塞来昔布对H22肝癌和Lewis肺癌抑制作用及其机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景:肿瘤流行病学资料显示,肝癌和肺癌是威胁人类健康与生命的恶性疾病,其发病率和死亡率极高。目前普遍采用的放化疗以及手术切除治疗,均存在敏感性较差,不能根治的缺点。因此,寻找有效的治疗方法是亟待解决的问题。塞来昔布(celecoxib)属于新一代非甾体类抗炎药物(nonsteroidal anti-inflammatory drugs,NSAIDs),大量的研究结果表明,塞来昔布具有潜在的抑制肿瘤发生、发展以及转移等作用。塞来昔布为环氧合酶-2(cyclooxygenase-2,COX-2)选择性抑制剂,通过抑制COX-2表达、降低前列腺素E2(prostaglandin E2,PGE2)水平促进肿瘤细胞凋亡,而近年来人们还发现了一些COX-2非依赖的靶点,但是其抗肿瘤作用机制目前尚未完全阐明。
     研究目的:阐述塞来昔布对H22肝癌和Lewis肺癌的治疗作用及其作用机制,为其作为抗肿瘤药物的临床应用提供理论依据。
     研究内容:建立小鼠实体瘤模型后灌胃给药,计算肿瘤体积和重量,检测在体水平对肿瘤的抑制;MTT方法检测对肿瘤细胞增殖抑制作用;流式细胞仪手段检测肿瘤细胞凋亡和线粒体膜电位变化;Western blot方法检测细胞花生四烯酸(arachidonic acid,AA)代谢通路关键酶磷脂酶A2(cytosolic phospholipase A2,cPLA2)和COX-2,线粒体凋亡通路关键蛋白Bcl-2相关X蛋白(Bcl-2 Associated X protein,Bax)、B细胞淋巴瘤/白血病-2(B cell lymphoma/lewkmia-2,Bcl-2)、凋亡诱导因子(apoptosis-inducing factor,AIF)、细胞色素c (cytochrome c,Cyto c)、半胱氨酸天冬氨酸酶-3(cysteine-aspartic proteases,caspase-3)、半胱氨酸天冬氨酸酶-9(cysteine-aspartic proteases,caspase-9),过氧化物酶体增殖物激活受体( peroxisome proliferator activated receptor,PPARγ),核因子-κB( nuclear factor kappa B,NF-κB)的表达;RT-PCR方法检测细胞中cPLA2和COX-2的mRNA转录情况,采用高效液相色谱(high performance liquid chromatography,HPLC)法检测24h细胞培养上清液AA水平的变化,酶联免疫吸附试验(enzyme-linked immunosorbent assay,ELISA)方法检测24h细胞培养上清液PGE2水平的变化。
     研究结果:
     1、塞来昔布对H22肝癌抑制作用
     塞来昔布抑制H22肝癌细胞增殖及其移植瘤生长;高剂量塞来昔布抑制cPLA2和COX-2表达,增加AA和降低PEG2水平;低剂量塞来昔布增加cPLA2表达,对COX-2表达无影响,AA含量增高、对PEG2水平无影响;各剂量组塞来昔布均增加AA与PGE2比值;塞来昔布作用于线粒体凋亡途径,破坏线粒体膜,影响凋亡信号蛋白表达,促进肿瘤细胞凋亡;塞来昔布对PPARγ/NF-κB途径无影响。
     2、塞来昔布对Lewis肺癌抑制作用
     塞来昔布抑制Lewis肺癌移植瘤生长及细胞增殖;能够促进细胞凋亡、降低线粒体膜电位;塞来昔布抑制COX-2表达,增加cPLA2表达,提高AA含量,降低PEG2水平;各剂量组塞来昔布均增加AA与PGE2比值;塞来昔布作用于PPARγ/NF-κB途径,增加PPARγ表达,降低NF-κB表达。
     研究结论:
     塞来昔布对H22肝癌细胞及Lewis肺癌细胞花生四稀酸代谢途径关键酶及其产物调节不同,但均增加AA与PGE2比值;AA与PGE2比值可能反映非甾体抗炎药抗肿瘤作用;塞来昔布可能通过调节COX-2依赖及非依赖途径、激活线粒体凋亡通路,促进细胞凋亡,从而发挥抗H22肝癌作用;塞来昔布抗Lewis肺癌可能通过调节COX-2依赖途径及PPARγ/NF-κB途径,促进细胞凋亡实现。
Liver and lung cancer are malignant diseases that threaten health and life of human, and their incidence and mortality keep very high nowadays, according to epidemiology data from cancer. Though many kinds of chemotherapies and surgical resections are used in clinical, there is no sensitive and efficiency method to overcome the problems. Therefore, it is very urgent to find some appropriate ways for the patients.
     Recent studies have found that, cancer development had close relationship with disorders of cPLA2 and COX-2, the two key enzymes in the AA metabolic pathway. The membrane phospholipids are catalyzed into AA by cPLA2, and then AA is converted into PGE2, which has the activities of inhibiting apoptosis, stimulating division and angiogenesis, promoting invasion and metastasis on tumor cells. It was reported that COX-2 and cPLA2 were over-expressed in cancers, such as colorectal cancer, liver cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer etc. These findings suggested that COX-2 and cPLA2 played a very important part in the development and differentiation of liver cancer and lung cancer.
     In addition, PPARγ/NF-kB pathway also had very close relationship with tumorigenesis and development. The PPARs are some clasess of nuclear transcription factors, and activated by their ligands. After being activated, the PPARs will bind to the specific sequences of DNA in heterodimer forms and regulate the transcription of the NF-κB. NF-κB activation can inhibit cell apoptosis and promote cell proliferation. The expressions of PPARγand NF-kB in tumor tissue were higher than those in adjacent tissues, depending on tumor histological type, cell differentiation and postoperative TNM staging. The agonists of PPARγpioglitazone and blocking NF-kB could inhibit tumor proliferation and promote apoptosis of tumor cells. Pathw.
     The mitochondria is an important place for apoptosis. With the stimulations of apoptosis signal external or internal in tumor cells, the mitochondrial membrane permeability was changed, and then cytochrome c, AIF and other apoptotic factors were released into the cytoplasm. The cytochrome c would lead to cell apoptosis after forming apoptosome with the Apaf-1 and caspase-9, activating caspase-3, caspase-6 or caspase-7 in the downstream. The Bcl-2 family proteins, including anti-apoptotic protein Bcl-2 and pro-apoptotic protein Bax, are some important factors that regulated the changes of the mitochondrial membrane permeability, and play a very important role in the mitochondrial apoptosis pathway.
     Celecoxib belongs to the new generation of non-steroidal anti-inflammatory drugs, and a large number of studies declare the potential inhibition effects of celecoxib on occurrence, development and transfer of tumor. Celecoxib is a kind of selective inhibitors of COX-2. It promoted tumor cell apoptosis by inhibiting the expression of COX-2 and decreasing the level of PGE2 in traditional theory. But, in recent years, some COX-2 independent pathways were also found a number of studies. Those showed that its anti-tumor mechanisms were not yet fully understood. In the present study, we attempted to elucidate the inhibition effects of celecoxib on the H22 hepatoma and Lewis lung cancer and their mechanisms by COX-2 dependent and -independent Pathway in vivo and in vitro. Moreover, we tried to explain the preventive and therapeutic effect of celecoxib on cancer through its regulations on the AA metabolic pathway, the PPARγ/NF-kB pathway and the mitochondrial apoptosis pathway. These findings will provide some theoretical basis to its clinical application as antitumor drugs in the future.
     1 Inhibition Effects of Celecoxib on H22 Hepatoma and its Mechanism
     Forty BALB/c mice receiving tumor implantation were divided randomly into control group and celecoxib groups at low, middle, high dosage (100、200、400mg/kg). All the groups were gavaged continuously with normal saline or celecoxib on the third day after implantation. The mice were killed on the 15th day, and the volume and weight of the tumor tissues were calculated. The protein expressions of cPLA2 and COX-2 were examined in the tumor tissues by Western blot analysis. The proliferation of H22 Hepatoma cells, treated with different doses (0, 25, 50, 100, 200μM) of celecoxib for 12, 24, 48 and 72 h, was measured by MTT assay. After being treated with celecoxib of different doses (0, 50, 100, 200 and 400μM) for 24h, the H22 cells were measured as followed: The apoptosis ratio and the mitochondrial membrane potential were detected by flow cytometry; The protein expressions of cPLA2, COX-2, Bax, Bcl-2, AIF, cytochrome c, caspase-3 and caspase-9 were examined by Western blot analysis, and the mRNA levels of cPLA2 and COX-2; The concentrations of arachidonic acid and prostaglandin E2 in the culture supernatants for 24h were measured by the methods of RP-HPLC and PGE2-specific ELISA respectively.
     The results in vivo showed that: The solid tumor volume and weight of treatment groups were lower than those of control group, and there was significant difference among the groups (P<0.05). The tumor inhibitory ratios of treatment groups were more than 30%, especially 75% in high dose group. The protein expressions of cPLA2 and COX-2 in treatment groups were lower than control group in a dose-dependent manner.
     The results in vitro showed that: Celecoxib exposure significantly reduced the viability of H22 cells in a dose- and time-dependent manner. IC50 values of celecoxib were 134.7±0.5μM, 112.4±0.3μM, 78.0±0.6μM and 56.9±0.8μM at 12, 24, 48, and 72h; Compared with controlgroup, the apoptosis ratio of tumor cell in treated groups notably raised and mitochondrial membrane potential decreased; The mRNA and protein expression of COX-2 were not altered in H22 cells treated with celecoxib at 50 and 100μM, however the COX-2 mRNA and protein levels were significantly decreased in H22 cells at 200 and 400μM dosage of celecoxib when compared to untreated H22 cells. Exposure to celecoxib caused an enhancement of the mRNA and protein levels of cPLA2 at dosages of 50 and 100μM, but resulted in an inhibition on the expression of cPLA2 at dosages of 200 and 400μM; The concentration of AA was elevated in the H22 cells after incubation with celecoxib at a dose of 50-400μM for 24h, and AA concentration was the highest at 100μM celecoxib, and the PGE2 production in H22 cells was not significantly influenced by treatment with 50 and 100μM celecoxib, but he levels of PGE2 production were significantly decreased when the cells were treated with 200 and 400μM celecoxib; The ratio of AA to PGE2 concentration was increased by celecoxib treatment in a dose-dependent manner in H22 cells; Compared with control group, Bax、caspase-3、caspase-9 were up-regulated and Bcl-2 was down-regulated; The protein levels of cytochrome c and AIF were up-regulated in the cytoplasmic, and down-regulated in the mitochondrias of the cells.
     2 Inhibition Effects of Celecoxib on Lewis lung cancer and its Mechanism
     Twenty C57BL/6 mice receiving tumor implantation were divided randomly into control group and treatment (200 mg/kg) groups. All the groups were gavaged continuously with normal saline or celecoxib on the third day after implantation. The mice were killed on the 15th day, and the volume and weight of the tumor tissues were calculated. The proliferation of Lewis lung carcinoma cells, treated with different doses (50, 100, 200μM) of celecoxib for 12, 24, 48 and 72 h, was measured by MTT assay. The apoptosis and mitochondrial membrane potential were detected by flow cytometry. The protein expression of cPLA2, COX-2, PPARγand NF-κB was examined using Western blot analysis. The concentrations of AA and PGE2 in culture supernatants were measured by the methods of RP-HPLC and PGE2-specific ELISA respectively.
     The results in vivo showed that: The solid tumor volume and weight of treatment groups were lower than those of control group, and there was significant difference among the groups (P<0.05). The tumor inhibitory ratio of treatment groups was 50%.
     The results in vitro showed that: The proliferation of Lewis lung carcinoma cells was inhibited by celecoxib in a dose and time-dependent manner. The IC50 value of 72 h was 134.06±2.97μM; the apoptosis ratio of tumor cell in treated groups notably raised; The expressions of cPLA2 and PPARγwere up-regulated, but COX-2 and NF-κB were down-regulated in the Lewis cells exposed to celecoxib. The amount of AA was increased and PGE2 was decreased in the culture supernatant respectively. The ratio of AA to PGE2 was increased in a dose-dependent manner.
     In summary, our findings suggest that celecoxib could adjust the productions of AA and PGE2 by regulating on cPLA2 and COX-2, the two key enzymes in AA metabolic pathway. Although the regulations were different in H22 hepatoma and Lewis lung cancer cells, the ratio of AA and PGE2 was increased in both cell lines treated by celecoxib. The ratio of AA and PGE2 maybe was the a key factor in tumor development and reflecting the anti-tumor effect of celecoxib and other NSAIDs. Celecoxib could significantly inhibit and Lewis lung cancer in vivo and in vitro. Celecoxib may inhibited the H22 hepatoma by COX-2 dependent, non-dependent pathway, and activating mitochondrial apoptosis pathway. Celecoxib may inhibited the Lewis lung cancer by COX-2 dependent pathway, and PPARγ/ NF-κB channels.
引文
[1] Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; 100 (1):57-70.
    [2] Kuper H, Adami HO, Trichopoulos D. Infections as a major preventable cause of human cancer. J Intern Med. 2000; 248(3):171-83.
    [3] Sameni M, Dosescu J, Moin K, et al. Functional imaging of proteolysis: stromal and inflammatory cells increase tumor proteolysis. Mol Imaging. 2003; 2(3):159-75.
    [4]李菁,朱元民,刘玉兰.炎症与肿瘤关系研究进展.中国医药导刊, 2007; 9(3): 217-219.
    [5] Daniel D, Meyer-Morse N, Bergsland EK, et al. Immune enhancement of skin carcinogenesis by CD4+ T cells. J Exp Med. 2003; 197(8):1017-28.
    [6] Boland CR, Luciani MG, Gasche C, et al. Infection, inflammation, and gastrointestinal cancer. Gut. 2005; 54(9):1321-31.
    [7] Balsinde J, Balboa MA. Cellular regulation and pro-posed biological functions of group VIA calcium-independent phospholipase A2 in activated cells. Cell Signal 2005; 17(9): 1052-62.
    [8] Wang XH, Yan GT, Wang LH, et al. The mediating role of cPLA2 in IL-1 beta and IL-6 release in LPS-induced HeLa cells. Cell Bio-chem Funct. 2004; 22(1): 41-4.
    [9] Shmelzer Z, Haddad N, Admon E, et al. Unique targeting of cytosolic phospholipase A2 to plasma membranes mediated by the NADPH oxidase in phagocytes. J Cell Biol. 2003; 162(4): 683-92.
    [10] Wang S, Zhou W, Wei M, et al. Effects of lead on NO, NOS, SOD, MDA in rat cerebral cortex. Wei Sheng Yan Jiu. 2002; 31(4): 226-8.
    [11] Graff JR, Konieek BW, Deddens JA, et al. Expression of groupⅡa secretory phospholipaseA2 inereases with prostate tumor grade. Clin Cancer Res. 2001; 7(12): 3857-61.
    [12] Panel V, Bo?lle PY, Ayala-Sanmartin J, et al. Cytoplasmic phospholipase A2 expression in human colon adenocarcinoma is correlated with cyclooxygenase-2 expression and contributes to prostaglandin E2 production. Cancer Lett. 2006; 243(2): 255-63.
    [13] Soydan AS, Tavares IA, Weech PK, et al. High molecular weight phospholipase A2 andfatty acids in human colon tumours and associated normaltissue. Eur J Cancer. 1996; (10): 1781-7.
    [14] Denizot Y, Chianea T, Labrousse F, et al. Platelet-activating factor and human thyroidcancer. Eur J Endocrinol. 2005; 153(1): 31-40.
    [15] Dong Q, Patel M, Scott KF, et al. Oncogenic action of phospholipase A2 in prostate cancer. Cancer Lett. 2006; 240(1): 9-16.
    [16] Laye JP, Gill JH. Phospholipase A2 expression in tumours: a target for therapeutic intervention? Drug Discov Today. 2003; 8(15): 710-6.
    [17] Williams CS, Mann M, Dubois RN. The role of cyclooxygenases in inflammation, cancer, and development. Oncogene. 1999; 18(55): 7908-16.
    [18] Fosslien E. Molecular pathology of cyclooxygenase-2 in neoplasia. Ann Clin Lab Sci. 2000; 30(1): 3-21.
    [19] Nzeako UC, Guicciardi ME, Yoon JH, et al. COX-2 inhibits Fas-mediated apoptosis in cholangiocarcinoma cells. Hepatology. 2002; 35(3): 552-9.
    [20] Li G, Yang T, Yan J. Cyclooxygenase-2 increased the angiogenic and metastatic potential of tumor cells. Biochem Biophys Res Commun. 2002; 299(5): 886-90.
    [21] Leahy KM, Koki AT, Masferrer JL. Role of cyclooxygenases in angiogenesis. Curr Med Chem. 2000; 7(11):1163-70.
    [22] Manchana T,Triratnachat S,Sirisabya N,et al. Prevalence and prognostic significance of COX-2 expression in stage IB cervical cancer.Gynecol Oncol. 2006; 100(3): 556-60.
    [23] Kim JY, Lim SJ, Park K, et al. Cyclooxygenase-2 and cerbB-2 expression in uterine cervical neoplasm assessed using tissue mi-croarrays.Gynecol Oncol. 2005; 97(2): 337-41.
    [24]唐丽霞,王敏,马坚伟.环氧合酶-2蛋白表达与卵巢癌生物学行为的相关性.中华肿瘤杂志. 2003; 25(3): 261-4.
    [25]刘锐,李明众. COX-2和Fas在宫颈癌中的表达及其临床意义.现代肿瘤医学. 2008; 16(2): 271-3.
    [26]李晓艳,王泽华.环氧合酶-2在卵巢癌组织中的表达.实用医学杂志. 2005; 21(20): 2254-5.
    [27] Yoshimatsu K, Altorki NK, Golijanin D, et al. Inducible prostaglandin E synthase is overexpressed in non2small cell lung cancer. Clin Cancer Res. 2001; 7(9): 2669-74.
    [28] Cheng J, Imanishi H, Iijima H, et al. Expression of cyclooxygenase2 and cytosoli phospholipase A2 in the liver tissue of patients with chronic hepatitis and liver cirrhosis. Hepatol. Res. 2002; 23(3):185-95.
    [29] Cheng ASL, Chan HLY, Leung NWY, et al. Expression of cyclooxygenase2 in chronic hepatitis B and the effects of anti-viraltherapy. Aliment Pharmacol Ther. 2002; 16(2): 251-60.
    [30] Dannenberg AJ, Altorki NK, Boyle JK, et al. Cyclooxygenase2: a pharmacological target for prevention of cancer. Lancet Onco1. 2001; 2(9): 544-51.
    [31] Rao CV, Reddy BS. NSAIDs and chemoprevention. Curr Cancer Drug Targets. 2004; 4(1): 29-42.
    [32] Evans JF, Kargman SL. Cancer and cyclooxygenase-2 (COX-2) inhibition. Curr Pharm Des. 2004; 10(6): 627-34.
    [33] Willson TM, Brown PJ, Sternbach DD, et al. The PPARs: from orphan receptors to drug discovery. J Med Chem. 2000; 43(4):527-50.
    [34]张敏,邹萍,白明,等.活化的过氧化物酶体增生物激活受体-γ诱导人类肺癌细胞凋亡及其机制的研究.中华医学杂志, 2003; 83(13): 1169-72.
    [35]李燕,陈晓光. PPARγ配体的抗肿瘤作用.哈尔滨商业大学学报(自然科学版). 2005; 21(3): 267-9.
    [36] Lovekamp-Swan T, Chaffin CL. The peroxisome proliferator-activated receptor gamma ligand troglitazone induces apoptosis and p53 in rat granulosa cells. Mol Cell Endocrinol. 2005; 233(1-2): 15-24.
    [37]潘华锋,陈孝平. Ciglitazone抑制人肝癌HepG2细胞增殖活性的实验研究.中华实验外科杂志. 2004; 21(8): 934-36.
    [38] Nagamine M, Okumura T, Tanno S, et al. PPARγligand-in-duced apoptosis through a p53-dependent mechanism in human gastric cancer cells. Cancer Sci. 2003; 94(4): 338-43.
    [39] Piva R, Gianferretti P, Ciucci A, et al.15-Deoxy-{Delta}12,14-prostaglandin J2 induces apoptosis in human malignant B cells: an effect associated with inhibition of NF-kappa B activ-ity and down-regulation of antiapoptotic proteins. Blood. 2005; 105(4): 1750-8.
    [40] Ceni E, Mello T, Tarocchi M, et al. Antidiabetic thiazolidinedi-ones induce ductaldifferentiation but not apoptosis in pancre-atic cancer cells.World J Gastroenterol. 2005; 11(8): 1122-30.
    [41] Margeli A, Kouraklis G, Theocharis S. Peroxisome proliferatoractivated receptor-c (PPAR-c) ligands and angiogenesis. Angiogenesis. 2003; 6(3): 165-9.
    [42] Ferruzzi P, Ceni E, Tarocchi M, et al. Thiazolidinediones inhibit growth and invasiveness of the human adrenocortical cancercell line H295R. J Clin Endocrinol Metab. 2005; 90(3): 1332-9.
    [43] Su CG, Wen X, Bailey ST, et al. A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response. J Clin Invest. 1999; 104(4): 383-9.
    [44] Konstantinopoulos PA, Vandoros GP, Sotiropoulou-Bonikou G, et al. NF-kappaB/PPAR gamma and/or AP-1/PPAR gamma 'on/off' switches and induction of CBP in colon adenocarcinomas: correlation with COX-2 expression. Int J Colorectal Dis. 2007; 22(1): 57-68.
    [45] Sen R, Baltimore D. Multiple nuclear factors interact with theimmunoglobulin enhancer sequences.Cell. 1986; 46(5): 705-16.
    [46] Ghosh S, Karin M. Missing pieces in the NF-kappaB puzzle. Cell. 2002; 109 Suppl: S81-96.
    [47] Beg AA, Sha WC, Bronson RT, et al. Embryoniclethality and liver degeneration in mice lacking the RelAcomponent of NF-kappa B. Nature. 1995; 376: 167-70.
    [48] Waddick KG, Uckun FM. Innovative treatment programsagainst cancer:II.Nuclear factor-kappaB(NF-kappaB) as a molecular target. Biochem Pharmacol. 1999; 57(1): 9-17.
    [49] Hiscott J, Kwon H, Genin P. Hostile takeovers: viral appropriation of the NF-kappaB pathway. J Clin Invest. 2001; 107:143-51.
    [50] Lwin T, Hazlehurst LA, Li Z, et al. Bone marrow stromal cells prevent apoptosis of lymphoma cells by upregulation of anti-apoptotic proteins associated with activation of NF-kappaB(RelB/p52)in non-Hodgkin\s lymphoma cells. Leukemia. 2007; 21: 1521-31.
    [51] Sweeney CJ, Mehrotra S, Sadaria MR, et al. The sesquiterpene lactone parthenolide in combination with docetaxel reduces metastasis and improves survival in a xenograft model of breast cancer. Mol Cancer Ther. 2005; 4(6): 1004-12.
    [52] Halestrap A. Mitochondria and cell death. The Biochemist, 2000; 21(2): 19-24.
    [53]尹立国,崔建忠.线粒体通透性转换孔的研究现状.华北煤炭医学院学报. 2007; 9(4): 486.
    [54] Haworth RA, Hunter DR, Berkoff HA. The isolation of Ca2+-reaifltant myocytes from the adult rat. J Mol Cell Cardiol. 1980; 12(1): 715-24.
    [55] Grinberg M, Schwarz M, Zaltsman Y, et al. Mitochondrial carrier homolog 2 is a target of tBID in cells signaled to die by tumor necrosis factor alpha. Mol Cell Bio. 2005; 25(11): 4579-90.
    [56] Kumarswamy R, Chandna S. Putative partners in Bax mediated cytochrome-C release:ANT, CypD, VDAC or none of them. Mitochondrion. 2009; 9(1): 1-8.
    [57] Dejean LM, Martinez-Caballero S, Guo L, et al. Oligomeric Bax is a component of the putative cytochrome C release channel MAC,mitochondrial apoptosis-induced channel.Mol Biol Cell. 2005; 16(5): 2424-32.
    [58] Cheng EH, Wei MC, Weiler S, et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX-and BAK-mediated mitochondrial apoptosis. Mol Cell. 2001; 8(3): 705-11.
    [59] Yu X, Wang L, Acehan D, et al. Three-dimensional structure of a double apoptosome formed by the Drosophila Apaf-1 related killer. J Mol Biol. 2006; 355(3): 577-58.
    [60] Otera H, Ohsakaya S, Nagaura Z, et al. Export of mitochondrial AIF in response to proapoptotic stimuli depends on processing at the intermembrane space.EBMO J. 2005; 24(7): 1375-86.
    [61]周艺群,谷志远.线粒体与细胞凋亡.解剖科学进展. 2006; 12(1): 60-2.
    [62] Linda E, Frank AE, Kruyt E, et al. Cell death independent of caspases: A review. Clin Cancer Res. 2005; 11(9): 3155-62.
    [63] Roberts DL, Merrison W, MacFarlane M, et al. The inhibitor of apoptosis protein-binding domain of smac is not essential for its proapoptotic activety. J Cellular Biochem. 2001; 153(1): 221-8.
    [64]王晓樑,王瑾,吕小萍,等.粒体丝氨酸蛋白酶Omi/HtrA2与细胞凋亡.生理科学进展. 2006; 37(3): 285-8.
    [65] Simonnel H, Alazard N, Pfeiffer K, et al. Low mitochondrial respiratory chain content correlates with tumor aggressiveness in renal cell carcinoma.Carcinogenesis. 2002; 23(5):759-68.
    [66] Young TW, Mei FC, Yang G, et al. Activation of antioxidant pathways in ras-mediated oncogenic transformation of human surface ovarian epithelial cells revealed by functional proteomics and mass spectrometrv. Cancer Res. 2004; 64(13): 4577-84.
    [67] Kawanishi S, Hiraku Y, Oikawa S. Mechanism of guanine-specific DNA damage by oxidative stress and its role incarcinogenesis and aging. Mutat Res. 2001; 488(1): 65-76.
    [68] Zhu W, Qin W, Sauter ER. Large-scale mitochondrial DNA deletion mutations and nuclear genome instability in human breast cancer.Cancer Detect Prev, 2004, 28 (2):119-26.
    [69] Le Mellay V, Troppmair J, Benz R, et al. Negative regulation of mitochondrial VDAC channels by C-Raf kinase. BMC Cell Biol. 2002; 3: 14.
    [70] Kolb JP, Kern C, Quiney C, et al. Re-establishment of a normal apoptotic process as a therapeutic approach in B-CLL. Curr Drug Targets Cardiovasc Haematol Disord. 2003; 3(4): 261-86.
    [71] Kim M, Park SY, Pai HS, et al. Hypoxia inhibits tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by blocking Bax translocation. Cancer Res. 2004; 64(12): 4078-81.
    [72] Subbegowda R, Froommmel TO. Aspirin toxicity for human colonic tumor cells from necrosis and is accompanied by cell cycle arrest. Cancer Res. 1998; 58(13): 2772-6.
    [73] Shiff SJ, Qiao L, Tsai LL, et al. Sulindac sulfide, an aspirin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells. J Clin Invest. 1995; 96(1): 491-503.
    [74] Han EK, Arber N, Yamamoto H, et al. Effects of sulindac and its metabolites on growth and apoptosis in human mammary epithelial and breast carcinoma cell lines. Breast Cancer Res Treat. 1998; 48(3): 195-203.
    [75]柯小龙,李红良,陈丹丹等. SC-58125对HepG2细胞增殖和凋亡的影响.中国病理生理杂志. 2002; 18(1): 51-4.
    [76] Li HX, Li XK, Cai SH et al. Synergistic effects of nimesulide and 5-fluorouracil on tumor growth and apoptosis in the implanted hepatoma in mice. World J Gastroentero. 2003; 9(5): 936-40.
    [77]徐美虹,吴开春,吴汉平等.非甾体类抗炎药对胃癌细胞增殖的抑制作用评论推荐.中华消化杂志. 2002; 22(4): 199-201.
    [78]张坚,徐卓群,胡强等.选择性环氧合酶2抑制剂对人膀胱癌细胞T24增殖和凋亡的作用.癌症. 2007; 26(4): 377-81.
    [79]钟鹰,李国庆.非甾体类抗炎药抗肿瘤作用研究进展.现代医药卫生. 2009; 25(23): 3584-6.
    [80]任先达,吕艳青,叶开和,等.非甾体类抗炎药抗肿瘤作用的分子机制.中国药理学通报. 2005; 21(5): 527-31.
    [81] Jana NR. NSAIDs and apoptosis. Cell Mol Life Sci. 2008; 65(9): 1295-301.
    [82]张超.塞来昔布对人结肠癌细胞裸鼠移植瘤肝转移和血管生成的影响.第三军医大学学报. 2006; 28(5): 451-3.
    [83] Lanza-Jacoby S, Miller S, Flynn J, et al. The cyclooxygenase-2 inhibitor, celecoxib, prevents the development of mammary tumors inHer-2 /neumice Cancer. Epidemiol Biomarkers JP. 2003; 12(12): 1486-91.
    [84] Abou-Issa HM, Alshafie GA, Seibe rt K, et al. Dose response efects of the COX-2 inhibitor, celecoxib, on the chemoprevention of mammary carcinogenesis. Anticancer Res. 2001; 21(5): 3425-32.
    [85]张岩,王杨,闫秀欣.阿司匹林和塞来昔布对乳腺癌细胞MCF-7增殖的影响.吉林大学学报(医学版). 2007; 33(5): 901-4.
    [86] Kem MA, Schubert D, Sahi D, et al. Proapoptoti candan tiprolif-erative potential of selective cyclooxygenase-2 inhibitors in humanliver tumor cells. Hepatology. 2002; 36(4Pt1): 885-94.
    [87] Altorki NK, Keresztes RS, Port JL, et al. Celecoxib, a selectivecyclooxygenase-2 inhibitors, enhances the response to preoperativepaclitaxel and carboplatin in early stage non-small-cell lungcancer. JClin Oncol. 2003; 21(14): 2645-50.
    [88] Qiu DK, Ma X, Peng YS, et al. Sigificance of cyclooxygenase-2 expression in human primary hepatocallular carcinoma.World J Gastroenterol. 2002; 8(5): 815-7.
    [89] Bae SH, Jung ES, Park YM, et al. Expression of cyclooxygenase-2(COX-2) in hpatocelluar carcinoma and growth inhibition of hepatoma celllines by a COX-2inhibitor, NS 2398. ClinCancerRes. 2001; 7(5): 1410-8.
    [90]廖璞,付晓,廖雪松,等.原发性肝癌血清磷脂酶A2活性变化及临床意义.重庆医学.2000; 29(3): 203-4
    [91]陈巍,章月丽,唐存存.肺癌患者血清磷脂酶A2的测定.海峡医学1991; 15(4): 324-5.
    [92] Yamashita S, Yamashita J, Ogawa M. Overexpression of group II phospholipase A2 in human breast cancer tissues is closely associated with their malignant potency. Br J Cancer. 1994; 69(6): 1166-70.
    [93] Marquez-Rosado L, Trejo-Soli MC, Garcia-Cuellar CM, et al. Celecoxib, a cyclooxygenase-2 inhibitor, prevents induction of liver preneoplastic lesions in rats. J Hepatology. 2005; 43(4): 653- 60.
    [94] Adrain C, Martin SJ. The mitochondrial apoptosome: a killer unleashed by the cytochromeseas. Trends Biochem Sci. 2001; 26(6):390-7.
    [95]丁斌,谢勇,周南,等. PPAR-γ的激动剂吡咯列酮抑制肝癌细胞生长和诱导凋亡的实验研究.江西医药. 2008; 43(7): 654-6.
    [96]张素霞,官阳,杨木兰,等. PARγ激动剂对肝癌细胞HepG2增殖的影响及其机制探讨.华中科技大学学报(医学版. 2009; 38(5): 627-31.
    [97]于洪波,姚登福,吴玮,等.肝癌及癌周组织核转录因子-κB表达的免疫组织化学分析.现代医药卫生. 2008; 24(12): 1746-8.
    [98]苏小琴,姚登福,章建国,等.肝细胞癌变核转录因子-κB及肿瘤坏死因子-α动态表达与改变.南通大学学报(医学版). 2006; 2(3): 163-6.
    [99] Khammanit R., Chantakru S., Kitiyanant Y., et al. Effect of serum starvation and chemical inhibitors on cell cycle synchronization of canine dermal fibroblasts. Theriogenology. 2008; 70(1), 27-34.
    [100]黄作君,徐刚,黎月玲,等.高效液相色谱法测定大鼠血浆中游离脂肪酸浓度.广东药学院学报. 2004; 22(6): 656-8.
    [101]Taketo MM. Cyclooxygenase-2 inhibitors in tumorigenesis (partII). J Natl Cancer Inst.1998 ; 90(21): 1609-20.
    [102]Yano T, Zissel G, Muller-Qernheim J, et al. Prostaglandin E2 reinforces the activation of Ras signal pathway in lung adenocarcinoma cells via EP3. FEBS Lett. 2002; 518(1-3): 154-8.
    [103]Pai R, Soreghan B, Szabo H, et al. Prostaglandin E2 tranactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. Nat Med.2002 ; 8(3): 289-93.
    [104]Balsinde J, Balboa MA, Insel PA, et al. Regulation and inhibition of phospholipase A2. Annu Rev Pharmacol Toxicol. 1999; 39: 175-189.
    [105]Murakami M, Kudo I. Phospholipase A2. J Biochem. 2002; 131(3): 285-92.
    [106]Ikeda T, Tozuka S, Hasumura Y, et al. Prostaglandin-E-producing hepatocellular carcinoma with hypercalcemia. Cancer.1988; 61(9): 1813-4.
    [107]Smith WL, Langenbach R. Why there are two cyclooxygenase isozymes. J Clin Invest. 2001; 107(12):1491-5.
    [108]Gupta RA, Dubois RN. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nat Rev Cancer. 2001; 1(1): 11-21.
    [109]Dommels YE, Haring MM, Keestra NG, et al. The role of cyclooxygenase in n-6 and n-3 polyunsaturated fatty acid mediated effects on cell proliferation, PGE(2) synthesis and cytotoxicity in human colorectal carcinoma cell lines. Carcinogenesis. 2003; 24(3): 385-92.
    [110]顾宇平,束永前,张炜明,等.非小细胞肺癌中COX-2、VEGF和MVD的表达及其意义.苏州大学学报(医学版). 2006; 26(4): 625-8.
    [111]安莉,王静,俞森洋,等. COX-2和p53蛋白在非小细胞肺癌中的表达及与微血管生成的相关性.中国肿瘤临床与康复. 2007; 14(1): 1-4.
    [112]Kim WJ, Yoon JM, Lee KH, et al. Effect of uteroglobin on cPLA2, COX-2 expression and ERK activation in NSCLC Cells. Tuberc Respir Dis. 2004; 56(6): 638-45.
    [113]何晓燕,张敏,陈智超,等. PPAR-γ在肺癌中的表达及在肺癌凋亡中的作用研究.中国肺癌杂志. 2006; 9(1): 35-9.
    [114]张敏,游泳,邹萍,等. PPAR-γ在肺癌中的表达及在肺癌凋亡中的作用研究.中德临床肿瘤学杂志(英文版). 2005; 4(1): 36-9.
    [115]马路,李成继,周厚清,等.转录因子NF-kB在非小细胞肺癌中的表达分析.中国医师杂志. 2008; 10(6): 822-4.
    [116]马国栋,刘志勇,邹卫,等.核转录因子kB在非小细胞肺癌中的表达及意义.临床肺科杂志. 2008; 13(9): 1178-80.
    [117]Cui W, Hu SX, Tang ZY, et al. In-vivo effects and mechanisms of celecoxib-reduced growth of cyclooxygenase-2 (COX-2) expressing versus COX-2-deleted human HCCxenografts in nude mice. Anticancer Drugs. 2008; 19(9): 891-7.
    [118]Schonthal AH. Induction of apoptosis by celecoxib in cell culture: an uncertain role for cyclooxygenase-2. Cancer Res. 2007; 67(11): 5575-6.
    [119]孙凤林,魏晓红,徐世东,等. Celecoxib对肺癌细胞生物学行为影响的研究.中国肿瘤临床. 2005; 32(11): 621-4.
    [120]龙伟,周锐. Celecoxib通过抑制Survivin表达促进肺癌A549细胞凋亡.中国肿瘤. 2006; 15(5): 325-8.
    [121]Bonovas S, Tsantes A, Drosos T, et al. Cancer chemoprevention: a summary of the current evidence. Anticancer Res. 2008 ; 28(3B): 1857-66.
    [122]Cheng J, Imanishi H, Amuro Y, et al. NS-398, a selective cyclooxygenase 2 inhibitor, inhibited cell growth and induced cell cycle arrest in human hepatocellular carcinoma cell lines. Int J Cancer. 2002 ; 99(5): 755-61.
    [123]Cui W, Yu CH, Hu KQ, et al. In vitro and in vivo effects and mechanisms of celecoxib-induced growth inhibition of human hepatocellular carcinoma cells. Clin Cancer Res. 2005; 11(22): 8213-21.
    [124]Maier TJ, Schilling K, Schmidt R, et al. Cyclooxygenase-2 (COX-2)-dependent and -independent anticarcinogenic effects of celecoxib in human colon carcinoma cells. Biochem.Pharmacol. 2004; 67(8): 1469-78.
    [125]刘纯伦,欧武陵.塞来昔布对肝癌SMMC27721细胞的生长抑制作用.第三军医大学学报. 2006; 8(26): 682-5.
    [126]唐保东,周奇,林箐华,等.塞来昔布对人肝癌细胞增殖的抑制作用.南方医科大学学报. 2007; 27(10):1511-3.
    [127]徐志远,戴朝六,王宁,等.塞来昔布对人肝癌细胞株增殖及凋亡的影响.肿瘤学杂志. 2008; 14(11): 902-5.
    [128]Grosch S, Maier TJ, Schiffmann S, et al. Cyclooxygenase-2 (COX-2) -independent anticarcinogenic effects of selective COX-2 inhibitors. J Natl Cancer Inst. 2006; 98(11): 736-47.
    [129]Kashfi K, Rigas B. Non-COX-2 targets and cancer: expanding the molecular target repertoire of chemoprevention. Biochem.Pharmacol. 2005; 70(7): 969-86.
    [130]Han C, Michalopoulos GK, Wu T, et al. Prostaglandin E2 receptor EP1 transactivatesEGFR/MET receptor tyrosine kinases and enhances invasiveness in human hepatocellular carcinoma cells. J Cell Physiol. 2006; 207(1): 261-70.
    [131]梁莉延.磷脂酶A2的研究进展.国外医学(生理、病理科学与临床分册). 2000; 20(6): 474-6.
    [132]Aggarwal A, Guo DL, Hoshida Y, et al. Topological and Functional Discovery in a Gene Coexpression Meta-Network of Gastric Cancer. Cancer Res. 2006; 66(1): 232-41.
    [133]Shin SM, Kim SG. Inhibition of arachidonic acid and iron-induced mitochondrial dysfunction and apoptosis by oltipraz and novel 1,2-dithiole-3-thione congeners. Mol Pharmacol. 2009; 75(1): 242-53.
    [134]Scorrano L, Penzo D, Petronilli V, et al. Arachidonic acid causes cell death through the mitochondrial permeability transition. Implications for tumor necrosis factor-alpha aopototic signaling. J Biol Chem. 2001; 276(15): 12035-40.
    [135]Nakanishi M, Rosenberg DW. Roles of cPLA2 alpha and arachidonic acid in cancer. Biochim Biophys Acta. 2006; 1761(11): 1335-43.
    [136]Gately S, Kerbel R. Therapeutic potential of selective cyclooxygenase-2 inhibitors in the management of tumor angiogenesis. Prog Exp Res. 2003; 37: 179-92.
    [137]Pang L, Nie M, Corbett L, et al. Cyclooxygenase-2 expression by nonsteroidal anti-inflammatory drugs in human airway smooth muscle cells: role of peroxisome proliferator-activated receptors. J Immunol. 2003; 170(2): 1043-51.
    [138]王春晖,欧阳钦,唐承薇.非甾体类抗炎药经转录活化蛋白-1及核因子-κB信号传导通路抑制结肠癌细胞生长.中华消化杂志. 2005; 25(8): 483-7.
    [139]Schroeder CP, Kadara H, Lotan D, et al. Involvement of mitochondrial and Akt signaling pathways in augmented apoptosis induced by a combination of low doses of celecoxib and N-(4-hydroxyphenyl) retinamide in premalignant human bronchial epithelial cells. Cancer Res. 2006; 66(19): 9762-70.
    [140]Ding H, Han C, Zhu J, et al. Celecoxib derivatives induce apoptosis via the disruption of mitochondrial membrane potential and activation of caspase 9. Int J Cancer. 2005; 113(5): 803-10.
    [141]Jendrossek V, Handrick R, Belka C. Celecoxib activates a novel mitochondrial apoptosis signaling pathway. FASEB J. 2003; 17(11):1547-9.
    [142]张桂英,彭杰,肖志强,等.塞来昔布诱导大肠癌细胞凋亡与细胞色素C依赖性信号途径活化相关.中华消化杂志. 2004; 24(9): 537-40.
    [143]俞万钧,修清玉,李兵,等.塞来昔布对Lewis肺癌的抑制作用.第二军医大学学报. 2005; 25(6): 505-7.
    [144]Stolina M, Sharma S, Lin Y, et al. Spectific inhibition of cyclooxygenase-2 restore antitumor reactivity by altering the balance of IL-10 and IL-12 synthesis. J Immunol, 2000; 164(2): 361-70.
    [145]张京伟,周云峰,胡名柏,等.塞来昔布对Lewis肺癌细胞增殖及移植瘤的抑制作用.武汉大学学报(医学版). 2008; 29(4): 485-8.
    [146]Cummings BS. Phospholipase A2 as targets for anti-cancer drugs. Biochem Pharmacol. 2007; 74(7): 949-59.
    [147]Krey G, Braissant O, L'Horset F, et al. Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator- dependent receptor ligand assay. Mol Endocrinol. 1997; 11(6): 779-91.
    [148]Forman BM, Chen J, and Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl Acad Sci U S A. 1997; 94(9): 4312-7.
    [149]Lehmann JM, Lenhard JM, Oliver BB, et al. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti- inflam- matory drugs. 1997; J Biol Chem 272(6): 3406-10.
    [150]Shaik MS, Chatterjee A, Jackson T, et al. Enhancement of antitumor activity of docetaxel by celecoxib in lung tumors. Int J Cancer. 2006; 118(2): 396-404.
    [151]Nikitakis NG, Hebert C, Lopes MA, et al. PPARgamma-mediated antineoplastic effect of NSAID sulindac on human oral squamous carcinoma cells. Int J Cancer. 2002; 98(6): 817-23.
    [152]Wick M, Hurteau G, Dessev C, et al. Peroxisome proliferator-activated receptor-gamma is a target of nonsteroidal anti-inflammatory drugs mediating cyclooxygenase-independent inhibition of lung cancer cell growth. Mol Pharmacol 2002; 62(5): 1207-14.
    [153]Wright HM, Clish CB, Mikami T, et al. A synthetic antagonist for the peroxisome proliferator-activated receptor gamma inhibits adipocyte differentiation. J Biol Chem.2000; 275(3): 1873-7.
    [154]Elrod HA, Sun SY. PPARgamma and Apoptosis in Cancer. PPAR Res. 2008; 2008: 704165.

© 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