用户名: 密码: 验证码:
羧胺三唑的抗癌新机制抑制肿瘤相关巨噬细胞中促炎细胞因子的释放
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
     羧胺三唑(carboxyamidotriazole, CAI)是一种非细胞毒类药物,能够抑制体外多种肿瘤细胞系的增殖,诱导肿瘤细胞凋亡。CAI还被认为能够抑制血管的形成和抑制肿瘤的转移。这些作用被推测可能与CAI抑制钙离子跨膜内流有关,但其具体的分子机制尚无定论。
     我们实验室近年来发现,CAI不仅对肿瘤细胞有直接作用,还在一系列急慢性炎症模型中表现出良好的抗炎作用。我们通过巴豆油致小鼠耳肿胀模型、棉球植入致大鼠肉芽肿实验模型和大鼠佐剂性关节炎模型分别观察到CAI对急性炎症、慢性增殖相和自身免疫性炎症均有明显的抑制作用,并且能明显下调炎症部位或炎症动物血浆中促炎细胞因子的含量。
     炎症和癌症之间有着重要的关系。早在19世纪,Virchow提出了慢性炎症可能会导致细胞增殖从而产生肿瘤的假说。进入21世纪以来,炎癌关系重新引起了科学家们极大的兴趣。流行病学研究结果表明感染和慢性炎症与肿瘤的发生有很大的关联。另有很多研究证实,在肿瘤出现后的生长过程中,肿瘤微环境中的炎症细胞和介质也会扮演肿瘤细胞“帮凶”的角色,其中肿瘤相关巨噬细胞(TAMs)和肿瘤坏死因子-α(TNF-α)等促炎细胞因子的作用得到广泛认同。
     综合以上背景,我们提出了以下设想:具有抗癌和抗炎双重作用的CAI是否能够作用于在炎、癌发展中均发挥重要作用的巨噬细胞?除能抑制炎症模型中促炎细胞因子,是否还能抑制肿瘤微环境中促炎细胞因子的产生?CAI抑制肿瘤微环境中的促炎细胞因子是否是其抗肿瘤作用的另一个机制?在本项课题,我们围绕这些问题展开了研究。
     研究方法
     我们提取佐剂性关节炎大鼠的腹腔巨噬细胞(动物连续21天给予溶剂或20mg/kg/d CAI),进行酶联免疫吸附试验(ELISA)测定其TNF-α的分泌。建立C57BL/6小鼠Lewis肺癌(LLC)模型,动物连续14天给予溶剂或20mg/kg/d CAI,对其肿瘤组织中的TNF-α进行免疫组化染色,用ELISA方法测定其肿瘤组织匀浆中TNF-α的含量,并获取肿瘤组织中的巨噬细胞,进行逆转录-聚合酶链式反应试验(RT-PCR)测定其中TNF-αmRNA的表达。另外,我们参考文献,将原代培养的小鼠腹腔巨噬细胞或小鼠巨噬细胞系RAW264.7细胞用Lewis肺癌细胞(LLC细胞)的条件培养基进行诱导,或将这些巨噬细胞直接与LLC细胞共培养,用RT-PCR方法和ELISA方法检测药物对这些经肿瘤条件诱导的巨噬细胞合成和释放TNF-α的影响。在这些实验中,我们采用影响多种细胞因子的药物——地塞米松(Dexamethasone, DEX)作为工具,研究了DEX对TNF-α的影响及其对CAI效应的增强作用。
     我们在巨噬细胞与LLC细胞共培养的体系中,用CCK-8试剂测定腹腔巨噬细胞对LLC细胞增殖的促进作用,并观察DEX增强共培养体系中CAI对LLC细胞增殖的抑制作用;通过结晶紫染色方法观察药物预处理的RAW264.7巨噬细胞对LLC细胞迁移和侵袭的影响,并在该体系中加入TNF-α抗体观察TNF-α在巨噬细胞影响LLC细胞迁移过程中的作用。
     我们用CCK-8试剂还观察了在单独培养LLC细胞时,DEX对CAI抑制LLC细胞增殖作用的影响;并在小鼠LLC模型中,观察了DEX对CAI抑制肿瘤生长作用的影响,对肿瘤组织冰冻切片进行CD31分子的免疫组化染色,观察血管生长的情况。
     为验证DEX对CAI抗肿瘤效应的增强较具有普遍性,我们利用A549裸鼠移植瘤模型对二者联合用药的效应再次进行了研究,并检测了血管生长的情况、肿瘤组织匀浆中TNF-α含量的变化和联合用药对荷瘤动物腹腔巨噬细胞中TNF-α释放的影响。
     研究结果
     CAI对佐剂性关节炎大鼠腹腔巨噬细胞中炎症介质TNF-α的释放有显著的抑制作用。不仅如此,肿瘤组织免疫组化染色、ELISA和荧光定量PCR实验结果显示,它也能明显抑制LLC肿瘤组织巨噬细胞中TNF-α的表达和释放。对肿瘤组织内的CD31分子进行染色发现,CAI对LLC肿瘤组织中血管的形成有抑制作用。
     体外实验中,我们发现正常C57BL/6小鼠的腹腔巨噬细胞在与LLC肿瘤细胞共培养时,p65向细胞核处聚集,可能表示有炎症反应在一定程度上被激活。CAI能够显著抑制与LLC肿瘤细胞共培养后的小鼠腹腔巨噬细胞TNF-α释放的增加,对经LLC肿瘤条件培养基诱导的小鼠腹腔巨噬细胞IL-10生成有上调作用。不仅如此,CAI还能显著抑制受LLC肿瘤细胞条件培养基诱导后的小鼠巨噬细胞样细胞RAW264.7中TNF-α、IL-6表达和释放的增加。
     在体外共培养模型中,我们发现正常C57BL/6小鼠的腹腔巨噬细胞能够显著刺激同源的LLC肿瘤细胞的增殖,而DEX和CAI能够有效抑制腹腔巨噬细胞的这种作用。RAW264.7细胞显著促进LLC肿瘤细胞的迁移和侵袭,在这个体系中加入TNF-α抗体能够有效抑制RAW264.7细胞引起的LLC细胞迁移,经过CAI和/或DEX预处理的RAW264.7细胞促进LLC细胞迁移和侵袭的作用减弱。
     CAI能够直接作用于LLC肿瘤细胞,显著抑制其增殖,而DEX未能促进CAI对LLC肿瘤细胞的直接抑制作用。CAI与DEX联合用药显著抑制C57BL/6小鼠中LLC肿瘤的生长,延长荷瘤动物的生存期,并且对动物的体重和生存状态没有产生明显影响。
     在裸鼠A549移植瘤模型中,CAI与DEX联合用药能够显著抑制A549移植瘤的生长,显著减少A549抑制瘤组织中血管的生成和下调A549抑制瘤组织匀浆TNF-α和TGF-β的含量。通过对荷瘤动物腹腔巨噬细胞进行研究,发现CAI和DEX联合用药将腹腔巨噬细胞中TNF-α的释放降至极低水平。
     研究结论
     CAI不仅对肿瘤细胞有直接作用,影响炎症模型中的细胞因子从而发挥抗炎作用,还能够对肿瘤环境中巨噬细胞合成或释放细胞因子产生影响,这些细胞包括体外经肿瘤条件诱导的巨噬细胞和体内移植瘤组织内的巨噬细胞。在这些模型中,能够广泛影响细胞因子的药物——DEX也能够在一定程度上抑制肿瘤的生长,并对CAI的抗肿瘤效应有着很强的促进作用。因此,影响肿瘤微环境中细胞因子的产生可能是CAI抗肿瘤作用的机制之一。总结我们在炎症模型和肿瘤模型中发现的CAI对炎症介质的影响,我们认为CAI成为研究炎癌关系非常良好的工具药物。小剂量地塞米松与CAI的联合应用在肿瘤发展的增殖、迁移、侵袭和血管生长等环节都表现出很强的抑制作用,对于优化CAI的抗肿瘤作用有着重要意义,将来可能能够成为临床上理想的联合用药方案之一。
Backgound
     Carboxyamidotriazole (CAI) is a non-cytotoxic anticancer drug in development. It inhibits proliferation and induces apoptosis of some cancer cells. A series of clinical trails have shown that the adverse effects of the drug are slight comparing with other anticancer drugs, although its effect is still in investigation. It has been suggested that CAI is a voltage-independent calcium channel inhibitor of cancer cells, but the exact anti-cancer mechanism is not quite clear yet.
     In our previous study, we found that CAI also possessed great anti-inflammation action in a variety of animal models of acute and chronic inflammation. In the adjuvant-induced arthritis (AA) model, the decrease of the pro-inflammatory cytokines at the site of inflammation and in serums by CAI was observed.
     Considering CAI has both anti-cancer and anti-inflammation activities, we then focused on its effects on macrophages, which were important both for tumor growth and inflammation processes. We wondered if CAI might also have effects on the pro-inflammatory cytokines production in tumor associated macrophages (TAMs), which might be one of the mechanisms for the anti-cancer actions of CAI.
     Methods
     Adjuvant-Induced Arthritis (AA) Model was established and peritoneal macrophages were isolated from AA rats on day 21. Tumor necrosis factor-a (TNF-α) production in these macrophages was measured by ELISA assay. Lewis Lung Carcinoma (LLC) Model was established and the tumor tissues were obtained on day 14. TNF-αand CD31 in the tumor tissues was stained immunohistochemically and the concentration of TNF-a was measured by ELISA assay. The macrophages were isolated from the tumor tissues with collagenase and TNF-a mRNA level in these macrophages was determined by RT-PCR assay. The peritoneal macrophages from C57 mice or RAW264.7 macrophages were co-cultured with LLC cells or LLC conditioned medium. The TNF-a and Interleukin-6 (IL-6) expression in these macrophages was tested by ELISA and RT-PCR assay. In the above tests, dexamethasone (DEX) was combined with CAI as it is known to inhibit the pro-inflammatory cytokines.
     The proliferation of the LLC cells cultured alone or with macrophages was measured with CCK-8 agent. The migration and invasion of the LLC cells cultured alone or with macrophages was measured with crystal violet agent. The TNF-a neutralizing antibody was used to verify the action of TNF-a.
     A549 xenograft model in nude mice was established to see whether the great inhibition caused by the combination of CAI and DEX was a much commen effect. The blood vessel growth and TNF-a concentration in tumor tissues were also tested.
     Results
     CAI suppressed TNF-a production in peritoneal macrophages isolated from rats of adjuvant-induced arthritis.
     CAI decreased TNF-a in LLC tumor tissues and suppressed the expression of mRNA for TNF-a in tumor tissue macrophages. It also inhibited the angiogenesis in LLC tumor tissues.
     CAI inhibited TNF-a and IL-6 production in macrophages induced by LLC cells or its conditioned medium. Proliferation of LLC cells was promoted by peritoneal macrophages; DEX enhanced the inhibition of CAI on the proliferation of LLC cells co-cultured with peritoneal macrophages instead of LLC cells cultured alone. RAW264.7 cells promoted LLC cells invasion and the activity was inhibited if RAW264.7 cells were pre-treated with CAI. The activity was also inhibited if the TNF-a neutralizing antibody was added in the co-culture system.
     DEX did not affect the direct inhibition of CAI on the proliferation of LLC cells cultured alone. But the combination treatment of CAI and DEX significantly inhibited the LLC tumor growth in mice with the survival time prolonged. The addition of DEX did not influence the body weights.
     In the A549 xenograft model, it is also observed that combination of CAI and DEX significantly inhibited the tumor growth and blood vessels growth in the tumor tissues. The concentrations of TNF-αand TGF-βin tumor homogenates and TNF-αproduction in peritoneal macrophages of A549 tumor bearing mice were all decreased by CAI and/or DEX.
     Conclusion
     In summary, we have found that CAI can not only act on tumor cells directly, but also on macrophages, including those in arthritis model and TAMs. It can suppress TNF-a and IL-6 production, break the "smoldering" inflammation balance in TAMs, and thus inhibit tumor growth indirectly. The findings suggest that CAI exerts anti-cancer activities may be due to its direct actions on both tumor cells and TAMs. The newly found anti-inflammation activity may make CAI one of effective tools to study the relationship between inflammation and cancer, and the combination with low dose DEX may be of great importance for clinical application of CAI.
引文
1. Coussens LM and Werb Z. Inflammation and cancer. Nature,2002,20:860-867.
    2. Shacter E and Weitzman SA. Chronic inflammation and cancer. Oncology,2002,16: 217-226.
    3. Hussain SP, Hofseth LJ and Harris, CC. Radical causes of cancer. Nat Rev,2003,3: 276-285.
    4. Fox JG and Wang TC. Inflammation, atrophy, and gastric cancer. J Clin,2007,117: 60-69.
    5. Dobrovolskaia MA and Kozlov SV. Inflammation and cancer:when NF-kappaB amalgamates the perilous partnership. Curr Cancer Drug Targets,2005,5:325-344.
    6. Balkwill F, Charles KA and Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell,2005,7:211-217.
    7. Karin M and Greten FR. NF-kB:linking inflammation and immunity to cancer development and progression. Nat Rev,2005,5:749-759.
    8. Karin M. Nuclear factor-kB in cancer development and progression. Nature,2006,441: 431-436.
    9. Ben-Baruch A. Inflammation-associated immune suppression in cancer:the roles played by cytokines, chemokines and additional mediators. Semin Cancer Biol,2006,16: 38-52.
    10. Smyth MJ, Cretney E, Kershaw MH and Hayakawa Y. Cytokines in cancer immunity and immunotherapy. Immunol Rev,2004,202:275-293.
    11. Kim R, Emi M, Tanabe K and Arihiro K. Tumor-driven evolution of immunosuppressive networks during malignant progression. Cancer Res,2006,66: 5527-5536.
    12. Hadden JW. Immunodeficiency and cancer:prospects for correction. Int Immunopharmacol,2003,3:1061-1071.
    13. Enfissi A, Prigent S, Colosetti P, Capiod T. The blocking of capacitative calcium entry by 2-aminoethyl diphenylborate (2-APB) and carboxyamidotriazole (CAI) inhibits proliferation in Hep G2 and Huh-7 human hepatoma cells. Cell Calcium,2004,36: 459-467.
    14. Jacobs W, Mikkelsen T, Smith R, et al. Inhibitory effects of CAI in glioblastoma growth and invasion. J. Neurooncol,1997,32:93-101.
    15. Guo L, Li ZS, Wang HL, et al. Carboxyamido-triazole inhibits proliferation of human breast cancer cells via G2/M cell cycle arrest and apoptosis. Eur J Pharmacol,2006, 538(1-3):15-22.
    16. Moody TW, Chiles J, Moody E, et al. CAI inhibits the growth of small cell lung cancer cells. Lung Cancer,2003,39:279-288.
    17. Perabo FG, Wirger A, Kamp S, et al. Carboxyamido-triazole (CAI), a signal transduction inhibitor induces growth inhibition and apoptosis in bladder cancer cells by modulation of Bcl-2. Anticancer Res,2004,24:2869-2877.
    18. Wasilenko JK, Shinn, CA, Willis CR, et al. Carboxyamido-triazole (CAI)--a novel "static" signal transduction inhibitor induces apoptosis in human B-cell chronic lymphocytic leukemia cells. Leuk. Lymphoma,2001.42:1049-1053.
    19. Guo L, Ye CY, Chen WY, et al. Anti-inflammatory and analgesic potency of carboxyamidotriazole, a tumorostatic agent. J Pharmacol Exp Ther,2008,325:10-16.
    20. Lin WW and Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. The Journal of Clinical Investigation,2007,117:1175-1183
    21. Johnson WJ, Muirhead KA, Meunier PC, et al. Macrophage activation in rat models of inflammation and arthritis:Systemic activation precedes arthritis induction and progression. Arthritis & Rheumatism,1986,29:1122-1130.
    22. Balkwill F. Tumour necrosis factor and cancer. Nature Rev,2009,9:361-370.
    23. Duff MD, Mestre J, Maddali S, et al. Analysis of gene expression in the tumor-associated macrophage. J Surg Res,2007,142:119-128.
    24. Plytycz B, Pedersen A, BΦgwald J and Seljelid R. In vitro interactions of murine peritoneal macrophages and sarcoma cells. Virchows Arch,1986,50:285-292.
    25. Lee CH, Wu CL and Shiau AL. Toll-like receptor 4 signaling promotes tumor growth. J Immunother,2010,33:73-82.
    26. Schottelius AJ, Mayo MW, Sartor RB, and Baldwin AS. Interleukin-10 signaling blocks inhibitor of kappaB kinase activity and nuclear factor kappaB DNA binding. J Biol Chem,1999,274:31868-31874.
    27. Green CE, Liu T, Montel V, et al. Chemoattractant signaling between tumor cells and macrophages regulates cancer cells migration, metastasis and neovascularization. Plos One,2009,4:1-15.
    28. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer,2004,4:71-78.
    29. Balkwill F. Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev,2002,13:135-141.
    30. Hodge DR, Hurt EM, and Farrar WL. The role of IL-6 and STAT3 in inflammation and cancer. Eur J Cancer,2005,41:2502-2512.
    31. Kohn EC, Alessandro R, Spoonster J, et al. Angiogenesis:role of calcium-mediated signal transduction. Proc Natl Acad Sci,1995,92:1307-1311.
    32. Qin LX, Tang ZY, Li XM, et al. Effect of antiangiogenic agents on experimental animal models of hepatocellular carcinoma. Ann Acad Med Singapore,1999,28(1): 147-151.
    33. Mignen O, Brink C, Enfissi A, et al. Carboxyamidotriazole-induced inhibition of mitochondrial calcium import blocks capacitative calcium entry and cell proliferation in HEK-293 cells. Journal of Cell Science,2005,118:5615-5623.
    34. Ryuto M, Ono M, Izumi H, et al. Induction of vascular endothelial growth factor by tumor necrosis factor alpha in human glioma cells. Possible roles of SP-1. J Biol Chem, 1996,271(45):28220-28228.
    35. Alessandro R, Fontana S, Giordano M, et al. Effects of carboxyamidotriazole on in vitro models of imatinib-resistant chronic myeloid leukemia. J Cell Physiol,2008,215: 111-121.
    36. Backwill F and Mantovani A. Inflammation and cancer:back to Virchow? Lancet, 2001,357:539-545.
    37. Karin M. Nuclear factor-κB in cancer development and progression. Nature,2006, 441:431-436.
    38. Hanahan D and Weinberg RA. The hallmarks of cancer. Cell,2000,100:57-70.
    39. Mantovani A. Inflaming metastasis. Nature,2009,457:36-37.
    40. Borrello MG, et al. Induction of a proinflammatory program in normal human thyrocytes by the RET/PTC1 oncogene. Proc Natl Acad Sci,2005,102:14825-14830.
    41. Guerra C, et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-RAS oncogenes in adult mice. Cancer Cell,2007,11:291-302.
    42. Sparmann A and Bar-Sagi D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell,2004,6:447-458.
    43. Sumimoto H, Imabayashi F, Iwata T and Kawakami Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med, 2006,203:1651-1656.
    44. Shchors K, et al. The Myc-dependent angiogenic switch in tumors is mediated by interleukin 1β.Genes Dev,2006,20:2527-2538.
    45. Bingle L, Brown NJ and Lewis CE. The role of tumor-associated macrophages in tumor progression:implications for new anticancer therapies. J Pathol 2002,196: 254-265.
    46. Maeda H and Akaike T. Nitric oxide and oxygen radicals in infection, inflammation, and cancer. Biochemistry,1998,63:854-865.
    47. Fulton AM, Loveless SE and Heppner GH. Mutagenic activity of tumor-associated macrophages in Salmonella typhimurium strains TA98 and TA100. Cancer Res,1984,44: 4308-4311.
    48. Arnott CH, et al. Tumour necrosis factor-a mediates tumour promotion via PKCa-and AP-1 dependent pathway. Oncogene,2002,21:4728-4738.
    49. Ogmundsdottir HM, Petursdottir Ⅰ and Gudmundsdottir Ⅰ. Interactions between the immune system and breast cancer. Acta Oncol,1995,34:647-650.
    50. Banciu M, Metselaar JM, Schiffelers RM and Storm G. Antitumor activity of liposomal prednisolone phosphate depends on the presence of functional tumor-associated macrophages in tumor tissue. Neoplasia 2008; 10:108-117.
    51. Escandell JM, Carmen Recio M, Manez S, et al. Cucurbitacin R reduces the inflammation and bone damage associated with adjuvant arthritis in Lewis rats by suppression of tumor necrosis factor-a in T lymphocytes and macrophages [J]. J Pharmacol Exp Ther,2007,320:581-590.
    52. Ginderachter J, Movahedi K, Hassanzadeh G, et al. Classical and alternative activation of mononuclear phagocytes:Picking the best of both worlds for tumor promotion. Immunology,2006,211:487-501.
    53. Wu S, Boyer CM, Whitaker RS, et al. Tumor necrosis factor alpha as an autocrine and paracrine growth factor for an ovarian cancer:monokine induction of tumor cell proliferation and tumor necrosis factor alpha expression. Cancer Res,1993,53: 1939-1944.
    54. Gaiotti D, Chung J, Iglesias M, et al. Tumor necrosis factor-alpha promotes human papillomavirus (HPV) E6/E7 RNA expression and cyclin-dependent kinase activity in HPV-immortalized keratinocytes by a ras-dependent pathway. Mol Carcinog,2000,27: 97-109.
    55. Kulbe H, Hagemann T, Szlosarek PW, et al. The inflammatory cytokine tumor necrosis factor-alpha regulates chemokine receptor expression on ovarian cancer cells. Cancer Res,2005,65:10355-10362.
    56. Chuang MJ, Sun KH, Tang SJ, et al. Tumor-derived tumor necrosis factor-alpha promotes progression and epithelial-mesenchymal transition in renal cell carcinoma cells. Cancer Sci,2008,99:905-913.
    57. Wu Y and Zhou BP. TNF-α/NF-κB/Snail pathway in cancer cell migration and invasion. Brit J Cancer,2010,102:639-644.
    58. Bromberg JF, Wrzeszczynska MH, Devgan G, et al. Stat3 as an oncogene. Cell,1999, 98:295-303.
    59. Li B, Vincent A, Cates J, et al. Low levels of tumor necrosis factor a increases tumor growth by inducing an endothelial phenotype of monocytes recruited to the tumor site. Cancer Res,2009,69:338-348.
    60. Kohn EC, Alessandro R, Spoonster J, et al. Angiogenesis:role of calcium-mediated signal transduction. Proc Natl Acad Sci,1995,92:1307-1311.
    61. Goulding NJ. The molecular complexity of glucocorticoid actions in inflammation — a four-ring circus. Curr Opin Pharmacol,2004,4:629-636.
    62. Ovali E, Ozdemir F, Aydin F, et al. The effects of dexamethasone on leukemic cells derived from patients with AML. J Exp Clin Cancer Res,2003,22:85-89.
    63. Eyre HJ, Ward JH. Control of cancer chemotherapy-induced nausea and vomiting. Cancer,1984,54:2642-2648.
    64. Whiteman KR, Johnson H, Xu SQ, et al. Combination therapy with IMGN901 and Lenalidomide plus low-dose Dexamethasone is highly effective in multiple myeloma xenograft models. AACR 100th Annual Meeting 2009.
    65. Rajkumar SV, Jacobus S, Callander NS, et al. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma:an open-label randomised controlled trial. Lancet Oncol,2010,11:29-37.
    66. Dutcher JP, Leon L, Manola J, et al. Phase Ⅱ study of carboxyamidotriazole in patients with advanced renal cell carcinoma refractory to immunotherapy:E4896, an Eastern Cooperative Oncology Group Study. Cancer,2005,104:2392-2399.
    67. Johnson EA, Marks RS, Mandrekar SJ, et al. Phase Ⅲ randomized, double-blind study of maintenance CAI or placebo in patients with advanced non-small cell lung cancer (NSCLC) after completion of initial therapy (NCCTG 97-24-51). Lung Cancer, 2008,60:200-207.
    68. Mikkelsen T, Lush R, Grossman SA, et al. Phase Ⅱ clinical and pharmacologic study of radiation therapy and carboxyamido-triazole (CAI) in adults with newly diagnosed glioblastoma multiforme. Invest New Drugs,2007,25:259-263.
    1. Hanahan D, and Weinberg RA. The hallmarks of cancer. Cell,2000,100:57-70.
    2. Coussens LM, and Werb Z. Inflammation and cancer. Nature,2002,420:860-867.
    3. Dobrovolskaia MA, and Kozlov SV. Inflammation and cancer:when NF-kappaB amalgamates the perilous partnership. Curr. Cancer Drug Targets,2005,5:325-344.
    4. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature, 2008,454:436-444.
    5. Balkwill F, Charles KA, and Mantovani, A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell,2005,7:211-217.
    6. Karin M, and Greten FR. NF-kB:linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol.,2005,5:749-759.
    7. Karin M. Nuclear factor-kB in cancer development and progression. Nature,2006,441: 431-436.
    8. Mantovani A. Cancer:Inflaming Metastasis. Nature,2009,457(1):36-37.
    9. Luster AD, Alon R and von Andrian, UH. Immune cell migration in inflammation: present and future therapeutic targets. Nat. Immunol.,2005,6:1182-1190.
    10. Balkwill F, and Mantovani A. Inflammation and cancer:back to Virchow? Lancet, 2001,357:539-545.
    11. Mantovani A, Sozzani S, Locati M, et al. Macrophage polarization:tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol, 2002,23:549-555.
    12. de Visser KE, Eichten A and Coussens LM. Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer.,2006,6:24-37.
    13. Ben-Baruch A. Inflammation-associated immune suppression in cancer:the roles played by cytokines, chemokines and additional mediators. Semin. Cancer Biol,2006,16: 38-52.
    14. Smyth MJ, Cretney E, Kershaw MH and Hayakawa Y. Cytokines in cancer immunity and immunotherapy. Immunol. Rev.,2004,202:275-293.
    15. Luo JL, Kamata H and Karin M. IKK/NF-kB signaling:balancing life and death-a new approach to cancer therapy. J. Clin. Invest.,2005,115:2625-2632.
    16. Karin M, Cao Y, Greten FR and Li ZW. NF-kB in cancer:from innocent bystander to major culprit. Nat. Rev. Cancer.,2002,2:301-310.
    17. Karin M. NF-kB and cancer:mechanisms and targets. Mol. Carcinog.,2006,45: 355-361.
    18. Wu Y and Zhou BP. TNF-a/NF-KB/Snail pathway in cancer cell migration and invasion. Brit J Cancer,2010,102:639-644.
    19. Pidgeon GP, et al. The role of endotoxin/lipopolysaccharide in surgically induced tumour growth in a murine model of metastatic disease. Br. J. Cancer.,1999,81: 1311-1317.
    20. Taketomi A, et al. Circulating intercellular adhesion molecule-1 in patients with hepatocellular carcinoma before and after hepatic resection. Hepatogastroenterology, 1997,44:477-483.
    21. Akira S, Uematsu S and Takeuchi O. Pathogen recognition and innate immunity. Cell., 2006,124:783-801.
    22. Fritz JH, Ferrero RL, Philpott DJ and Girardin SE. Nod-like proteins in immunity, inflammation and disease. Nat. Immunol.,2006,7:1250-1257.
    23. Robinson MJ, Sancho D, Slack EC, et al. Myeloid C-type lectins in innate immunity. Nat. Immunol.,2006,7:1258-1265.
    24. Klesney-Tait J, Turnbull IR and Colonna M. The TREM receptor family and signal integration. Nat. Immunol.,2006,7:1266-1273.
    25. Han J and Ulevitch RJ. Limiting inflammatory responses during activation of innate immunity. Nat. Immunol.,2005,6:1182-1189.
    26. Shacter E and Weitzman SA. Chronic inflammation and cancer. Oncology,2002,16: 217-226.
    27. Staib F, et al. The p53 tumor suppressor network is a key responder to microenvironmental components of chronic inflammatory stress. Cancer Res,2005,65: 10255-10264.
    28. Sun J, et al. Interactions of sequence variants in interleukin-1 receptor-associated kinase 4 and the toll-like receptor 6-1-10 gene cluster increase prostate cancer risk. Cancer Epidemiol. Biomarkers Prev,2006,15:480-485.
    29. Hugot JP, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature,2001,411:599-603.
    30. Podolsky DK. Inflammatory bowel disease. N. Engl. J. Med.,2002,347:417-429.
    31. Harmey JH, et al. Lipopolysaccharide induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int. J. Cancer., 2002,101:415-422.
    32. Luo JL, Maeda S, Hsu LC, et al. Inhibition of NF-kB in cancer cells converts inflammation-induced tumor growth mediated by TNF-a to TRAIL-mediated tumor regression. Cancer Cell,2004,6:297-305.
    33. Almasan A and Ashkenazi A. Apo2L/TRAIL:apoptosis signaling, biology, and potential for cancer therapy. Cytokine Growth Factor Rev,2003,14:337-348.
    34. Aderem A, Ulevitch RJ. Toll-like receptors in the induction of the innate immune response. Nature,2000,406:782-787.
    35. Lee CH, Wu CL and Shiau AL. Toll-like receptor 4 signaling promotes tumor growth. J Immunother 2010; 33:73-82.
    36. Aggarwal BB. Signalling pathways of the TNF superfamily:a double edged sword. Nat Rev Immunol,2003,3:745-756.
    37. Bradley JR. TNF-mediated inflammatory disease. J Pathol,2008,214:149-160.
    38. Lin WJ and Yeh WC. Implication of Toll like receptor and tumor necrosis factor alpha signaling in septic shock. Shock,2005,24:206-209.
    39. Mocellin S, Rossi CR, Pilati P, and Nitti D. Tumor necrosis factor, cancer and anticancer therapy. Cytokine Growth Factor Rev,2005,16:35-53.
    40. Hussain SP, Hofseth LJ and Harris CC, Radical causes of cancer. Nat. Rev. Cancer., 2003,3:276-285.
    41. Moore RJ, et al. Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat. Med.,1999,5:828-831.
    42. Arnott CH, et al. Expression of both TNF-a receptor subtypes is essential for optimal skin tumour development. Oncogene,2004,23:1902-1910.
    43. Pikarsky E, et al. NF-kB functions as a tumour promoter in inflammation-associated cancer. Nature,2004,431:461-466.
    44. Elgert KD, Alleva DG and Mullins DW. Tumor-induced immune dysfunction:the macrophage connection. J. Leukoc. Biol.,1998,64:275-290.
    45. Shishodia S and Aggarwal BB. Nuclear factor-kappaB activation mediates cellular transformation, proliferation, invasion angiogenesis and metastasis of cancer. Cancer Treat Res,2004,119:139-173.
    46. Johnston DA, Dong B, Hughes CC. TNF induction of jagged-1 in endothelial cells is NFkappaB-dependent. Gene,2009,435:36-44.
    47. Li B, Vincent A, Cates J, et al. Low levels of tumor necrosis factor a increases tumor growth by inducing an endothelial phenotype of monocytes recruited to the tumor site. Cancer Res,2009,69 (1):338-347.
    48. Devoogdt N, Revets H, Kindt A, et al. The tumor-promoting effect of TNF-a involves the induction of secretory leukocyte protease inhibitor. The Journal of Immunology,2006, 177:8046-8052.
    49. Thiery JP, Sleeman JP. Complex networks orchestrate epithelialmesenchymal transitions. Nat Rev Mol Cell Biol,2006,7:131-142
    50. Cardiff RD. Epithelial to mesenchymal transition tumors:fallacious or Snails pace? Clin Cancer Res,2005,11:8534-8537.
    51. Thompson EW, Newgreen DF, Tarin D. Carcinoma invasion and metastasis:a role for epithelial-mesenchymal transition? Cancer Res,2005,65:5991-5995; discussion 5995.
    52. Parker BS, Argani P, Cook BP, et al. Alterations in vascular gene expression in invasive breast carcinoma. Cancer Res,2004,64:7857-7866.
    53. Martin TA, Goyal A, Watkins G, Jiang WG. Expression of the transcription factors snail, slug, and twist and their clinical significance in human breast cancer. Ann Surg Oncol,2005,12:488-496.
    54. Barbera MJ, Puig I, Dominguez D, et al. Regulation of Snail transcription during epithelial to mesenchymal transition of tumor cellsOncogene,2004,23:7345-7354.
    55. Wu Y, Deng J, Rychahou PG, et al. Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell,2009,15:416-428.
    56. Ishihara K, and Hirano T. IL-6 in autoimmune disease and chronic inflammatory proliferative disease. Cytokine Growth Factor Rev,2002,13:357-368.
    57. Hodge DR, Hurt EM and Farrar WL. The role of IL-6 and STAT3 in inflammation and cancer. Eur. J. Cancer.,2005,41:2502-2512.
    58. O'Shea JJ, Gadina M and Schreiber RD. Cytokine signaling in 2002:new surprises in the Jak/Stat pathway. Cell,2002,109(Suppl.):S121-S131.
    59. Haura EB, Turkson J and Jove R. Mechanisms of disease:insights into the emerging role of signal transducers and activators of transcription in cancer. Nat. Clin. Pract. Oncol.,2005,2:315-324.
    60. Huang et al. Effects of IL-6 and AG490 on regulation of Stat3 signaling pathway and invasion of human pancreatic cancer cells in vitro Journal of Experimental & Clinical Cancer Research,2010,29:51.
    61. Guilian Niu, Kenneth L Wright, Mei Huang, et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene,2002,21:2000-2008.
    62. Xie TX, Wei DY, Liu MG, et al. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene,2004,23:3550-3560.
    63. Osborne J, Moore PS, and Chang Y. KSHV encoded viral IL-6 activates multiple human IL-6 signalling pathways. Hum. Immunol,1999,60:921-927.
    64. Bommert K, Bargou RC and Stuhmer T. Signalling and survival pathways in multiple myeloma. Eur. J. Cancer.,2006,42:1574-1580.
    65. Cozen W, et al. IL-6 levels and genotype are associated with risk of young adult Hodgkin lymphoma. Blood,2004,103:3216-3221.
    66. Gado K, et al. Mouse plasmacytoma:an experimental model of human multiple myeloma. Haematologica,2001,86:227-236.
    67. Chauhan D, et al. Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-κB. Blood,1996,87:1104-1112.
    68. Jego G, et al. Pathogen-associated molecular patterns are growth and survival factors for human myeloma cells through Toll-like receptors. Leukemia,2006,20:1130-1137.
    69. Feng J, et al. The rational designed antagonist derived from the complex structure of interleukin-6 and its receptor affectively blocking interleukin-6 might be a promising treatment in multiple myeloma. Biochimie,2006,88:1265-1273.
    70. Becker C, et al. TGFβ suppresses tumor progression in colon cancer by inhibition of IL-6 trans-signaling. Immunity,2004,21:491-501.
    71. Greten FR, et al. IKKβ links inflammation and tumorigenesis in a mouse model of colitisassociated cancer. Cell,2004,118:285-296.
    72. Rose-John S, Scheller J, Elson G and Jones SA. Interleukin-6 biology is coordinated by membrane-bound and soluble receptors:role in inflammation and cancer. J. Leukoc. Biol.,2006,80:227-236.
    73. Abroun S, et al. Receptor synergy of interleukin-6 (IL-6) and insulin-like growth factor-I in myeloma cells that highly express IL-6 receptor a. Blood,2004,103: 2291-2298.
    74. Pestka S, et al. Interleukin-10 and related cytokines and receptors. Annu. Rev. Immunol.,2004,22:929-979.
    75. Schottelius AJ, Mayo MW, Sartor RB and Baldwin AS. Interleukin-10 signaling blocks inhibitor of kappaB kinase activity and nuclear factor kappaB DNA binding. J. Biol. Chem.,1999,274:31868-31874.
    76. Hoentjen F, Sartor RB, Ozaki M and Jobin C. STAT3 regulates NF-kB recruitment to the IL-12p40 promoter in dendritic cells. Blood,2005,105:689-696.
    77. Moore KW, et al. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol.,2001,19:683-765.
    78. Berg DJ, et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4+ TH1-like responses. J. Clin. Invest.,1996,98:1010-1020.
    79. Sellon RK, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect. Immun.,1998,66:5224-5231.
    80. Erdman SE, et al. CD4+CD25+ regulatory T lymphocytes inhibit microbially-induced colon cancer in Rag2-deficient mice. Am. J. Pathol.,2003,162: 691-702.
    81. Maloy K, et al. CD4+CD25+ Tr cells suppress innate immune pathology through cytokinedependent mechanisms. J. Exp. Med.,2003,197:111-119.
    82. Erdman SE, et al. CD4+CD25+ regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res,2003,63:6042-6050.
    83. Erdman SE, et al. CD4+CD25+ regulatory lymphocytes induce regression of intestinal tumors in ApcMin/+ mice. Cancer Res,2005,65:3998-4004.
    84. Larmonier N, et al. Tumor-derived CD4+CD25+ regulatory T cell suppression of dendritic cell function involves TGF-beta and IL-10. Cancer Immunol. Immunother., 2007,56:48-59.
    85. Kundu N and Fulton AM. Interleukin-10 inhibits tumor metastasis, down-regulates MHC class I, and enhances NK lysis. Cell. Immunol.,1997,180:55-61.
    86. Blankenstein T. The role of tumor stroma in the interaction between tumor and immune system. Curr. Opin. Immunol,2005,17:180-186.
    87. Sredni B, et al. Ammonium trichloro(dioxoethylene-o,o')tellurate (AS101) sensitizes tumors to chemotherapy by inhibiting the tumor interleukin 10 autocrine loop. Cancer Res,2004,64:1843-1852.
    88. Ogden CA, et al. Enhanced apoptotic cell clearance capacity and B cell survival factor production by IL-10-activated macrophages:implications for Burkitt's lymphoma. J. Immunol.,2005,174:3015-3023.
    89. Garcia-Hernandez ML, et al. Interleukin-10 promotes B16-melanoma growth by inhibition of macrophage functions and induction of tumour and vascular cell proliferation. Immunology,2002,105:231-243.
    90. Chen WJ and Wahl SM. TGF-β:the missing link in CD4+CD25+ regulatory T cell-mediated immunosuppression. Cytokine Growth Factor Rev,2003,14:85-89.
    91. Becker C, Fantini MC and Neurath MF. TGF-β as a T cell regulator in colitis and colon cancer. Cytokine Growth Factor Rev,2006,17:97-106.
    92. Ghiringhelli F, et al. Tumor cells convert immature myeloid dendritic cells into TGF-β-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J. Exp. Med., 2005,202:919-929.
    93. Derynck R and Zhang YE. Smad-dependent and Smad-independent pathways in TGF-β family signaling. Nature,2003,425:577-584.
    94. Derynck R, Akhurst RJ and Balmain A. TGF-β signaling in tumor suppression and cancer progression. Nat. Genet.,2001,29:117-129.
    95. Parsons R, et al. Microsatellite instability and mutations of the transforming growth factor type Ⅱ receptor gene in colorectal cancer. Cancer Res,1995,55:5548-5550.
    96. Kim SJ, Im YH, Markowitz SD and Bang YJ. Molecular mechanisms of inactivation of TGF-P receptors during carcinogenesis. Cytokine Growth Factor Rev,2000,11: 159-168.
    97. Markowitz S, et al. Inactivation of the type Ⅱ TGF-P receptor in colon cancer cells with microsatellite instability. Science,1995,268:1336-1338.
    98. Yang X, et al. Targeted disruption of Smad3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-β. EMBO J,1999,18:1280-1291.
    99. Jenkins BJ, et al. Hyperactivation of Stat3 in gp130 mutant mice promotes gastric hyperproliferation and desensitizes TGF-P signaling. Nat. Med.,2005,11:845-852.
    100. Chen ML, et al. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-β signals in vivo. Proc. Natl. Acad. Sci,2005,102:419-424.
    101. Mangan PR, et al. Transforming growth factor-β induces development of the TH17 lineage. Nature,2006,441:231-234.
    102. Cho ML, et al. STAT3 and NF-κB signal pathway is required for IL-23-mediated IL-17 production in spontaneous arthritis animal model IL-1 receptor antagonist-deficient mice. J. Immunol,2006,176:5652-5661.
    103. Moseley TA, Haudenschild DR, Rose L, and Reddi AH. Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor Rev,2003,14:155-174.
    104. Witowski J, Ksiazek K and Jorres A. Interleukin-17:a mediator of inflammatory responses. Cell Mol. Life Sci.,2004,61:567-579.
    105. Langrish CL, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med.,2005,201:233-240.
    106. Park H, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol.,2005,6:1133-1141.
    107. Ruddy MJ, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-α is mediated by CCAAT/enhancer-binding protein family members. J. Biol. Chem.,2004,279:2559-2567.
    108. Tartour E, et al. Interleukin 17, a T-cell-derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice. Cancer Res,1999,59:3698-3704.
    109. Numasaki M, et al. IL-17 enhances the net angiogenic activity and in vivo growth of human non-small cell lung cancer in SCID mice through promoting CXCR-2-dependent angiogenesis. J. Immunol.,2005,175:6177-6189.
    110. Numasaki M, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood, 2003,101:2620-2727.
    111. Benchetrit F, et al. Interleukin-17 inhibits tumor cell growth by means of a T-cell-dependent mechanism. Blood,2002,99:2114-2121.
    112. LeBlanc HN and Ashkenazi A. Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ,2003,10:66-75.
    113. Takeda K, et al. Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development. J. Exp. Med.,2002,195: 161-169.
    114. Watford WT, et al. Signaling by IL-12 and IL-23 and the immunoregulatory roles of STAT4. Immunol. Rev.,2004,202:139-156.
    115. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev.,2003,3:133-146.
    116. Hao JS, and Shan, B.E. Immune enhancement and anti-tumour activity of IL-23. Cancer Immunol. Immunother,2006,55:1426-1431.
    117. Langowski JL, et al. IL-23 promotes tumour incidence and growth. Nature,2006, 442:461-465.
    118. Ugai S, et al. Transduction of the IL-21 and IL-23 genes in human pancreatic carcinoma cells produces natural killer cell-dependent and -independent antitumor effects. Cancer Gene Ther,2003,10:771-778.
    119. Lo CH, et al. Antitumour and antimetastatic activity of IL-23. J. Immunol,2003, 171:600-607.
    1.Wightman B. et al. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell,1993,75:855-862.
    2. Pasquinelli AE. et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature,2000,408:86-89.
    3. Selbach M. et al. Widespread changes in protein synthesis induced by microRNAs. Nature,2008,455:58-63.
    4. Baek D. et al. The impact of microRNAs on protein output. Nature,2008,455:64-71.
    5. Lu LF. et al. MicroRNA in the immune system, microRNA as an immune system. Immunology,2009,127:291-298.
    6. Moschos SA. et al. Expression profiling in vivo demonstrates rapid changes in lung microRNA levels following lipopolysaccharideinduced inflammation but not in the anti-inflammatory action of glucocorticoids. BMC Genomics,2007,8:240.
    7. Taganov KD. et al. NF-kappaB-dependent induction of micro-RNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl Acad. Sci. USA,2006,103:12481-12486.
    8. Bhaumik D. et al. MicroRNAs:an important player in maintaining a balance between inflammation and tumor suppression. Cell Cycle,2009,8:1822.
    9. Perry MM. et al. Rapid changes in microRNA-146a expression negatively regulate the IL-1beta-induced inflammatory response in human lung alveolar epithelial cells. J. Immunol.,2008,180:5689-5698.
    10. Xiao B. et al. Induction of microRNA-155 during Helicobacter pylori infection and its negative regulatory role in the inflammatory response. J. Infect. Dis,2009,200: 916-925.
    11.Worm J. et al. Silencing of microRNA-155 in mice during acute inflammatory response leads to derepression of c/ebp Beta and downregulation of G-CSF. Nucleic Acids Res,2009,37:5784-5792.
    12. Hu G. et al. MicroRNA-98 and let-7 confer cholangiocyte expression of cytokine-inducible Src homology 2-containing protein in response to microbial challenge. J. Immunol.,2009,183:1617-1624.
    13. Bazzoni F. et al. Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals. Proc. Natl Acad. Sci. USA,2009,106: 5282-5287.
    14.Gong AY. et al. MicroRNA-513 regulates B7-H1 translation and is involved in IFN-gamma-induced B7-H1 expression in cholangiocytes. J. Immunol.,2009,182: 1325-1333.
    15. Lebwohl M. Psoriasis. Lancet,2003,361:1197-1204.
    16. Sonkoly E. et al. MicroRNAs:novel regulators involved in the pathogenesis of psoriasis? PLoS ONE,2007,2:e610.
    17. Lu TX. et al. MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. J. Immunol.,2009,182:4994-5002.
    18. Nakasa T. et al. Expression of microRNA-146 in rheumatoid arthritis synovial tissue. Arthritis Rheum.,2008,58:1284-1292.
    19. Padgett KA. et al. Primary biliary cirrhosis is associated with altered hepatic microRNA expression. J. Autoimmun.,2009,32:246-253.
    20. Wu F. et al. MicroRNAs are differentially expressed in ulcerative colitis and alter expression of macrophage inflammatory peptide-2 alpha. Gastroenterology,2008,135: 1624-1635.
    21. Calin GA. et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl Acad. Sci. USA,2002 99:15524-15529.
    22. Calin GA. et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc. Natl Acad. Sci. USA,2004,101:2999-3004.
    23. Costinean S. et al. Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc. Natl Acad. Sci. USA,2006,103: 7024-7029.
    24. Esquela-Kerscher A. et al. The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle,2008,7:759-764.
    25. Takamizawa J. et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res.,2004,64: 3753-3756.
    26. Calin GA. et al. A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N. Engl. J. Med.,2005,353:1793-1801.
    27. Yanaihara N. et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell,2006,9:189-198.
    28. He L. et al. A microRNA polycistron as a potential human oncogene. Nature,2005, 435:828-833.
    29. Johnson SM. et al. RAS is regulated by the let-7 microRNA family. Cell,2005,120: 635-647.
    30. Chan JA. et al. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res,2005,65:6029-6033.
    31.Dews M. et al. Augmentation of tumor angiogenesis by a Mycactivated microRNA cluster. Nat. Genet.,2006,38:1060-1065.
    32. Loffler D. et al. Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood,2007:110,1330-1333.
    33. Seike M. et al. MiR-21 is an EGFR-related anti-apoptotic factor in lung cancer from never-smokers. Proc. Natl Acad. Sci. USA,2009:106,12085-12090.
    34. Krichevsky AM. et al. miR-21:a small multi-faceted RNA. J. Cell. Mol. Med.,2009, 13:39-53.
    35. Frankel LB. et al. Programmed cell death 4 (PDCD4) is an important functional target of the micro RNA miR-21 in breast cancer cells. J. Biol. Chem.,2008,283: 1026-1033.
    36. Zhu S. et al. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J. Biol. Chem.,2007,282:14328-14336.
    37. Meng F. et al. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology,2007,133:647-658.
    38. Liu M. et al. Regulation of the cell cycle gene, BTG2, by miR-21 in human laryngeal carcinoma. Cell Res.,2009,19:828-837.
    39. Si ML. et al. miR-21-mediated tumor growth. Oncogene,2007,26:2799-2803.
    40. Schetter AJ. et al. Association of inflammation-related and micro-RNA gene expression with cancer-specific mortality of colon adenocarcinoma. Clin. Cancer Res., 2009,15:5878-5887.
    41. Faraoni I. et al. miR-155 gene:a typical multifunctional micro-RNA. Biochim. Biophys. Acta,2009,1792:497-505.
    42. Motsch N. et al. Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) induces the expression of the cellular microRNA miR-146a. RNA Biol.,2007,4: 131-137.
    43. O'Connell RM. et al. MicroRNA-155 is induced during the macrophage inflammatory response. Proc. Natl Acad. Sci. USA,2007,104:1604-1609.
    44. Tili E. et al. Modulation of miR-155 and miR-125b levels following lipopolysaccharide/TNF-alpha stimulation and their possible roles in regulating the response to endotoxin shock. J. Immunol.,2007,179:5082-5089.
    45. Wang X. et al. Inducible nitric oxide synthase expression is regulated by MAP kinase phosphatase-1. J. Biol. Chem,2009,284:27123-27134.
    46. Gironella M. et al. Tumor protein 53-induced nuclear protein 1 expression is repressed by miR-155, and its restoration inhibits pancreatic tumor development. Proc. Natl Acad. Sci. USA,2007,104:16170-16175.
    47. Tilia E, et al. Mutator activity induced by microRNA-155 (miR-155) links inflammation and cancer. PNAS,2011,108(12):4908-4913.

© 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