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...

In Vivo Effect of Arsenic Trioxide on Keap1-p62-Nrf2 Signaling Pathway in Mouse Liver: Expression of Antioxidant Responsive Element-Driven Genes Related to Glutathione Metabolism

DOI: 10.1155/2013/817693

Full-Text   Cite this paper   Add to My Lib

Abstract:

Arsenic is a Group I human carcinogen, and chronic arsenic exposure through drinking water is a major threat to human population. Liver is one of the major organs for the detoxification of arsenic. The present study was carried out in mice in vivo after arsenic treatment through drinking water at different doses and time of exposure. Arsenic toxicity is found to be mediated by reactive oxygen species. Nuclear factor (erythroid-2 related) factor 2 (Nrf2)/Keap1 (Kelch-like ECH-associated protein 1)/ARE (antioxidant response element)—driven target gene system protects cells against oxidative stress and maintains cellular oxidative homeostasis. Our result showed 0.4?ppm, 2?ppm, and 4?ppm arsenic trioxide treatment through drinking water for 30 days and 90 days induced damages in the liver of Swiss albino mice as evidenced by histopathology, disturbances in liver function, induction of heat shock protein 70, modulation of trace elements, alteration in reduced glutathione level, glutathione-s-transferase and catalase activity, malondialdehyde production, and induction of apoptosis. Cellular Nrf2 protein level and mRNA level increased in all treatment groups. Keap1 protein as well as mRNA level decreased concomitantly in arsenic treated mice. Our study clearly indicates the important role of Nrf2 in activating ARE driven genes related to GSH metabolic pathway and also the adaptive response mechanisms in arsenic induced hepatotoxicity. 1. Introduction Arsenic (As), a Group I human carcinogen, is the major source of ground water contamination all over the world. The permissible limit of As, set by World Health Organization (WHO) is 10 parts per billion (ppb). However, in many countries including India and Bangladesh, people are consuming As through drinking water at much higher level. Up to 50?ppm of As is reported in many states in the USA [1]. Fu et al. [2] estimated that 13 million Americans were exposed to more than 0.01?ppm of arsenic through public water systems by 2006. According to the report of the Prevention and Treatment Academy of China, this number reached 14.66 million in China [3] where in many places individuals were exposed up to a level of 1?ppm of As [4]. In West Bengal, India As concentrations in some tube wells is as high as 3.4?ppm [5]. Chronic arsenic exposure has become a great concern than acute exposure mainly because of its carcinogenic effects [6, 7]. Environmental exposure to arsenic is generally in the form of either arsenite (As3+) or arsenate (As5+) which undergoes redox conversion, where arsenite is the predominant form in

References

[1]  P. B. Tchounwou, A. K. Patlolla, and J. A. Centeno, “Carcinogenic and systemic health effects associated with arsenic exposure: a critical review,” Toxicologic Pathology, vol. 31, no. 6, pp. 575–588, 2003.
[2]  J. Fu, C. G. Woods, E. Yehuda-Shnaidman et al., “Low-level arsenic impairs glucose-stimulated insulin secretion in pancreatic beta cells: involvement of cellular adaptive response to oxidative stress,” Environmental Health Perspectives, vol. 118, no. 6, pp. 864–870, 2010.
[3]  G. Sun, X. Li, J. Pi et al., “Current research problems of chronic arsenicosis in China,” Journal of Health, Population and Nutrition, vol. 24, no. 2, pp. 176–181, 2006.
[4]  G. Sun, Y. Xu, X. Li, Y. Jin, B. Li, and X. Sun, “Urinary arsenic metabolites in children and adults exposed to arsenic in drinking water in inner Mongolia, China,” Environmental Health Perspectives, vol. 115, no. 4, pp. 648–652, 2007.
[5]  D. N. G. Mazumder, R. Haque, N. Ghosh et al., “Arsenic levels in drinking water and the prevalence of skin lesions in West Bengal, India,” International Journal of Epidemiology, vol. 27, no. 5, pp. 871–877, 1998.
[6]  World Health Organization International Agency for Research on Cancer, Some Metals and Metallic Compounds: IARC Monographs on the Evaluation of Carcinogenic Risk of Chemicals to Man, vol. 23, WHO Press, IARC, Lyon, France, 1980.
[7]  Z. Wang and T. G. Rossman, “The carcinogenicity of arsenic,” in Toxicology of Metals, W. C. Louis, Ed., pp. 219–227, CRC Press, Boca Raton, Fla, USA, 1996.
[8]  H. Tinwell, S. C. Stephens, and J. Ashby, “Arsenite as the probable active species in the human carcinogenicity of arsenic: mouse micronucleus assays on Na and K arsenite, orpiment, and Fowler's solution,” Environmental Health Perspectives, vol. 95, pp. 205–210, 1991.
[9]  J. Liu and M. P. Waalkes, “Liver is a target of arsenic carcinogenesis,” Toxicological Sciences, vol. 105, no. 1, pp. 24–32, 2008.
[10]  J. Liu, L. Yu, E. J. Tokar et al., “Arsenic-induced aberrant gene expression in fetal mouse primary liver-cell cultures,” Annals of the New York Academy of Sciences, vol. 1140, pp. 368–375, 2008.
[11]  J. Wu, J. Liu, M. P. Waalkes et al., “High dietary fat exacerbates arsenic-induced liver fibrosis in mice,” Experimental Biology and Medicine, vol. 233, no. 3, pp. 377–384, 2008.
[12]  T. Jiang, Z. Huang, J. Y. Chan, and D. D. Zhang, “Nrf2 protects against As(III)-induced damage in mouse liver and bladder,” Toxicology and Applied Pharmacology, vol. 240, no. 1, pp. 8–14, 2009.
[13]  National Research Council and National Academy of Sciences, Arsenic in Drinking Water, National Academy Press, Washington, DC, USA, 1999.
[14]  D. N. G. Mazumder, “Effect of chronic intake of arsenic-contaminated water on liver,” Toxicology and Applied Pharmacology, vol. 206, no. 2, pp. 169–175, 2005.
[15]  Y. Xu, Y. Wang, Q. Zheng et al., “Clinical manifestations and arsenic methylation after a rare subacute arsenic poisoning accident,” Toxicological Sciences, vol. 103, no. 2, pp. 278–284, 2008.
[16]  A. Santra, J. Das Gupta, B. K. De, B. Roy, and D. N. G. Mazumder, “Hepatic manifestations in chronic arsenic toxicity,” Indian Journal of Gastroenterology, vol. 18, no. 4, pp. 152–155, 1999.
[17]  S. Roy and S. Bhattacharya, “Arsenic-induced histopathology and synthesis of stress proteins in liver and kidney of Channa punctatus,” Ecotoxicology and Environmental Safety, vol. 65, no. 2, pp. 218–229, 2006.
[18]  H. Shi, X. Shi, and K. J. Liu, “Oxidative mechanism of arsenic toxicity and carcinogenesis,” Molecular and Cellular Biochemistry, vol. 255, no. 1-2, pp. 67–78, 2004.
[19]  S. Das, A. Santra, S. Lahiri, and D. N. Guha Mazumder, “Implications of oxidative stress and hepatic cytokine (TNF-α and IL-6) response in the pathogenesis of hepatic collagenesis in chronic arsenic toxicity,” Toxicology and Applied Pharmacology, vol. 204, no. 1, pp. 18–26, 2005.
[20]  B. Li, X. Li, B. Zhu et al., “Sodium arsenite induced reactive oxygen species generation, nuclear factor (erythroid-2 related) factor 2 activation, heme oxygenase-1 expression, and glutathione elevation in Chang human hepatocytes,” Environmental Toxicology, vol. 13, no. 4, 2011.
[21]  K. Jomova, Z. Jenisova, M. Feszterova et al., “Arsenic: toxicity, oxidative stress and human disease,” Journal of Applied Toxicology, vol. 31, no. 2, pp. 95–107, 2011.
[22]  M. Delnomdedieu, M. M. Basti, J. D. Otvos, and D. J. Thomas, “Reduction and binding of arsenate and dimethylarsinate by glutathione: a magnetic resonance study,” Chemico-Biological Interactions, vol. 90, no. 2, pp. 139–155, 1994.
[23]  M. F. Hughes, “Arsenic toxicity and potential mechanisms of action,” Toxicology Letters, vol. 133, no. 1, pp. 1–16, 2002.
[24]  A. Lau, N. F. Villeneuve, Z. Sun, P. K. Wong, and D. D. Zhang, “Dual roles of Nrf2 in cancer,” Pharmacological Research, vol. 58, no. 5-6, pp. 262–270, 2008.
[25]  J. D. Hayes and M. McMahon, “NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer,” Trends in Biochemical Sciences, vol. 34, no. 4, pp. 176–188, 2009.
[26]  L. Baird and A. T. Dinkova-Kostova, “The cytoprotective role of the Keap1-Nrf2 pathway,” Archives of Toxicology, vol. 85, no. 4, pp. 241–272, 2011.
[27]  J. Pi, W. Qu, J. M. Reece, Y. Kumagai, and M. P. Waalkes, “Transcription factor Nrf2 activation by inorganic arsenic in cultured keratinocytes: involvement of hydrogen peroxide,” Experimental Cell Research, vol. 290, no. 2, pp. 234–245, 2003.
[28]  X. He, M. G. Chen, G. X. Lin, and Q. Ma, “Arsenic induces NAD(P)H-quinone oxidoreductase I by disrupting the Nrf2 × Keap1 × Cul3 complex and recruiting Nrf2 × Maf to the antioxidant response element enhancer,” The Journal of Biological Chemistry, vol. 281, no. 33, pp. 23620–23631, 2006.
[29]  X.-J. Wang, Z. Sun, W. Chen, K. E. Eblin, J. A. Gandolfi, and D. D. Zhang, “Nrf2 protects human bladder urothelial cells from arsenite and monomethylarsonous acid toxicity,” Toxicology and Applied Pharmacology, vol. 225, no. 2, pp. 206–213, 2007.
[30]  X.-J. Wang, Z. Sun, W. Chen, Y. Li, N. F. Villeneuve, and D. D. Zhang, “Activation of Nrf2 by arsenite and monomethylarsonous acid is independent of Keap1-C151: enhanced Keap1-Cul3 interaction,” Toxicology and Applied Pharmacology, vol. 230, no. 3, pp. 383–389, 2008.
[31]  H. Endo, Y. Sugioka, Y. Nakagi, Y. Saijo, and T. Yoshida, “A novel role of the NRF2 transcription factor in the regulation of arsenite-mediated keratin 16 gene expression in human keratinocytes,” Environmental Health Perspectives, vol. 116, no. 7, pp. 873–879, 2008.
[32]  D. Meng, X. Wang, Q. Chang et al., “Arsenic promotes angiogenesis in vitro via a heme oxygenase-1-dependent mechanism,” Toxicology and Applied Pharmacology, vol. 244, no. 3, pp. 291–299, 2010.
[33]  E. Beutler, O. Duron, and B. M. Kelly, “Improved method for the determination of blood glutathione,” The Journal of Laboratory and Clinical Medicine, vol. 61, pp. 882–888, 1963.
[34]  W. H. Habig, M. J. Pabst, and W. B. Jakoby, “Glutathione S transferases. The first enzymatic step in mercapturic acid formation,” Journal of Biological Chemistry, vol. 249, no. 22, pp. 7130–7139, 1974.
[35]  J. A. Buege and S. D. Aust, “Microsomal lipid peroxidation,” in Methods in Enzymology, S. Fleisher and L. Packer, Eds., pp. 302–310, Academic Press, New York, NY, USA, 1978.
[36]  H. Aebi, “Catalase in vitro,” in Methods in Enzymology, L. Packer, Ed., pp. 121–126, Academic Press, Orlando, Fla, USA, 1984.
[37]  N. Kawamura, “Catalase,” in Experimental Protocols for Reactive Oxygen and Nitrogen Species, J. M. C. Gutteridge and N. Taniguchi, Eds., pp. 77–78, Oxford University Press, New York, NY, USA, 1999.
[38]  A. Chattopadhyay, S. Podder, S. Agarwal, and S. Bhattacharya, “Fluoride-induced histopathology and synthesis of stress protein in liver and kidney of mice,” Archives of Toxicology, vol. 85, no. 4, pp. 327–335, 2011.
[39]  O. H. Lowry, N. J. Rosebrough, A. L. Farr, and R. J. Randall, “Protein measurement with the folin phenol reagent,” The Journal of Biological Chemistry, vol. 193, no. 1, pp. 265–275, 1951.
[40]  N. Li, J. Alam, M. I. Venkatesan et al., “Nrf2 is a key transcription factor that regulates antioxidant defense in macrophages and epithelial cells: protecting against the proinflammatory and oxidizing effects of diesel exhaust chemicals,” Journal of Immunology, vol. 173, no. 5, pp. 3467–3481, 2004.
[41]  K. Shimano, M. Satake, A. Okaya et al., “Hepatic oval cells have the side population phenotype defined by expression of ATP-binding cassette transporter ABCG2/BCRP1,” American Journal of Pathology, vol. 163, no. 1, pp. 3–9, 2003.
[42]  J. Aono, T. Yanagawa, K. Itoh et al., “Activation of Nrf2 and accumulation of ubiquitinated A170 by arsenic in osteoblasts,” Biochemical and Biophysical Research Communications, vol. 305, no. 2, pp. 271–277, 2003.
[43]  H. Harada, R. Sugimoto, A. Watanabe et al., “Differential roles for Nrf2 and AP-1 in upregulation of HO-1 expression by arsenite in murine embryonic fibroblasts,” Free Radical Research, vol. 42, no. 4, pp. 297–304, 2008.
[44]  N. Singh, D. Kumar, K. Lal, S. Raisuddin, and A. P. Sahu, “Adverse health effects due to arsenic exposure: modification by dietary supplementation of jaggery in mice,” Toxicology and Applied Pharmacology, vol. 242, no. 3, pp. 247–255, 2010.
[45]  A. Santra, A. Maiti, S. Das, S. Lahiri, S. K. Charkaborty, and D. N. Guha Mazumder, “Hepatic damage caused by chronic arsenic toxicity in experimental animals,” Journal of Toxicology, vol. 38, no. 4, pp. 395–405, 2000.
[46]  Z. Drobná, F. S. Walton, D. S. Paul, W. Xing, D. J. Thomas, and M. Styblo, “Metabolism of arsenic in human liver: the role of membrane transporters,” Archives of Toxicology, vol. 84, pp. 3–16, 2010.
[47]  N. Tandan, M. Roy, and S. Roy, “Ameliorative potential of Psidium guajava on hemato-biochemical alterations in arsenic-exposed wistar rats,” Toxicology International, vol. 19, pp. 121–124, 2012.
[48]  S. J. S. Flora, S. C. Pant, P. R. Malhotra, and G. M. Kannan, “Biochemical and histopathological changes in arsenic-intoxicated rats coexposed to ethanol,” Alcohol, vol. 14, no. 6, pp. 563–568, 1997.
[49]  R. Ferzand, J. A. Gadahi, S. Saleha, and Q. Ali, “Histological and haematological disturbance caused by arsenic toxicity in mice model,” Pakistan Journal of Biological Sciences, vol. 11, no. 11, pp. 1405–1413, 2008.
[50]  R. N. P. Das Neves, F. Carvalho, M. Carvalho et al., “Protective activity of hesperidin and lipoic acid against sodium arsenite acute toxicity in mice,” Toxicologic Pathology, vol. 32, no. 5, pp. 527–535, 2004.
[51]  K. T. Kitchin, “Recent advances in arsenic carcinogenesis: modes of action, animal model systems, and methylated arsenic metabolites,” Toxicology and Applied Pharmacology, vol. 172, no. 3, pp. 249–261, 2001.
[52]  S. J. S. Flora, S. Bhadauria, S. C. Pant, and R. K. Dhaked, “Arsenic induced blood and brain oxidative stress and its response to some thiol chelators in rats,” Life Sciences, vol. 77, no. 18, pp. 2324–2337, 2005.
[53]  S. J. S. Flora, S. Bhadauria, G. M. Kannan, and N. Singh, “Arsenic induced oxidative stress and the role of antioxidant supplementation during chelation: a review,” Journal of Environmental Biology, vol. 28, no. 2, pp. 333–347, 2007.
[54]  M. E. Vahter, “Interactions between arsenic-induced toxicity and nutrition in early life,” Journal of Nutrition, vol. 137, no. 12, pp. 2798–2804, 2007.
[55]  E. O. Farombi, O. A. Adelowo, and Y. R. Ajimoko, “Biomarkers of oxidative stress and heavy metal levels as indicators of environmental pollution in African cat fish (Clarias gariepinus) from Nigeria Ogun River,” International Journal of Environmental Research and Public Health, vol. 4, no. 2, pp. 158–165, 2007.
[56]  S. Bashir, Y. Sharma, M. Irshad, S. D. Gupta, and T. D. Dogra, “Arsenic-induced cell death in liver and brain of experimental rats,” Basic and Clinical Pharmacology and Toxicology, vol. 98, no. 1, pp. 38–43, 2006.
[57]  M. Mittal and S. J. S. Flora, “Effects of individual and combined exposure to sodium arsenite and sodium fluoride on tissue oxidative stress, arsenic and fluoride levels in male mice,” Chemico-Biological Interactions, vol. 162, no. 2, pp. 128–139, 2006.
[58]  C.-H. Guo, W.-S. Ko, P.-C. Chen, G.-S. W. Hsu, C.-Y. Lin, and C.-L. Wang, “Alterations in trace elements and oxidative stress in uremic patients with dementia,” Biological Trace Element Research, vol. 131, no. 1, pp. 13–24, 2009.
[59]  Z. Y. Zhang, N. Q. Liu, F. L. Li et al., “Characterization of Fe, Cu and Zn in organs of PDAPP transgenic mice by XRF spectrometry,” X-Ray Spectrometry, vol. 35, no. 4, pp. 253–256, 2006.
[60]  T. Kowalik-Jankowska, M. Ruta-Dolejsz, K. Wisniewska, L. Lankiewicz, and H. Kozlowski, “Possible involvement of Copper(II) in Alzheimer disease,” Environmental Health Perspectives, vol. 110, no. 5, pp. 869–870, 2002.
[61]  O. A. Levander, “Metabolic interrelationships between arsenic and selenium,” Environmental Health Perspectives, vol. 19, pp. 159–164, 1977.
[62]  T. G. Rossman and A. N. Uddin, “Selenium prevents spontaneous and arsenite-induced mutagenesis,” International Congress Series, vol. 1275, pp. 173–179, 2004.
[63]  Y. Molin, P. Frisk, and N.-G. Ilb?ck, “Sequential effects of daily arsenic trioxide treatment on essential and nonessential trace elements in tissues in mice,” Anti-Cancer Drugs, vol. 19, no. 8, pp. 812–818, 2008.
[64]  Y. Molin, P. Frisk, and N.-G. Ilback, “Arsenic trioxide affects the trace element balance in tissues in infected and healthy mice differently,” Anticancer Research, vol. 29, no. 1, pp. 83–90, 2009.
[65]  M. Haidari, E. Javadi, A. Sanati, M. Hajilooi, and J. Ghanbili, “Association of increased ferritin with premature coronary stenosis in men,” Clinical Chemistry, vol. 47, no. 9, pp. 1666–1672, 2001.
[66]  Z. Durackova, L. Bergendi, A. Liptakova, and J. Muchova, “Free radicals derived from oxygen and medicine,” Bratislava Medical Journal, vol. 94, pp. 419–434, 1993.
[67]  M. Ahmad, M. A. Khan, and A. S. Khan, “Oxidative stress and level of-iron indices in coronary heart disease patients,” Journal of Ayub Medical College, Abbottabad, vol. 21, no. 2, pp. 56–59, 2009.
[68]  A. Jain, T. Lamark, E. Sj?ttem et al., “p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription,” Journal of Biological Chemistry, vol. 285, no. 29, pp. 22576–22591, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133