2(9):717-26.
    [123]POLAKIS P. Wnt signaling and cancer [J]. Genes & development,2000,14(15):1837-51.
    [124]NOORT M, CLEVERS J. TCF transcription factors, mediators of Wnt-signaling in development and cancer [J]. Dev Biol,2002,244:1-8.
    [125]NELSON W J, NUSSE R. Convergence of Wnt, β-catenin, and cadherin pathways [J]. Science,2004, 303(5663):1483-7.
    [126]KUBBUTAT M H G, JONES S N, VOUSDEN K H. Regulation of p53 stability by Mdm2 [J]. Nature,1997,387(6630):299-303.
    [127]LEVINE A J, MOMAND J, FINLAY C A. The p53 tumour suppressor gene [J]. Nature,1991, 351(6326):453.
    [128]刘波.Tautomycetin新型衍生物的分离鉴定及其生物合成机制的初步探讨[D].大连:大连理工大学生命学院,2011.
    [129]MATSUZAWA S, KAWAMURA T, MITSUHASHI S, et al. Thyrsiferyl 23-acetate and its derivatives induce apoptosis in various T-and B-leukemia cells [J]. Bioorganic & medicinal chemistry,1999,7(2):381-7.
    [130]NAKAI E, PARK K, YAWATA T, et al. Enhanced MDR1 expression and chemoresistance of cancer stem cells derived from glioblastoma [J]. Cancer investigation,2009,27(9):901-8.
    [131]ABAAN O D, MUTLU P K, BARAN Y, et al. Multidrug resistance mediated by MRP1 gene overexpression in breast cancer patients [J]. Cancer investigation,2009,27(2):201-5.
    [132]KNUEFERMANN C, LU Y, LIU B, et al. HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells [J]. Oncogene,2003,22(21):3205-12.
    [133]Malabika S, Carmen RR, Gema R, et al. Stimulation of the mitogen-activated protein kinase pathway antagonizes TRAIL-induced apoptosis downstream of BID cleavage in human breast cancer MCF-7 cells [J]. Oncogene,2002,21(7):4323-4327.
    [134]LIANG Y, YAN C, SCHOR N F. Apoptosis in the absence of caspase 3 [J]. Oncogene,2001,20(45): 6570.
    [135]CUENDA A, NEBREDA A R. p38 [delta] and PKD1:Kinase Switches for Insulin Secretion [J]. Cell, 2009,136(2):209-10.
    [136]TOTROV M, ABAGYAN R. Flexible ligand docking to multiple receptor conformations:a practical alternative [J]. Current opinion in structural biology,2008,18(2):178-84.
    [137]SARKER M, RUIZ-RUIZ C, ROBLEDO G, et al. Stimulation of the mitogen-activated protein kinase pathway antagonizes TRAIL-induced apoptosis downstream of BID cleavage in human breast cancer MCF-7 cells [J]. Oncogene,2002,21(27):4323-7.
    [138]FRIDMAN J S, LOWE S W. Control of apoptosis by p53 [J]. Oncogene,2003,22(56):9030-40.
    [139]SAKAMAKI J, DAITOKU H, UENO K, et al. Arginine methylation of BCL-2 antagonist of cell death (Bad) counteracts its phosphorylation and inactivation by Akt [J]. Proceedings of the National Academy of Sciences,2011,108(15):6085.
    [140]ZHANG C, CAI T, ZHU H, et al. Synergistic antitumor activity of gemcitabine and ABT-737 in vitro and in vivo through disrupting the interaction of USP9X and Mcl-1 [J]. Molecular cancer therapeutics,2011,10(7):1264.
    [141]FUKUDA M, ASANO S. NAKAMURA T, et al. CRM1 is responsible for intracellular transport mediated by the nuclear export signal [J]. Nature,1997,390(6657):308-11.
    [142]FORNEROD M, OHNO M, YOSHIDA M, et al. CRM1 is an export receptor for leucine-rich nuclear export signals [J]. Cell,1997,90(6):1051-60.
    [143]KUDO N, WOLFF B, SEKIMOTO T, et al. Leptomycin B inhibition of signal-mediated nuclear export by direct binding to CRM1 [J]. Experimental cell research,1998,242(2):540-7.
    [144]SANTI D V, ZHOU Y. Combination therapies using leptomycin B [M]. Google Patents.2005.
    [145]VIGNERI P, WANG J Y J. Induction of apoptosis in chronic myelogenous leukemia cells through nuclear entrapment of BCR-ABL tyrosine kinase [J]. Nature medicine,2001,7(2):228-34.

© 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