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...

NADPH Oxidases in Chronic Liver Diseases

DOI: 10.1155/2014/742931

Full-Text   Cite this paper   Add to My Lib

Abstract:

Oxidative stress is a common feature observed in a wide spectrum of chronic liver diseases including viral hepatitis, alcoholic, and nonalcoholic steatohepatitis. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are emerging as major sources of reactive oxygen species (ROS). Several major isoforms are expressed in the liver, including NOX1, NOX2, and NOX4. While the phagocytic NOX2 has been known to play an important role in Kupffer cell and neutrophil phagocytic activity and inflammation, the nonphagocytic NOX homologues are increasingly recognized as key enzymes in oxidative injury and wound healing. In this review, we will summarize the current advances in knowledge on the regulatory pathways of NOX activation, their cellular distribution, and their role in the modulation of redox signaling in liver diseases. 1. Introduction In chronic liver diseases, hepatocyte injury triggers Kupffer cell activation and hepatic stellate cell (HSC) transdifferentiation to matrix-producing myofibroblasts, and the accumulation of extracellular matrix leads to fibrosis and cirrhosis [1, 2]. Most if not all pathogenic insults in the liver can cause oxidative stress, inducing lipid peroxidation, protein oxidation, and DNA damage, leading to hepatocyte mitochondrial dysfunction, amplifying inflammation and initiating fibrosis [3]. As important second messengers, ROS can have an impact on cell death/survival pathways [4]. In the liver there are several important sources of ROS production. While the role of cytochrome P4502E1 (CYP2E1), the mitochondrial respiratory chain, arachidonic acid oxidation, and the xanthine oxidase system have been extensively studied in the past [5–7], recently the group of NOX enzymes have been emerging as major sources of ROS production. H2O2 is one of the main oxidative radicals during liver diseases and it is generated by the NADPH oxidases or complex III of the mitochondrial respiratory chain. In physiological situations the amount of H2O2 is under a tight control by the peroxiredoxins and glutathione peroxidases as well as by catalase [8]. During chronic liver injury however this balance is perturbed and parenchymal cells are exposed to increasing concentrations of ROS. Among the seven NOX homologues found in mammals (NOX1, NOX2, NOX3, NOX4, NOX5, DUOX1, and DUOX2), the main ROS-producing NOXs in the liver are NOX1, NOX2, and NOX4 [9] (Table 1). NOX1 and 2 are mainly producing superoxide whereas NOX4 directly produces H2O2. To form active complexes, NOX1 and NOX2 bind with their structural subunits: NOX1 associates

References

[1]  Y. Lee and S. L. Friedman, “Fibrosis in the liver: acute protection and chronic disease,” Progress in Molecular Biology and Translational Science, vol. 97, pp. 151–200, 2010.
[2]  J. X. Jiang and N. J. Torok, “Liver injury and the activation of the hepatic myofibroblasts,” Current Pathobiology Reports, vol. 1, pp. 215–223, 2013.
[3]  V. Sánchez-Valle, N. C. Chávez-Tapia, M. Uribe, and N. Méndez-Sánchez, “Role of oxidative stress and molecular changes in liver fibrosis: a review,” Current Medicinal Chemistry, vol. 19, no. 28, pp. 4850–4860, 2012.
[4]  J. X. Jiang, K. Mikami, S. Venugopal, Y. Li, and N. J. T?r?k, “Apoptotic body engulfment by hepatic stellate cells promotes their survival by the JAK/STAT and Akt/NF-κB-dependent pathways,” Journal of Hepatology, vol. 51, no. 1, pp. 139–148, 2009.
[5]  H. Jaeschke, “Reactive oxygen and mechanisms of inflammatory liver injury: present concepts,” Journal of Gastroenterology and Hepatology, vol. 26, supplement 1, pp. 173–179, 2011.
[6]  C. S. Lieber, “Role of oxidative stress and antioxidant therapy in alcoholic and nonalcoholic liver diseases,” Advances in Pharmacology, vol. 38, pp. 601–628, 1997.
[7]  A. I. Cederbaum, “Iron and CYP2E1-dependent oxidative stress and toxicity,” Alcohol, vol. 30, no. 2, pp. 115–120, 2003.
[8]  H. Sies, “Role of metabolic H2O2 generation: redox signaling and oxidative stress,” Journal of Biological Chemistry, vol. 289, no. 13, pp. 8735–8741, 2014.
[9]  Y.-H. Paik, J. Kim, T. Aoyama, S. de Minicis, R. Bataller, and D. A. Brenner, “Role of NADPH oxidases in liver fibrosis,” Antioxidants & Redox Signaling, vol. 20, no. 17, pp. 2854–2872, 2014.
[10]  J. D. Lambeth, “NOX enzymes and the biology of reactive oxygen,” Nature Reviews Immunology, vol. 4, no. 3, pp. 181–189, 2004.
[11]  A. Mizrahi, Y. Berdichevsky, Y. Ugolev et al., “Assembly of the phagocyte NADPH oxidase complex: chimeric constructs derived from the cytosolic components as tools for exploring structure-function relationships,” Journal of Leukocyte Biology, vol. 79, no. 5, pp. 881–895, 2006.
[12]  J. I. Cohen, X. Chen, and L. E. Nagy, “Redox signaling and the innate immune system in alcoholic liver disease,” Antioxidants and Redox Signaling, vol. 15, no. 2, pp. 523–534, 2011.
[13]  S. C. Lu, “Glutathione synthesis,” Biochimica et Biophysica Acta, vol. 1830, no. 5, pp. 3143–3153, 2013.
[14]  L. Yuan and N. Kaplowitz, “Glutathione in liver diseases and hepatotoxicity,” Molecular Aspects of Medicine, vol. 30, no. 1-2, pp. 29–41, 2009.
[15]  T. W. Kensler, N. Wakabayashi, and S. Biswal, “Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway,” Annual Review of Pharmacology and Toxicology, vol. 47, pp. 89–116, 2007.
[16]  G. J. Kops, T. B. Dansen, P. E. Polderman, et al., “Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress,” Nature, vol. 419, no. 6904, pp. 316–321, 2002.
[17]  A. I. Malik and K. B. Storey, “Transcriptional regulation of antioxidant enzymes by FoxO1 under dehydration stress,” Gene, vol. 485, no. 2, pp. 114–119, 2011.
[18]  Y. H. Paik, K. Iwaisako, E. Seki et al., “The nicotinamide adenine dinucleotide phosphate oxidase (NOX) homologues NOX1 and NOX2/gp91(phox) mediate hepatic fibrosis in mice,” Hepatology, vol. 53, no. 5, pp. 1730–1741, 2011.
[19]  W. Cui, K. Matsuno, K. Iwata et al., “NOX1/nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) oxidase promotes proliferation of stellate cells and aggravates liver fibrosis induced by bile duct ligation,” Hepatology, vol. 54, no. 3, pp. 949–958, 2011.
[20]  J. X. Jiang, S. Venugopal, N. Serizawa et al., “Reduced nicotinamide adenine dinucleotide phosphate oxidase 2 plays a key role in stellate cell activation and liver fibrogenesis in vivo,” Gastroenterology, vol. 139, no. 4, pp. 1375.e4–1384.e4, 2010.
[21]  H. E. Boudreau, S. U. Emerson, A. Korzeniowska, M. A. Jendrysik, and T. L. Leto, “Hepatitis C virus (HCV) proteins induce NADPH oxidase 4 expression in a transforming growth factor β-dependent manner: a new contributor to HCV-induced oxidative stress,” Journal of Virology, vol. 83, no. 24, pp. 12934–12946, 2009.
[22]  Q. Liu, H. Li, N. Wang et al., “Polymorphism of rs1836882 in NOX4 gene modifies associations between dietary caloric intake and ROS levels in peripheral blood mononuclear cells,” PLoS ONE, vol. 8, no. 12, Article ID e85660, 2013.
[23]  H. Vlassara and J. Uribarri, “Advanced glycation end products (AGE) and diabetes: cause, effect, or both?” Current Diabetes Reports, vol. 14, no. 1, article 453, 2014.
[24]  J. X. Jiang, X. Chen, H. Fukada, N. Serizawa, S. Devaraj, and N. J. T?r?k, “Advanced glycation endproducts induce fibrogenic activity in nonalcoholic steatohepatitis by modulating TNF-α-converting enzyme activity in mice,” Hepatology, vol. 58, no. 4, pp. 1339–1348, 2013.
[25]  S. de Minicis, E. Seki, Y.-H. Paik et al., “Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis,” Hepatology, vol. 52, no. 4, pp. 1420–1430, 2010.
[26]  R. S. Tacke, H.-C. Lee, C. Goh et al., “Myeloid suppressor cells induced by hepatitis C virus suppress T-cell responses through the production of reactive oxygen species,” Hepatology, vol. 55, no. 2, pp. 343–353, 2012.
[27]  R. Reinehr, S. Becker, A. Eberle, S. Grether-Beck, and D. H?ussinger, “Involvement of NADPH oxidase isoforms and Src family kinases in CD95-dependent hepatocyte apoptosis,” The Journal of Biological Chemistry, vol. 280, no. 29, pp. 27179–27194, 2005.
[28]  P. Sancho, J. Mainez, E. Crosas-Molist et al., “NADPH oxidase NOX4 mediates stellate cell activation and hepatocyte cell death during liver fibrosis development,” PLoS ONE, vol. 7, no. 9, Article ID e45285, 2012.
[29]  J. X. Jiang, X. Chen, N. Serizawa et al., “Liver fibrosis and hepatocyte apoptosis are attenuated by GKT137831, a novel NOX4/NOX1 inhibitor in vivo,” Free Radical Biology and Medicine, vol. 53, no. 2, pp. 289–296, 2012.
[30]  K.-H. Kim, C.-C. Chen, R. I. Monzon, and L. F. Lau, “Matricellular protein CCN1 promotes regression of liver fibrosis through induction of cellular senescence in hepatic myofibroblasts,” Molecular and Cellular Biology, vol. 33, no. 10, pp. 2078–2090, 2013.
[31]  C. Bureau, J. Bernad, N. Chaouche et al., “Nonstructural 3 protein of hepatitis C virus triggers an oxidative burst in human monocytes via activation of NADPH oxidase,” Journal of Biological Chemistry, vol. 276, no. 25, pp. 23077–23083, 2001.
[32]  F. Thorén, A. Romero, M. Lindh, C. Dahlgren, and K. Hellstrand, “A hepatitis C virus-encoded, nonstructural protein (NS3) triggers dysfunction and apoptosis in lymphocytes: role of NADPH oxidase-derived oxygen radicals,” Journal of Leukocyte Biology, vol. 76, no. 6, pp. 1180–1186, 2004.
[33]  N. S. R. de Mochel, S. Seronello, S. H. Wang et al., “Hepatocyte NAD(P)H oxidases as an endogenous source of reactive oxygen species during hepatitis C virus infection,” Hepatology, vol. 52, no. 1, pp. 47–59, 2010.
[34]  J. Colmenero, R. Bataller, P. Sancho-Bru, et al., “Effects of losartan on hepatic expression of nonphagocytic NADPH oxidase and fibrogenic genes in patients with chronic hepatitis C,” American Journal of Physiology—Gastrointestinal and Liver Physiology, vol. 297, no. 4, pp. G726–G734, 2009.
[35]  S. De Minicis and D. A. Brenner, “Oxidative stress in alcoholic liver disease: role of NADPH oxidase complex,” Journal of Gastroenterology and Hepatology, vol. 23, no. 1, pp. S98–S103, 2008.
[36]  H. Kono, I. Rusyn, M. Yin et al., “NADPH oxidase-derived free radicals are key oxidants in alcohol-induced liver disease,” The Journal of Clinical Investigation, vol. 106, no. 7, pp. 867–872, 2000.
[37]  M. D. Wheeler, H. Kono, M. Yin et al., “The role of kupffer cell oxidant production in early ethanol-induced liver disease,” Free Radical Biology and Medicine, vol. 31, no. 12, pp. 1544–1549, 2001.
[38]  F. J. Cubero and N. Nieto, “Ethanol and arachidonic acid synergize to activate Kupffer cells and modulate the fibrogenic response via tumor necrosis factor α, reduced glutathione, and transforming growth factor β-dependent mechanisms,” Hepatology, vol. 48, no. 6, pp. 2027–2039, 2008.
[39]  V. Thakur, M. T. Pritchard, M. R. McMullen, Q. Wang, and L. E. Nagy, “Chronic ethanol feeding increases activation of NADPH oxidase by lipopolysaccharide in rat Kupffer cells: Role of increased reactive oxygen in LPS-stimulated ERK1/2 activation and TNF-α production,” Journal of Leukocyte Biology, vol. 79, no. 6, pp. 1348–1356, 2006.
[40]  I. Levin, J. Petrasek, and G. Szabo, “The presence of p47phox in liver parenchymal cells is a key mediator in the pathogenesis of alcoholic liver steatosis,” Alcoholism: Clinical and Experimental Research, vol. 36, no. 8, pp. 1397–1406, 2012.
[41]  S. Yeligar, H. Tsukamoto, and V. K. Kalra, “Ethanol-induced expression of ET-1 and ET-BR in liver sinusoidal endothelial cells and human endothelial cells involves hypoxia-inducible factor-1α and microRNA-199,” The Journal of Immunology, vol. 183, no. 8, pp. 5232–5243, 2009.
[42]  A. E. Feldstein and S. M. Bailey, “Emerging role of redox dysregulation in alcoholic and nonalcoholic fatty liver disease,” Antioxidants and Redox Signaling, vol. 15, no. 2, pp. 421–424, 2011.
[43]  C. Guichard, R. Moreau, D. Pessayre, T. K. Epperson, and K.-H. Krause, “NOX family NADPH oxidases in liver and in pancreatic islets: a role in the metabolic syndrome and diabetes?” Biochemical Society Transactions, vol. 36, no. 5, pp. 920–929, 2008.
[44]  T. Adachi, H. Togashi, A. Suzuki et al., “NAD(P)H oxidase plays a crucial role in PDGF-induced proliferation of hepatic stellate cells,” Hepatology, vol. 41, no. 6, pp. 1272–1281, 2005.
[45]  R. Sugimoto, M. Enjoji, M. Kohjima et al., “High glucose stimulates hepatic stellate cells to proliferate and to produce collagen through free radical production and activation of mitogen-activated protein kinase,” Liver International, vol. 25, no. 5, pp. 1018–1026, 2005.
[46]  M. Carmiel-Haggai, A. I. Cederbaum, and N. Nieto, “A high-fat diet leads to the progression of non-alcoholic fatty liver disease in obese rats,” FASEB Journal, vol. 19, no. 1, pp. 136–138, 2005.
[47]  S. Chatterjee, D. Ganini, E. J. Tokar, et al., “Leptin is key to peroxynitrite-mediated oxidative stress and Kupffer cell activation in experimental non-alcoholic steatohepatitis,” Journal of Hepatology, vol. 58, no. 4, pp. 778–784, 2013.
[48]  J. X. Jiang, K. Mikami, V. H. Shah, and N. J. Torok, “Leptin induces phagocytosis of apoptotic bodies by hepatic stellate cells via a Rho guanosine triphosphatase-dependent mechanism,” Hepatology, vol. 48, no. 5, pp. 1497–1505, 2008.
[49]  S. de Minicis, E. Seki, C. Oesterreicher, B. Schnabl, R. F. Schwabe, and D. A. Brenner, “Reduced nicotinamide adenine dinucleotide phosphate oxidase mediates fibrotic and inflammatory effects of leptin on hepatic stellate cells,” Hepatology, vol. 48, no. 6, pp. 2016–2026, 2008.
[50]  I. García-Ruiz, E. Gómez-Izquierdo, T. Díaz-Sanjuán et al., “Sp1 and Sp3 transcription factors mediate leptin-induced collagen α1(I) gene expression in primary culture of male rat hepatic stellate cells,” Endocrinology, vol. 153, no. 12, pp. 5845–5856, 2012.
[51]  H. Vlassara and G. E. Striker, “AGE restriction in diabetes mellitus: a paradigm shift,” Nature Reviews Endocrinology, vol. 7, no. 9, pp. 526–539, 2011.
[52]  A. dela Pe?a, I. A. Leclercq, J. Williams, and G. C. Farrell, “NADPH oxidase is not an essential mediator of oxidative stress or liver injury in murine MCD diet-induced steatohepatitis,” Journal of Hepatology, vol. 46, no. 2, pp. 304–313, 2007.
[53]  A. E. Feldstein, A. Canbay, P. Angulo et al., “Hepatocyte apoptosis and fas expression are prominent features of human nonalcoholic steatohepatitis,” Gastroenterology, vol. 125, no. 2, pp. 437–443, 2003.
[54]  J. H. Yu, B.-M. Zhu, G. Riedlinger, K. Kang, and L. Hennighausen, “The liver-specific tumor suppressor STAT5 controls expression of the reactive oxygen species-generating enzyme NOX4 and the proapoptotic proteins PUMA and BIM in mice,” Hepatology, vol. 56, no. 6, pp. 2375–2386, 2012.
[55]  Y.-H. Paik, J. Kim, T. Aoyama, S. De Minicis, R. Bataller, and D. A. Brenner, “Role of NADPH oxidases in liver fibrosis,” Antioxidants and Redox Signaling, vol. 20, no. 17, pp. 2854–2872, 2014.
[56]  R. Bataller, R. F. Schwabe, Y. H. Choi et al., “NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic fibrosis,” Journal of Clinical Investigation, vol. 112, no. 9, pp. 1383–1394, 2003.
[57]  R. K. Ambasta, P. Kumar, K. K. Griendling, H. H. H. W. Schmidt, R. Busse, and R. P. Brandes, “Direct interaction of the novel Nox proteins with p22phox is required for the formation of a functionally active NADPH oxidase,” Journal of Biological Chemistry, vol. 279, no. 44, pp. 45935–45941, 2004.
[58]  I. Carmona-Cuenca, C. Roncero, P. Sancho et al., “Upregulation of the NADPH oxidase NOX4 by TGF-beta in hepatocytes is required for its pro-apoptotic activity,” Journal of Hepatology, vol. 49, no. 6, pp. 965–976, 2008.
[59]  T. Aoyama, Y. H. Paik, S. Watanabe et al., “Nicotinamide adenine dinucleotide phosphate oxidase in experimental liver fibrosis: GKT137831 as a novel potential therapeutic agent,” Hepatology, vol. 56, no. 6, pp. 2316–2327, 2012.
[60]  S. Senturk, M. Mumcuoglu, O. Gursoy-Yuzugullu, B. Cingoz, K. C. Akcali, and M. Ozturk, “Transforming growth factor-beta induces senescence in hepatocellular carcinoma cells and inhibits tumor growth,” Hepatology, vol. 52, no. 3, pp. 966–974, 2010.
[61]  E. Crosas-Molist, E. Bertran, P. Sancho, et al., “The NADPH oxidase NOX4 inhibits hepatocyte proliferation and liver cancer progression,” Free Radical Biology & Medicine, vol. 69, pp. 338–347, 2014.
[62]  P. Sancho and I. Fabregat, “NADPH oxidase NOX1 controls autocrine growth of liver tumor cells through up-regulation of the epidermal growth factor receptor pathway,” Journal of Biological Chemistry, vol. 285, no. 32, pp. 24815–24824, 2010.
[63]  M. Daugaard, R. Nitsch, B. Razaghi et al., “Hace1 controls ROS generation of vertebrate Rac1-dependent NADPH oxidase complexes,” Nature Communications, vol. 4, article 2180, 2013.
[64]  C. D. Klaassen and S. A. Reisman, “Nrf2 the rescue: effects of the antioxidative/electrophilic response on the liver,” Toxicology and Applied Pharmacology, vol. 244, no. 1, pp. 57–65, 2010.
[65]  S. M. Shin, J. H. Yang, and S. H. Ki, “Role of the Nrf2-are pathway in liver diseases,” Oxidative Medicine and Cellular Longevity, vol. 2013, Article ID 763257, 9 pages, 2013.
[66]  S. Schaedler, J. Krause, K. Himmelsbach, et al., “Hepatitis B virus induces expression of antioxidant response element-regulated genes by activation of Nrf2,” The Journal of Biological Chemistry, vol. 285, no. 52, pp. 41074–41086, 2010.
[67]  D. Burdette, M. Olivarez, and G. Waris, “Activation of transcription factor Nrf2 by hepatitis C virus induces the cell-survival pathway,” Journal of General Virology, vol. 91, no. 3, pp. 681–690, 2010.
[68]  K. C. Wu, J. Liu, and C. D. Klaassen, “Role of Nrf2 in preventing ethanol-induced oxidative stress and lipid accumulation,” Toxicology and Applied Pharmacology, vol. 262, no. 3, pp. 321–329, 2012.
[69]  J. Lamlé, S. Marhenke, J. Borlak, et al., “Nuclear factor-eythroid 2-related factor 2 prevents alcohol-induced fulminant liver injury,” Gastroenterology, vol. 134, no. 4, pp. 1159.e2–1168.e2, 2008.
[70]  J. Liu, K. C. Wu, Y.-F. Lu, E. Ekuase, and C. D. Klaassen, “NRF2 protection against liver injury produced by various hepatotoxicants,” Oxidative Medicine and Cellular Longevity, vol. 2013, Article ID 305861, 8 pages, 2013.
[71]  H. Sugimoto, K. Okada, J. Shoda et al., “Deletion of nuclear factor-E2-related factor-2 leads to rapid onset and progression of nutritional steatohepatitis in mice,” American Journal of Physiology: Gastrointestinal and Liver Physiology, vol. 298, no. 2, pp. G283–G294, 2010.
[72]  S. Chowdhry, M. H. Nazmy, P. J. Meakin et al., “Loss of Nrf2 markedly exacerbates nonalcoholic steatohepatitis,” Free Radical Biology and Medicine, vol. 48, no. 2, pp. 357–371, 2010.
[73]  R. Shimozono, Y. Asaoka, Y. Yoshizawa, et al., “Nrf2 activators attenuate the progression of nonalcoholic steatohepatitis-related fibrosis in a dietary rat model,” Molecular Pharmacology, vol. 84, no. 1, pp. 62–70, 2013.
[74]  U. A. Kohler, S. Kurinna, D. Schwitter, et al., “Activated Nrf2 impairs liver regeneration in mice by activation of genes involved in cell cycle control and apoptosis,” Hepatology, vol. 60, no. 2, pp. 670–678, 2013.
[75]  L. Hecker, N. J. Logsdon, D. Kurundkar et al., “Reversal of persistent fibrosis in aging by targeting Nox4-Nrf2 redox imbalance,” Science Translational Medicine, vol. 6, Article ID 231ra247, 2014.
[76]  I. Tikhanovich, J. Cox, and S. A. Weinman, “Forkhead box class O transcription factors in liver function and disease,” Journal of Gastroenterology and Hepatology, vol. 28, no. 1, pp. 125–131, 2013.
[77]  D. R. Calnan and A. Brunet, “The FoxO code,” Oncogene, vol. 27, no. 16, pp. 2276–2288, 2008.
[78]  S. Nemoto and T. Finkel, “Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway,” Science, vol. 295, no. 5564, pp. 2450–2452, 2002.
[79]  M. Adachi, Y. Osawa, H. Uchinami, T. Kitamura, D. Accili, and D. A. Brenner, “The forkhead transcription factor FoxO1 regulates proliferation and transdifferentiation of hepatic stellate cells,” Gastroenterology, vol. 132, no. 4, pp. 1434–1446, 2007.
[80]  M. Matsumoto, S. Han, T. Kitamura, and D. Accili, “Dual role of transcription factor FoxO1 in controlling hepatic insulin sensitivity and lipid metabolism,” The Journal of Clinical Investigation, vol. 116, no. 9, pp. 2464–2472, 2006.
[81]  V. S. Calvert, R. Collantes, H. Elariny et al., “A systems biology approach to the pathogenesis of obesity-related nonalcoholic fatty liver disease using reverse phase protein microarrays for multiplexed cell signaling analysis,” Hepatology, vol. 46, no. 1, pp. 166–172, 2007.
[82]  L. Deng, I. Shoji, W. Ogawa et al., “Hepatitis C virus infection promotes hepatic gluconeogenesis through an NS5A-mediated, FoxO1-dependent pathway,” Journal of Virology, vol. 85, no. 17, pp. 8556–8568, 2011.
[83]  G. Z. Tao, N. Lehwald, K. Y. Jang et al., “Wnt/β-catenin signaling protects mouse liver against oxidative stress-induced apoptosis through the inhibition of forkhead transcription factor FoxO3,” Journal of Biological Chemistry, vol. 288, no. 24, pp. 17214–17224, 2013.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413