全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Molecular Mechanisms of Liver Injury and Hepatocarcinogenesis: Focusing on the Role of Stress-Activated MAPK

DOI: 10.1155/2012/172894

Full-Text   Cite this paper   Add to My Lib

Abstract:

Hepatocellular carcinoma (HCC) is the third most common cause of cancer mortality. Short-term prognosis of patients with HCC has improved recently due to advances in early diagnosis and treatment, but long-term prognosis is still unsatisfactory. Therefore, obtaining a further understanding of the molecular carcinogenic mechanisms and the unique pathogenic biology of HCC is important. The most characteristic process in hepatocarcinogenesis is underlying chronic liver injury, which leads to repeated cycles of hepatocyte death, inflammation, and compensatory proliferation and subsequently provides a mitogenic and mutagenic environment leading to the development of HCC. Recent in vivo studies have shown that the stress-activated mitogen-activated protein kinase (MAPK) cascade converging on c-Jun NH2-terminal kinase (JNK) and p38 plays a central role in these processes, and it has attracted considerable attention as a therapeutic target. However, JNK and p38 have complex functions and a wide range of cellular effects. In addition, crosstalk with each other and the nuclear factor-kappaB pathway further complicate these functions. A full understanding is essential to bring these observations into clinical settings. In this paper, we discuss the latest findings regarding the mechanisms of liver injury and hepatocarcinogenesis focusing on the role of the stress-activated MAPK pathway. 1. Introduction Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third most common cause of cancer mortality worldwide [1]. The age-adjusted incidence rate of HCC varies geographically, with high rates in East Asia and moderate rates in Europe, North/South America, and Oceania. In addition, a definite increase in HCC incidence has recently been reported in Europe and North America [2]. Although the etiology of background liver disease varies geographically, chronic hepatitis B virus (HBV) or hepatitis C virus (HCV) infection is the main cause of HCC in most areas. Other major etiologies include alcoholic hepatitis, hemochromatosis, and nonalcoholic steatohepatitis (NASH) [1, 3]. Accumulating evidence indicates that a sustained inflammatory reaction in the liver is the major contributing factor to the development of HCC. For example, in chronic hepatitis C, the host immune responses to HCV are often not strong enough to completely clear the infection, resulting in chronic stimulation of an antigen-specific immune response [4]. Hepatocyte damage is induced by the continued expression of cytokines and recruitment of activated inflammatory cells to the liver,

References

[1]  H. B. El-Serag and K. L. Rudolph, “Hepatocellular carcinoma: epidemiology and molecular carcinogenesis,” Gastroenterology, vol. 132, no. 7, pp. 2557–2576, 2007.
[2]  R. Masuzaki, H. Yoshida, R. Tateishi, S. Shiina, and M. Omata, “Hepatocellular carcinoma in viral hepatitis: improving standard therapy,” Best Practice and Research, vol. 22, no. 6, pp. 1137–1151, 2008.
[3]  M. C. Yu and J. M. Yuan, “Environmental factors and risk for hepatocellular carcinoma,” Gastroenterology, vol. 127, pp. S72–S78, 2004.
[4]  A. Budhu and W. W. Xin, “The role of cytokines in hepatocellular carcinoma,” Journal of Leukocyte Biology, vol. 80, no. 6, pp. 1197–1213, 2006.
[5]  M. Levrero, “Viral hepatitis and liver cancer: the case of hepatitis C,” Oncogene, vol. 25, no. 27, pp. 3834–3847, 2006.
[6]  A. M. Elsharkawy and D. A. Mann, “Nuclear factor-κB and the hepatic inflammation-fibrosis-cancer axis,” Hepatology, vol. 46, no. 2, pp. 590–597, 2007.
[7]  H. Nakagawa, S. Maeda, H. Yoshida et al., “Serum IL-6 levels and the risk for hepatocarcinogenesis in chronic hepatitis C patients: an analysis based on gender differences,” International Journal of Cancer, vol. 125, no. 10, pp. 2264–2269, 2009.
[8]  V. Mazzaferro, E. Regalia, R. Doci et al., “Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis,” The New England Journal of Medicine, vol. 334, no. 11, pp. 693–699, 1996.
[9]  J. M. Llovet, M. Schwartz, and V. Mazzaferro, “Resection and liver transplantation for hepatocellular carcinoma,” Seminars in Liver Disease, vol. 25, no. 2, pp. 181–200, 2005.
[10]  R. Tateishi, S. Shiina, T. Teratani et al., “Percutaneous radiofrequency ablation for hepatocellular carcinoma: an analysis of 1000 cases,” Cancer, vol. 103, no. 6, pp. 1201–1209, 2005.
[11]  R. Masuzaki, H. Yoshida, and M. Omata, “Does chemotherapy prevent HCV-related hepatocellular carcinoma? Pros,” Digestive and Liver Disease, vol. 42, supplement 3, pp. 281–286, 2010.
[12]  J. M. Llovet, S. Ricci, V. Mazzaferro et al., “Sorafenib in advanced hepatocellular carcinoma,” The New England Journal of Medicine, vol. 359, no. 4, pp. 378–390, 2008.
[13]  A. Villanueva and J. M. Llovet, “Targeted therapies for hepatocellular carcinoma,” Gastroenterology, vol. 140, no. 5, pp. 1410–1426, 2011.
[14]  H. Yoshida, Y. Shiratori, M. Moriyama, et al., “Interferon therapy reduces the risk for hepatocellular carcinoma: national surveillance program of cirrhotic and noncirrhotic patients with chronic hepatitis C in Japan. IHIT Study Group. Inhibition of Hepatocarcinogenesis by Interferon Therapy,” Annals of Internal Medicine, vol. 131, pp. 174–181, 1999.
[15]  M. W. Fried, M. L. Shiffman, K. Rajender Reddy et al., “Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection,” The New England Journal of Medicine, vol. 347, no. 13, pp. 975–982, 2002.
[16]  T. T. Chang, R. G. Gish, R. De Man et al., “A comparison of entecavir and lamivudine for HBeAg-positive chronic hepatitis B,” The New England Journal of Medicine, vol. 354, no. 10, pp. 1001–1010, 2006.
[17]  J. G. McHutchison, G. T. Everson, S. C. Gordon et al., “Telaprevir with peginterferon and ribavirin for chronic HCV genotype 1 infection,” The New England Journal of Medicine, vol. 360, no. 18, pp. 1827–1838, 2009.
[18]  T. Sakurai, S. Maeda, L. Chang, and M. Karin, “Loss of hepatic NF-κB activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 28, pp. 10544–10551, 2006.
[19]  L. Hui, L. Bakiri, A. Mairhorfer et al., “p38α suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway,” Nature Genetics, vol. 39, no. 6, pp. 741–749, 2007.
[20]  L. Hui, K. Zatloukal, H. Scheuch, E. Stepniak, and E. F. Wagner, “Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation,” The Journal of Clinical Investigation, vol. 118, no. 12, pp. 3943–3953, 2008.
[21]  T. Sakurai, G. He, A. Matsuzawa et al., “Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis,” Cancer Cell, vol. 14, no. 2, pp. 156–165, 2008.
[22]  S. Maeda, “NF-κB, JNK, and TLR signaling pathways in hepatocarcinogenesis,” Gastroenterology Research and Practice, vol. 2010, Article ID 367694, 10 pages, 2010.
[23]  E. F. Wagner and A. R. Nebreda, “Signal integration by JNK and p38 MAPK pathways in cancer development,” Nature Reviews Cancer, vol. 9, no. 8, pp. 537–549, 2009.
[24]  L. Min, B. He, and L. Hui, “Mitogen-activated protein kinases in hepatocellular carcinoma development,” Seminars in Cancer Biology, vol. 21, no. 1, pp. 10–20, 2011.
[25]  J. M. Kyriakis and J. Avruch, “Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation,” Physiological Reviews, vol. 81, no. 2, pp. 807–869, 2001.
[26]  L. Chang and M. Karin, “Mammalian MAP kinase signalling cascades,” Nature, vol. 410, no. 6824, pp. 37–40, 2001.
[27]  R. J. Davis, “Signal transduction by the JNK group of MAP kinases,” Cell, vol. 103, no. 2, pp. 239–252, 2000.
[28]  E. Shaulian and M. Karin, “AP-1 as a regulator of cell life and death,” Nature Cell Biology, vol. 4, no. 5, pp. E131–E136, 2002.
[29]  C. R. Weston and R. J. Davis, “The JNK signal transduction pathway,” Current Opinion in Genetics and Development, vol. 12, no. 1, pp. 14–21, 2002.
[30]  H. M. Shen and Z. G. Liu, “JNK signaling pathway is a key modulator in cell death mediated by reactive oxygen and nitrogen species,” Free Radical Biology and Medicine, vol. 40, no. 6, pp. 928–939, 2006.
[31]  D. Conze, T. Krahl, N. Kennedy et al., “c-Jun NH2-terminal kinase (JNK)1 and JNK2 have distinct roles in CD8+ T cell activation,” Journal of Experimental Medicine, vol. 195, no. 7, pp. 811–823, 2002.
[32]  R. F. Schwabe and D. A. Brenner, “Mechanisms of liver injury. I. TNF-α-induced liver injury: role of IKK, JNK, and ROS pathways,” American Journal of Physiology, vol. 290, no. 4, pp. G583–G589, 2006.
[33]  D. R. Nelson, H. L. Lim, C. G. Marousis et al., “Activation of tumor necrosis factor-α system in chronic hepatitis C virus infection,” Digestive Diseases and Sciences, vol. 42, no. 12, pp. 2487–2494, 1997.
[34]  N. Sheron, J. Lau, H. Daniels et al., “Increased production of tumour necrosis factor alpha in chronic hepatitis B virus infection,” Journal of Hepatology, vol. 12, no. 2, pp. 241–245, 1991.
[35]  G. L. A. Bird, N. Sheron, A. K. J. Goka, G. J. Alexander, and R. S. Williams, “Increased plasma tumor necrosis factor in severe alcoholic hepatitis,” Annals of Internal Medicine, vol. 112, no. 12, pp. 917–920, 1990.
[36]  M. H. Jarrar, A. Baranova, R. Collantes et al., “Adipokines and cytokines in non-alcoholic fatty liver disease,” Alimentary Pharmacology and Therapeutics, vol. 27, no. 5, pp. 412–421, 2008.
[37]  K. R. Feingold, M. Soued, C. Grunfeld, A. H. Moser, J. A. Verdier, and H. G. DoVale, “Tumor necrosis factor stimulates DNA synthesis in the liver of intact rats,” Biochemical and Biophysical Research Communications, vol. 153, no. 2, pp. 576–582, 1988.
[38]  Y. Yamada, I. Kirillova, J. J. Peschon, and N. Fausto, “Initiation of liver growth by tumor necrosis factor: deficient liver regeneration in mice lacking type I tumor necrosis factor receptor,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 4, pp. 1441–1446, 1997.
[39]  A. Wullaert, G. Van Loo, K. Heyninck, and R. Beyaert, “Hepatic tumor necrosis factor signaling and nuclear factor-κB: effects on liver homeostasis and beyond,” Endocrine Reviews, vol. 28, no. 4, pp. 365–386, 2007.
[40]  S. Papa, C. Bubici, F. Zazzeroni, and G. Franzoso, “Mechanisms of liver disease: cross-talk between the NF-κB and JNK pathways,” Biological Chemistry, vol. 390, no. 10, pp. 965–976, 2009.
[41]  O. Micheau and J. Tschopp, “Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes,” Cell, vol. 114, no. 2, pp. 181–190, 2003.
[42]  C. A. Bradham, T. Qian, K. Streetz, C. Trautwein, D. A. Brenner, and J. J. Lemasters, “The mitochondrial permeability transition is required for tumor necrosis factor alpha-mediated apoptosis and cytochrome c release,” Molecular and Cellular Biology, vol. 18, no. 11, pp. 6353–6364, 1998.
[43]  R. F. Schwabe, H. Uchinami, T. Qian, B. L. Bennett, J. J. Lemasters, and D. A. Brenner, “Differential requirement for c-Jun NH2-terminal kinase in TNFalpha- and Fas-mediated apoptosis in hepatocytes.,” The FASEB Journal, vol. 18, no. 6, pp. 720–722, 2004.
[44]  L. Chang, H. Kamata, G. Solinas et al., “The E3 ubiquitin ligase itch couples JNK activation to TNFα-induced cell death by inducing c-FLIPL turnover,” Cell, vol. 124, no. 3, pp. 601–613, 2006.
[45]  S. Maeda, L. Chang, Z. W. Li, J. L. Luo, H. Leffert, and M. Karin, “IKKβ is required for prevention of apoptosis mediated by cell-bound but not by circulating TNFα,” Immunity, vol. 19, no. 5, pp. 725–737, 2003.
[46]  Y. Wang, R. Singh, J. H. Lefkowitch, R. M. Rigoli, and M. J. Czaja, “Tumor necrosis factor-induced toxic liver injury results from JNK2-dependent activation of caspase-8 and the mitochondrial death pathway,” The Journal of Biological Chemistry, vol. 281, no. 22, pp. 15258–15267, 2006.
[47]  H. M. Ni, X. Chen, Y. H. Shi et al., “Genetic delineation of the pathways mediated by bid and JNK in tumor necrosis factor-α-induced liver injury in adult and embryonic mice,” The Journal of Biological Chemistry, vol. 284, no. 7, pp. 4373–4382, 2009.
[48]  M. Takamura, Y. Matsuda, S. Yamagiwa et al., “An inhibitor of c-Jun NH2-terminal kinase, SP600125, protects mice from d-galactosamine/lipopolysaccharide-induced hepatic failure by modulating BH3-only proteins,” Life Sciences, vol. 80, no. 14, pp. 1335–1344, 2007.
[49]  C. Wang, L. Deng, M. Hong, G. R. Akkaraju, J. I. Inoue, and Z. J. Chen, “TAK1 is a ubiquitin-dependent kinase of MKK and IKK,” Nature, vol. 412, no. 6844, pp. 346–351, 2001.
[50]  H. Kamata, S. I. Honda, S. Maeda, L. Chang, H. Hirata, and M. Karin, “Reactive oxygen species promote TNFα-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases,” Cell, vol. 120, no. 5, pp. 649–661, 2005.
[51]  K. Tobiume, A. Matsuzawa, T. Takahashi, et al., “ASK1 is required for sustained activations of JNK/p38 MAP kinases and apoptosis,” EMBO Reports, vol. 2, no. 3, pp. 222–228, 2001.
[52]  M. Saitoh, H. Nishitoh, M. Fujii et al., “Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1,” The EMBO Journal, vol. 17, no. 9, pp. 2596–2606, 1998.
[53]  H. Nakagawa, S. Maeda, Y. Hikiba et al., “Deletion of apoptosis signal-regulating kinase 1 attenuates acetaminophen-induced liver injury by inhibiting c-Jun N-terminal kinase activation,” Gastroenterology, vol. 135, no. 4, pp. 1311–1321, 2008.
[54]  H. Nakagawa, Y. Hirata, K. Takeda et al., “Apoptosis signal-regulating kinase 1 inhibits hepatocarcinogenesis by controlling the tumor-suppressing function of stress-activated mitogen-activated protein kinase,” Hepatology, vol. 54, no. 1, pp. 185–195, 2011.
[55]  H. Okuyama, H. Nakamura, Y. Shimahara et al., “Overexpression of thioredoxin prevents acute hepatitis caused by thioacetamide or lipopolysaccharide in mice,” Hepatology, vol. 37, no. 5, pp. 1015–1025, 2003.
[56]  K. Moriya, K. Nakagawa, T. Santa et al., “Oxidative stress in the absence of inflammation in a mouse model for hepatitis C virus-associated hepatocarcinogenesis,” Cancer Research, vol. 61, no. 11, pp. 4365–4370, 2001.
[57]  H. Kaneto, Y. Nakatani, T. Miyatsuka et al., “Possible novel therapy for diabetes with cell-permeable JNK-inhibitory peptide,” Nature Medicine, vol. 10, no. 10, pp. 1128–1132, 2004.
[58]  H. Nakagawa, A. Isogawa, R. Tateishi, et al., “Serum gamma-glutamyltransferase level is associated with serum superoxide dismutase activity and metabolic syndrome in a Japanese population,” Journal of Gastroenterology, vol. 47, no. 2, pp. 187–194, 2012.
[59]  Y. Deng, X. Ren, L. Yang, et al., “A JNK-dependent pathway is required for TNFalpha-induced apoptosis,” Cell, vol. 115, pp. 61–70, 2003.
[60]  K. Yamamoto, H. Ichijo, and S. J. Korsmeyer, “BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G2/M,” Molecular and Cellular Biology, vol. 19, no. 12, pp. 8469–8478, 1999.
[61]  X. Deng, L. Xiao, W. Lang, et al., “Novel role for JNK as a stress-activated Bcl2 kinase,” The Journal of Biological Chemistry, vol. 276, pp. 23681–23688, 2001.
[62]  M. Fan, M. Goodwin, T. Vu, C. Brantley-Finley, W. A. Gaarde, and T. C. Chambers, “Vinblastine-induced phosphorylation of Bcl-2 and Bcl-X(L) is mediated by JNK and occurs in parallel with inactivation of the Raf-1/MEK/ERK cascade,” The Journal of Biological Chemistry, vol. 275, no. 39, pp. 29980–29985, 2000.
[63]  F. Tsuruta, J. Sunayama, Y. Mori et al., “JNK promotes Bax translocation to mitochondria through phosphorylation of 14-3-3 proteins,” The EMBO Journal, vol. 23, no. 8, pp. 1889–1899, 2004.
[64]  T. Kaufmann, P. J. Jost, M. Pellegrini et al., “Fatal hepatitis mediated by tumor necrosis factor TNFalpha requires caspase-8 and involves the BH3-only proteins Bid and Bim,” Immunity, vol. 30, no. 1, pp. 56–66, 2009.
[65]  K. Lei and R. J. Davis, “JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 5, pp. 2432–2437, 2003.
[66]  R. J. Youle and A. Strasser, “The BCL-2 protein family: opposing activities that mediate cell death,” Nature Reviews Molecular Cell Biology, vol. 9, no. 1, pp. 47–59, 2008.
[67]  A. Hoffmann and D. Baltimore, “Circuitry of nuclear factor κB signaling,” Immunological Reviews, vol. 210, pp. 171–186, 2006.
[68]  Z. Gao, P. Chiao, X. Zhang et al., “Coactivators and corepressors of NF-κB in IκBα gene promoter,” The Journal of Biological Chemistry, vol. 280, no. 22, pp. 21091–21098, 2005.
[69]  T. Luedde, N. Beraza, V. Kotsikoris et al., “Deletion of NEMO/IKKgamma in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma,” Cancer Cell, vol. 11, no. 2, pp. 119–132, 2007.
[70]  E. De Smaele, F. Zazzeroni, S. Papa et al., “Induction of gadd4β by NF-κB downregulates pro-apoptotic JNK signalling,” Nature, vol. 414, no. 6861, pp. 308–313, 2001.
[71]  S. Papa, F. Zazzeroni, C. Bubici et al., “Gadd45β mediates the NF-κB suppression of JNK signalling by targeting MKK7/JNKK2,” Nature Cell Biology, vol. 6, no. 2, pp. 146–153, 2004.
[72]  S. Papa, F. Zazzeroni, Y. X. Fu et al., “Gadd45β promotes hepatocyte survival during liver regeneration in mice by modulating JNK signaling,” The Journal of Clinical Investigation, vol. 118, no. 5, pp. 1911–1923, 2008.
[73]  S. Kaur, F. Wang, M. Venkatraman, and M. Arsura, “X-linked inhibitor of apoptosis (XIAP) inhibits c-Jun N-terminal kinase 1 (JNK1) activation by transforming growth factor β1 (TGF-β1) through ubiquitin-mediated proteosomal degradation of the TGF-β1-activated kinase 1 (TAK1),” The Journal of Biological Chemistry, vol. 280, no. 46, pp. 38599–38608, 2005.
[74]  E. G. Lee, D. L. Boone, S. Chai et al., “Failure to regulate TNF-induced NF-κB and cell death responses in A20-deficient mice,” Science, vol. 289, no. 5488, pp. 2350–2354, 2000.
[75]  U. Lademann, T. Kallunki, and M. J??ttel?, “A20 zinc finger protein inhibits TNF-induced apoptosis and stress response early in the signaling cascades and independently of binding to TRAF2 or 14-3-3 proteins,” Cell Death and Differentiation, vol. 8, no. 3, pp. 265–272, 2001.
[76]  M. B. Arvelo, J. T. Cooper, C. Longo et al., “A20 protects mice from D-galactosamine/lipopolysaccharide acute toxic lethal hepatitis,” Hepatology, vol. 35, no. 3, pp. 535–543, 2002.
[77]  C. G. Pham, C. Bubici, F. Zazzeroni et al., “Ferritin heavy chain upregulation by NF-κB inhibits TNFα-induced apoptosis by suppressing reactive oxygen species,” Cell, vol. 119, no. 4, pp. 529–542, 2004.
[78]  S. Maeda, H. Kamata, J. L. Luo, H. Leffert, and M. Karin, “IKKβ couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis,” Cell, vol. 121, no. 7, pp. 977–990, 2005.
[79]  J. Heinrichsdorff, T. Luedde, E. Perdiguero, A. R. Nebreda, and M. Pasparakis, “p38α MAPK inhibits JNK activation and collaborates with IκB kinase 2 to prevent endotoxin-induced liver failure,” EMBO Reports, vol. 9, no. 10, pp. 1048–1054, 2008.
[80]  P. C. F. Cheung, D. G. Campbell, A. R. Nebreda, and P. Cohen, “Feedback control of the protein kinase TAK1 by SAPK2a/p38α,” The EMBO Journal, vol. 22, no. 21, pp. 5793–5805, 2003.
[81]  H. Muniyappa and K. C. Das, “Activation of c-Jun N-terminal kinase (JNK) by widely used specific p38 MAPK inhibitors SB202190 and SB203580: a MLK-3-MKK7-dependent mechanism,” Cellular Signalling, vol. 20, no. 4, pp. 675–683, 2008.
[82]  S. Inokuchi, T. Aoyama, K. Miura et al., “Disruption of TAK1 in hepatocytes causes hepatic injury, inflammation, fibrosis, and carcinogenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 2, pp. 844–849, 2010.
[83]  K. Bettermann, M. Vucur, J. Haybaeck et al., “TAK1 suppresses a NEMO-dependent but NF-kappaB-independent pathway to liver cancer,” Cancer Cell, vol. 17, no. 5, pp. 481–496, 2010.
[84]  H. Malhi, S. F. Bronk, N. W. Werneburg, and G. J. Gores, “Free fatty acids induce JNK-dependent hepatocyte lipoapoptosis,” The Journal of Biological Chemistry, vol. 281, no. 17, pp. 12093–12101, 2006.
[85]  H. Malhi and G. J. Gores, “Molecular mechanisms of lipotoxicity in nonalcoholic fatty liver disease,” Seminars in Liver Disease, vol. 28, no. 4, pp. 360–369, 2008.
[86]  T. Uehara, B. Bennett, S. T. Sakata et al., “JNK mediates hepatic ischemia reperfusion injury,” Journal of Hepatology, vol. 42, no. 6, pp. 850–859, 2005.
[87]  B. K. Gunawan, Z. X. Liu, D. Han, N. Hanawa, W. A. Gaarde, and N. Kaplowitz, “c-Jun N-terminal kinase plays a major role in murine acetaminophen hepatotoxicity,” Gastroenterology, vol. 131, no. 1, pp. 165–178, 2006.
[88]  K. Sabapathy, K. Hochedlinger, S. Y. Nam, A. Bauer, M. Karin, and E. F. Wagner, “Distinct roles for JNK1 and JNK2 in regulating JNK activity and c-Jun-dependent cell proliferation,” Molecular Cell, vol. 15, no. 5, pp. 713–725, 2004.
[89]  A. Jaeschke, M. Karasarides, J. J. Ventura et al., “JNK2 is a positive regulator of the cJun transcription factor,” Molecular Cell, vol. 23, no. 6, pp. 899–911, 2006.
[90]  R. F. Schwabe, C. A. Bradham, T. Uehara et al., “c-Jun-N-terminal kinase drives cyclin D1 expression and proliferation during liver regeneration,” Hepatology, vol. 37, no. 4, pp. 824–832, 2003.
[91]  Y. Sugioka, T. Watanabe, Y. Inagaki et al., “c-Jun NH2-terminal kinase pathway is involved in constitutive matrix metalloproteinase-1 expression in a hepatocellular carcinoma-derived cell line,” International Journal of Cancer, vol. 109, no. 6, pp. 867–874, 2004.
[92]  C. Cellurale, G. Sabio, N. J. Kennedy et al., “Requirement of c-Jun NH2-terminal kinase for Ras-initiated tumor formation,” Molecular and Cellular Biology, vol. 31, no. 7, pp. 1565–1576, 2011.
[93]  A. S. Nateri, B. Spencer-Dene, and A. Behrens, “Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development,” Nature, vol. 437, no. 7056, pp. 281–285, 2005.
[94]  X. Wu, X. Tu, K. S. Joeng, M. J. Hilton, D. A. Williams, and F. Long, “Rac1 activation controls nuclear localization of beta-catenin during canonical Wnt signaling,” Cell, vol. 133, no. 2, pp. 340–353, 2008.
[95]  R. Sancho, A. S. Nateri, A. G. De Vinuesa et al., “JNK signalling modulates intestinal homeostasis and tumourigenesis in mice,” The EMBO Journal, vol. 28, no. 13, pp. 1843–1854, 2009.
[96]  C. Cellurale, C. R. Weston, J. Reilly et al., “Role of JNK in a Trp53-dependent mouse model of breast cancer,” PLoS One, vol. 5, no. 8, Article ID e12469, 2010.
[97]  D. Schramek, A. Kotsinas, A. Meixner et al., “The stress kinase MKK7 couples oncogenic stress to p53 stability and tumor suppression,” Nature Genetics, vol. 43, no. 3, pp. 212–219, 2011.
[98]  N. K. Saxena, P. P. Fu, A. Nagalingam et al., “Adiponectin modulates C-Jun N-terminal kinase and mammalian target of rapamycin and inhibits hepatocellular carcinoma,” Gastroenterology, vol. 139, no. 5, pp. 1762–e1765, 2010.
[99]  Q. Chang, Y. Zhang, K. J. Beezhold et al., “Sustained JNK1 activation is associated with altered histone H3 methylations in human liver cancer,” Journal of Hepatology, vol. 50, no. 2, pp. 323–333, 2009.
[100]  A. Erhardt, M. Hassan, T. Heintges, and D. H?ussinger, “Hepatitis C virus core protein induces cell proliferation and activates ERK, JNK, and p38 MAP kinases together with the MAP kinase phosphatase MKP-1 in a HepG2 Tet-Off cell line,” Virology, vol. 292, no. 2, pp. 272–284, 2002.
[101]  Y. Tanaka, F. Kanai, T. Ichimura et al., “The hepatitis B virus X protein enhances AP-1 activation through interaction with Jab1,” Oncogene, vol. 25, no. 4, pp. 633–642, 2006.
[102]  T. Tsutsumi, T. Suzuki, K. Moriya et al., “Alteration of intrahepatic cytokine expression and AP-1 activation in transgenic mice expressing hepatitis C virus core protein,” Virology, vol. 304, no. 2, pp. 415–424, 2002.
[103]  M. Murata, K. Matsuzaki, K. Yoshida et al., “Hepatitis B virus X protein shifts human hepatic transforming growth factor (TGF)-β signaling from tumor suppression to oncogenesis in early chronic hepatitis B,” Hepatology, vol. 49, no. 4, pp. 1203–1217, 2009.
[104]  M. Das, D. S. Garlick, D. L. Greiner, and R. J. Davis, “The role of JNK in the development of hepatocellular carcinoma,” Genes and Development, vol. 25, no. 6, pp. 634–645, 2011.
[105]  J. Hirosumi, G. Tuncman, L. Chang et al., “A central, role for JNK in obesity and insulin resistance,” Nature, vol. 420, no. 6913, pp. 333–336, 2002.
[106]  G. Sabio, M. Das, A. Mora et al., “A stress signaling pathway in adipose tissue regulates hepatic insulin resistance,” Science, vol. 322, no. 5907, pp. 1539–1543, 2008.
[107]  T. Ohki, R. Tateishi, S. Shiina et al., “Visceral fat accumulation is an independent risk factor for hepatocellular carcinoma recurrence after curative treatment in patients with suspected NASH,” Gut, vol. 58, no. 6, pp. 839–844, 2009.
[108]  F. Marra and C. Bertolani, “Adipokines in liver diseases,” Hepatology, vol. 50, no. 3, pp. 957–969, 2009.
[109]  E. J. Park, J. H. Lee, G. Y. Yu et al., “Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression,” Cell, vol. 140, no. 2, pp. 197–208, 2010.
[110]  T. Arano, H. Nakagawa, R. Tateishi et al., “Serum level of adiponectin and the risk of liver cancer development in chronic hepatitis C patients,” International Journal of Cancer, vol. 129, no. 9, pp. 2226–2235, 2011.
[111]  J. Han and P. Sun, “The pathways to tumor suppression via route p38,” Trends in Biochemical Sciences, vol. 32, no. 8, pp. 364–371, 2007.
[112]  B. Ge, H. Gram, F. Di Padova et al., “MAPKK-independent activation of p38α mediated by TAB1-dependent autophosphorylation of p38α,” Science, vol. 295, no. 5558, pp. 1291–1294, 2002.
[113]  L. Hui, L. Bakiri, E. Stepniak, and E. F. Wagner, “p38α: a suppressor of cell proliferation and tumorigenesis,” Cell Cycle, vol. 6, no. 20, pp. 2429–2433, 2007.
[114]  T. M. Thornton and M. Rincon, “Non-classical p38 map kinase functions: cell cycle checkpoints and survival,” International Journal of Biological Sciences, vol. 5, no. 1, pp. 44–52, 2009.
[115]  E. Stepniak, R. Ricci, R. Eferl et al., “c-Jun/AP-1 controls liver regeneration by repressing p53/p21 and p38 MAPK activity,” Genes and Development, vol. 20, no. 16, pp. 2306–2314, 2006.
[116]  J. S. Campbell, G. M. Argast, S. Y. Yuen, B. Hayes, and N. Fausto, “Inactivation of p38 MAPK during liver regeneration,” International Journal of Biochemistry and Cell Biology, vol. 43, no. 2, pp. 180–188, 2011.
[117]  I. Dolado, A. Swat, N. Ajenjo, G. De Vita, A. Cuadrado, and A. R. Nebreda, “p38alpha MAP kinase as a sensor of reactive oxygen species in tumorigenesis,” Cancer Cell, vol. 11, no. 2, pp. 191–205, 2007.
[118]  P. Sun, N. Yoshizuka, L. New et al., “PRAK is essential for ras-induced senescence and tumor suppression,” Cell, vol. 128, no. 2, pp. 295–308, 2007.
[119]  D. V. Bulavin, C. Phillips, B. Nannenga et al., “Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16Ink4a-p19 Arf pathway,” Nature Genetics, vol. 36, no. 4, pp. 343–350, 2004.
[120]  Y. H. Hsieh, T. T. Wu, C. Y. Huang, Y. S. Hsieh, J. M. Hwang, and J. Y. Liu, “p38 mitogen-activated protein kinase pathway is involved in protein kinase Cα-regulated invasion in human hepatocellular carcinoma cells,” Cancer Research, vol. 67, no. 9, pp. 4320–4327, 2007.
[121]  M. S. Kim, E. J. Lee, H. R. C. Kim, and A. Moon, “p38 kinase is a key signaling molecule for H-ras-induced cell motility and invasive phenotype in human breast epithelial cells,” Cancer Research, vol. 63, no. 17, pp. 5454–5461, 2003.
[122]  K. Iyoda, Y. Sasaki, M. Horimoto et al., “Involvement of the p38 mitogen-activated protein kinase cascade in hepatocellular carcinoma,” Cancer, vol. 97, no. 12, pp. 3017–3026, 2003.
[123]  H. Ichijo, E. Nishida, K. Irie et al., “Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways,” Science, vol. 275, no. 5296, pp. 90–94, 1997.
[124]  H. Huynh, S. P. Choo, H. C. Toh, et al., “Comparing the efficacy of sunitinib with sorafenib in xenograft models of human hepatocellular carcinoma: mechanistic explanation,” Current Cancer Drug Targets, vol. 11, no. 8, pp. 944–953, 2011.
[125]  C. Runchel, A. Matsuzawa, and H. Ichijo, “Mitogen-activated protein kinases in mammalian oxidative stress responses,” Antioxidants and Redox Signaling, vol. 15, no. 1, pp. 205–218, 2011.
[126]  G. R. Fanger, N. L. Johnson, and G. L. Johnson, “MEK kinases are regulated by EGF and selectively interact with Rac/Cdc42,” The EMBO Journal, vol. 16, no. 16, pp. 4961–4972, 1997.
[127]  D. N. Chadee and J. M. Kyriakis, “MLK3 is required for mitogen activation of B-Raf, ERK and cell proliferation,” Nature Cell Biology, vol. 6, no. 8, pp. 770–776, 2004.
[128]  E. Pikarsky, R. M. Porat, I. Stein et al., “NF-κB functions as a tumour promoter in inflammation-associated cancer,” Nature, vol. 431, no. 7007, pp. 461–466, 2004.
[129]  R. J. Mayer and J. F. Callahan, “p38 MAP kinase inhibitors: a future therapy for inflammatory diseases,” Drug Discovery Today, vol. 3, no. 1, pp. 49–54, 2006.
[130]  S. Kumar, J. Boehm, and J. C. Lee, “P38 MAP kinases: key signalling molecules as therapeutic targets for inflammatory diseases,” Nature Reviews Drug Discovery, vol. 2, no. 9, pp. 717–726, 2003.
[131]  S. Hagiwara, M. Kudo, H. Chung, et al., “Activation of c-Jun N-terminal kinase in non-cancerous liver tissue predicts a high risk of recurrence after hepatic resection for hepatocellular carcinoma,” Hepatology Research, vol. 42, no. 4, pp. 394–400, 2012.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133