1
|
Li S, Yin Y, Dong X, Xu L, Yang Z, Li H, Zou Y, Wu Z. Protective Effects of Lactoferrin Treatment Against Sodium Arsenite Exposure-Induced Nephrotoxicity. Biol Trace Elem Res 2025; 203:1539-1554. [PMID: 38833108 DOI: 10.1007/s12011-024-04256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
It is said that a wide range of renal functions are at risk from arsenic exposure. We examined how lactoferrin administration may mitigate inflammation, apoptosis, redox imbalance, and fibrosis in order to counteract arsenic-induced nephrotoxicity. Accordingly, male C57BL/6 mice (6 weeks) were divided into six experimental groups with six mice in each group. The first and second groups were intragastrically administered normal saline and sodium arsenite (NaAsO2) at 5 mg/kg body weight concentrations as the negative control (NC) and NaAsO2 groups. The third, fourth, and fifth groups were intragastrically administered lactoferrin at concentrations of 100, 200, and 400 mg/kg body weight in addition to NaAsO2 at concentrations of 5 mg/kg body weight. The sixth group was intragastrically administered lactoferrin at a concentration of 200 mg/kg body weight with the experimental group set as the lactoferrin group. After daily drug administration for 4 weeks, the lactoferrin concentrations were optimized based on the results of renal index and renal function. Histopathological, biochemical, and gene expression analyses were performed to evaluate the status of renal tissue architecture, redox imbalance, inflammation, apoptosis, and fibrosis to confirm the alleviative effect of lactoferrin treatment against the NaAsO2 exposure-induced nephrotoxicity. The results confirmed that the 200 mg/kg lactoferrin treatment mitigated these arsenic effects and maintained the normal renal frameworks. Conclusively, disrupting the renal redox balance and triggering inflammation, apoptosis, along with fibrosis is a milieu that arsenic, robustly exerts its nephrotoxic effect. Lactoferrin, probably by its direct and indirect control mechanism on these said pathways, can mitigate the nephrotoxicity and preserve the normal renal health.
Collapse
Affiliation(s)
- Shubin Li
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Yaning Yin
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Xingna Dong
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Limeng Xu
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Zehao Yang
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Hong Li
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Yanhui Zou
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China.
| | - Zhenli Wu
- Department of Geriatric Medical Center, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China.
| |
Collapse
|
2
|
Wang D, He R, Song Q, Diao H, Jin Y, Zhang A. Calcitriol Inhibits NaAsO 2 Triggered Hepatic Stellate Cells Activation and Extracellular Matrix Oversecretion by Activating Nrf2 Signaling Pathway Through Vitamin D Receptor. Biol Trace Elem Res 2024; 202:3601-3613. [PMID: 37968493 DOI: 10.1007/s12011-023-03957-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
Previous studies, including our own, have demonstrated that arsenic exposure can induce liver fibrosis, while the underlying mechanism remains unclear and there is currently no effective pharmacological intervention available. Recent research has demonstrated that vitamin D supplementation can ameliorate liver fibrosis caused by various etiologies, potentially through modulation of the Nrf2 signaling pathways. However, it remains unclear whether vitamin D intervention can mitigate arsenic-caused liver fibrosis. As is known hepatic stellate cells (HSCs) activation and extracellular matrix (ECM) deposition are pivotal in the pathogenesis of liver fibrosis. In this study, we investigated the intervention effect of calcitriol (a form of active vitamin D) on arsenite-triggered Lx-2 cells (a human hepatic stellate cell line) activation and ECM oversecretion. Additionally, we also elucidated the role and mechanism of Nrf2 antioxidant signaling pathway. Our results demonstrated that calcitriol intervention significantly inhibits Lx-2 cell activation and ECM oversecretion induced by arsenite exposure. Additionally, calcitriol activates Nrf2 and its downstream antioxidant enzyme expression in Lx-2 cells, thereby reducing ROS overproduction caused by arsenite exposure. Further investigation reveals that calcitriol activates the Nrf2 signaling pathway and inhibits arsenite-triggered Lx-2 cell activation and ECM oversecretion by targeting vitamin D receptor (VDR). In conclusion, this study has demonstrated that vitamin D intervention can effectively inhibit HSC activation and ECM oversecretion triggered by arsenite exposure through its antioxidant activity. This provides a novel strategy for targeted nutritional intervention in the treatment of arsenic-induced liver fibrosis.
Collapse
Affiliation(s)
- Dapeng Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China.
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China.
| | - Rui He
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Qian Song
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Heng Diao
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Ying Jin
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Aihua Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China.
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China.
| |
Collapse
|
3
|
Arsenic Induces Continuous Inflammation and Regulates Th1/Th2/Th17/Treg Balance in Liver and Kidney In Vivo. Mediators Inflamm 2022; 2022:8414047. [PMID: 35210942 PMCID: PMC8863494 DOI: 10.1155/2022/8414047] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/26/2022] [Indexed: 12/26/2022] Open
Abstract
Numerous studies on arsenic-induced hepatonephric toxicity including cancer have been reported. Given that chronic inflammatory response and immune imbalance are associated with oncogenesis, we investigated whether arsenic could influence the hepatic and nephritic expression of inflammatory factors and the differentiation of T cells. Mice were exposed to NaAsO2 (0, 25, and 50 mg/L) for 1 and 3 months. Our data showed the destruction of the structure and inflammatory infiltration in the liver. The arsenic markedly increased the activity of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST). The myeloperoxidase (MPO) activities increased in the liver at 25 and 50 mg/L arsenic for 3 months as well as in the kidney at both 1 and 3 months. An increased expression of inflammatory indicators (IL-1β, IL-12, and TNF-α) at 25 and 50 mg/L arsenic for 1 and 3 months in the liver and kidney, as well as IL-1β in the liver for 3 months and in the kidney at 50 mg/L for 1 and 3 months were demonstrated in our experiments. Besides, a definite tendency toward Th1/Th17 cytokines in the liver while Th2/Th17 cytokines in kidney was also observed by arsenic. Moreover, arsenic enhanced the expression of MAPK/Nrf2/NF-κB signaling molecules. In conclusion, the results of the study suggested that arsenic induces continuous immune-inflammatory responses in the liver and kidney.
Collapse
|
4
|
Inesta-Vaquera F, Navasumrit P, Henderson CJ, Frangova TG, Honda T, Dinkova-Kostova AT, Ruchirawat M, Wolf CR. Application of the in vivo oxidative stress reporter Hmox1 as mechanistic biomarker of arsenic toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 270:116053. [PMID: 33213951 DOI: 10.1016/j.envpol.2020.116053] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/02/2020] [Accepted: 11/06/2020] [Indexed: 05/26/2023]
Abstract
Inorganic arsenic (iAs) is a naturally occurring metalloid present in drinking water and polluted air exposing millions of people globally. Epidemiological studies have linked iAs exposure to the development of numerous diseases including cognitive impairment, cardiovascular failure and cancer. Despite intense research, an effective therapy for chronic arsenicosis has yet to be developed. Laboratory studies have been of great benefit in establishing the pathways involved in iAs toxicity and providing insights into its mechanism of action. However, the in vivo analysis of arsenic toxicity mechanisms has been difficult by the lack of reliable in vivo biomarkers of iAs's effects. To address this issue we have applied the use of our recently developed stress reporter models to study iAs toxicity. The reporter mice Hmox1 (oxidative stress/inflammation; HOTT) and p21 (DNA damage) were exposed to iAs at acute and chronic, environmentally relevant, doses. We observed induction of the oxidative stress reporters in several cell types and tissues, which was largely dependent on the activation of transcription factor NRF2. We propose that our HOTT reporter model can be used as a surrogate biomarker of iAs-induced oxidative stress, and it constitutes a first-in-class platform to develop treatments aimed to counteract the role of oxidative stress in arsenicosis. Indeed, in a proof of concept experiment, the HOTT reporter mice were able to predict the therapeutic utility of the antioxidant N-acetyl cysteine in the prevention of iAs associated toxicity.
Collapse
Affiliation(s)
- Francisco Inesta-Vaquera
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK.
| | - Panida Navasumrit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Colin J Henderson
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Tanya G Frangova
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Tadashi Honda
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY, 11794-3400, USA
| | - Albena T Dinkova-Kostova
- Department of Molecular Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - C Roland Wolf
- Department of Systems Medicine. School of Medicine. University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| |
Collapse
|
5
|
A Systematic Review of the Various Effect of Arsenic on Glutathione Synthesis In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9414196. [PMID: 32802886 PMCID: PMC7411465 DOI: 10.1155/2020/9414196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/29/2020] [Indexed: 01/03/2023]
Abstract
Background Arsenic is a toxic metalloid widely present in nature, and arsenic poisoning in drinking water is a serious global public problem. Glutathione is an important reducing agent that inhibits arsenic-induced oxidative stress and participates in arsenic methylation metabolism. Therefore, glutathione plays an important role in regulating arsenic toxicity. In recent years, a large number of studies have shown that arsenic can regulate glutathione synthesis in many ways, but there are many contradictions in the research results. At present, the mechanism of the effect of arsenic on glutathione synthesis has not been elucidated. Objective We will conduct a meta-analysis to illustrate the effects of arsenic on GSH synthesis precursors Glu, Cys, Gly, and rate-limiting enzyme γ-GCS in mammalian models, as well as the regulation of p38/Nrf2 of γ-GCS subunit GCLC, and further explore the molecular mechanism of arsenic affecting glutathione synthesis. Results This meta-analysis included 30 studies in vivo and 58 studies in vitro, among which in vivo studies showed that arsenic exposure could reduce the contents of GSH (SMD = -2.86, 95% CI (-4.45, -1.27)), Glu (SMD = -1.11, 95% CI (-2.20,-0.02)), and Cys (SMD = -1.48, 95% CI (-2.63, -0.33)), with no statistically significant difference in p38/Nrf2, GCLC, and GCLM. In vitro studies showed that arsenic exposure increased intracellular GSH content (SMD = 1.87, 95% CI (0.18, 3.56)) and promoted the expression of p-p38 (SMD = 4.19, 95% CI (2.34, 6.05)), Nrf2 (SMD = 4.60, 95% CI (2.34, 6.86)), and GCLC (SMD = 1.32, 95% CI (0.23, 2.41)); the p38 inhibitor inhibited the expression of Nrf2 (SMD = -1.27, 95% CI (-2.46, -0.09)) and GCLC (SMD = -5.37, 95% CI (-5.37, -2.20)); siNrf2 inhibited the expression of GCLC, and BSO inhibited the synthesis of GSH. There is a dose-dependent relationship between the effects of exposure on GSH in vitro. Conclusions. These indicate the difference between in vivo and in vitro studies of the effect of arsenic on glutathione synthesis. In vivo studies have shown that arsenic exposure can reduce glutamate and cysteine levels and inhibit glutathione synthesis, while in vitro studies have shown that chronic low-dose arsenic exposure can activate the p38/Nrf2 pathway, upregulate GCLC expression, and promote glutathione synthesis.
Collapse
|
6
|
Liu Y, Liang Y, Zheng B, Chu L, Ma D, Wang H, Chu X, Zhang J. Protective Effects of Crocetin on Arsenic Trioxide-Induced Hepatic Injury: Involvement of Suppression in Oxidative Stress and Inflammation Through Activation of Nrf2 Signaling Pathway in Rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1921-1931. [PMID: 32546959 PMCID: PMC7245440 DOI: 10.2147/dddt.s247947] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022]
Abstract
Purpose Arsenic trioxide (ATO) has been shown to induce hepatic injury. Crocetin is a primary constituent of saffron, which has been verified to have antioxidant and anti-inflammatory effects. In the current experiment, we evaluated the efficacy of crocetin against ATO-induced hepatic injury and explored the potential molecular mechanisms in rats. Methods Rats were pretreated with 25 or 50 mg/kg crocetin 6 h prior to treating with 5 mg/kg ATO to induce hepatic injury daily for 7 days. Results Treatment with crocetin attenuated ATO-induced body weight loss, decreases in food and water consumption, and improved ATO-induced hepatic pathological damage. Crocetin significantly inhibited ATO-induced alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) increases. Crocetin prevented ATO-induced liver malondialdehyde (MDA) and reactive oxygen species (ROS) levels. Crocetin abrogated the ATO-induced decrease of catalase (CAT) and superoxide dismutase (SOD) activity. Crocetin was found to significantly restore the protein levels of interleukin 6 (IL-6), interleukin 1β (IL-1β), and tumor necrosis factor-alpha (TNF-α). Furthermore, crocetin promoted the expression of nuclear factor erythroid 2 related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and NADP(H): quinone oxidoreductase 1 (NQO1). Conclusion These findings suggest that crocetin ameliorates ATO-induced hepatic injury in rats. In addition, the effect of crocetin might be related to its role in antioxidant stress, as an anti-inflammatory agent, and in regulating the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yanshuang Liu
- Department of Diagnostics, School of Integrated Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China.,Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Shijiazhuang 050200, Hebei, People's Republic of China
| | - Yingran Liang
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Bin Zheng
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Li Chu
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Donglai Ma
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Hongfang Wang
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Jianping Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Shijiazhuang 050200, Hebei, People's Republic of China.,Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| |
Collapse
|
7
|
Tian X, Feng J, Dong N, Lyu Y, Wei C, Li B, Ma Y, Xie J, Qiu Y, Song G, Ren X, Yan X. Subchronic exposure to arsenite and fluoride from gestation to puberty induces oxidative stress and disrupts ultrastructure in the kidneys of rat offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 686:1229-1237. [PMID: 31412519 DOI: 10.1016/j.scitotenv.2019.04.409] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/26/2019] [Accepted: 04/27/2019] [Indexed: 06/10/2023]
Abstract
Underground drinking water is commonly contaminated with arsenite (As) and fluoride (F) associated with chronic kidney diseases in humans; however, the combined renal toxicity of these pollutants and the underlying mechanisms are still unclear. The aim of the present study was to investigate the interaction between As and F regarding toxic effects on the kidney of rat offspring exposed to pollutants during prenatal and postnatal development. Pregnant rats were randomly divided into four groups that received NaAsO2 (50 mg/L), NaF (100 mg/L), NaAsO2 (50 mg/L) and NaF (100 mg/L) in drinking water, or clean water, respectively, during gestation and lactation. After weaning, six male pups were randomly selected from each group and continued on the same treatment as their mothers for up to three months. The results revealed that subchronic exposure to high-dose As and/or F decreased the organ coefficient of the kidneys and disrupted kidney ultrastructure, moreover inhibited the activity of antioxidant enzymes and increased the generation of malondialdehyde in the kidney. As exposure alone or combined with F led to an upregulation of nuclear factor erythroid 2-related factor-2 (Nrf2) and its regulatory targets (Ho-1, Gclc, and Nqo1), whereas the effect of F alone was not significant. These results suggest that the renal toxicity of As and F is associated with the induction of mitochondrial damage and oxidative stress, and alters the expression of Nrf2 and its regulatory targets. Furthermore, variance analysis results showed that an interaction between As and F in the toxicity process.
Collapse
Affiliation(s)
- Xiaolin Tian
- Shanxi Key Laboratory of Experimental Animal and Human Disease Animal Models, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Jing Feng
- Shanxi Key Laboratory of Experimental Animal and Human Disease Animal Models, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Nisha Dong
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Yi Lyu
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Cailing Wei
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Ben Li
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Yanqin Ma
- College of Life Science, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jiaxin Xie
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Yulan Qiu
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Guohua Song
- Shanxi Key Laboratory of Experimental Animal and Human Disease Animal Models, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Xuefeng Ren
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, The State University of New York, Buffalo, NY 14214, USA; Department of Pharmacology and Toxicology, School of Biomedical Sciences, The State University of New York, Buffalo, NY 14214, USA
| | - Xiaoyan Yan
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China.
| |
Collapse
|
8
|
Wang C, Niu Q, Ma R, Song G, Hu Y, Xu S, Li Y, Wang H, Li S, Ding Y. The Variable Regulatory Effect of Arsenic on Nrf2 Signaling Pathway in Mouse: a Systematic Review and Meta-analysis. Biol Trace Elem Res 2019; 190:362-383. [PMID: 30357758 DOI: 10.1007/s12011-018-1549-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023]
Abstract
Arsenic is known to cause oxidative damage. Nuclear factor E2-relate factor-2 (Nrf2) can resist this toxicity. Scholars demonstrated that Nrf2 pathway was activated by arsenic. In contrast, other articles established arsenic-induced inhibition of Nrf2 pathway. To resolve the contradiction and elucidate the mechanism of Nrf2 induced by arsenic, 39 publications involving mouse models were identified through exhaustive database retrieval and were analyzed. The pooled results suggested that arsenic obviously elevated transcription and translation levels of Nrf2 and its downstream genes, NAD(P)H dehydrogenase 1 (NQO1), heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and GST-glutathione-S-transferase1/2 (GSTO1/2). Arsenic increased the level of reactive oxygen species (ROS), but reduced the level of glutathione (GSH). Subgroup analysis between arsenic and control groups showed that the levels of Nrf2 and its downstream genes are greater in high dose than that in the low dose, higher in short-term exposure than long term, female subjects tolerated better than males, higher in mice than the rats, and greater in other organs than the liver. However, the contents of genes of Nrf2 pathway between the arsenic and control groups were lower in rats than in mice and were less for long-term exposure than the short term (P < 0.05). Conclusively, a variable regulation of arsenic on Nrf2 pathway is noted. Higher dose and short-term exposure of female mice organs except for liver promoted Nrf2 pathway. On the other hand, arsenic inhibited Nrf2 pathway for long-term exposure on rats.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Qiang Niu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Rulin Ma
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Guanling Song
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Yunhua Hu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Shangzhi Xu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Yu Li
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Haixia Wang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Shugang Li
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China.
| | - Yusong Ding
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China.
| |
Collapse
|
9
|
Cheng X, Hu J, Li J, Chen J, Wang H, Mao T, Xue B, Li B. The silk gland damage and the transcriptional response to detoxifying enzymes-related genes of Bombyx mori under phoxim exposure. CHEMOSPHERE 2018; 209:964-971. [PMID: 30114747 DOI: 10.1016/j.chemosphere.2018.06.167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 06/08/2023]
Abstract
Silk gland is a major organ of Bombyx mori for the synthesis and secretion of silk protein. Phoxim exposure can be toxic to B. mori and causes a decrease of fibroin synthesis, finally affecting the silk production in industry. To study the mechanism of metabolism and detoxification of silk gland under phoxim exposure, we measured the residual quantity of phoxim in silk gland and hemolymph after phoxim exposure, and the detoxifying enzymes-related genes and enzyme activity were also investigated. Results indicated that the residual amount of phoxim existed up to 24 h in silk gland compared with that in hemolymph, suggesting that phoxim can accumulate in the silk glands within a certain time course. The transcriptional levels of PI3K/Akt genes, including Akt, Tor1, p70s6k and 4e-bp, were up-regulated by 6.919, 1.358, 10.766 and 7.708-fold, respectively. The expression of two downstream genes (CncC and Keap1) was up-regulated by 1.939 and 3.373-fold, respectively. In addition, the transcriptional levels of detoxification-related genes including CYP6AB, CYP306A, CarE2, GST1 and GSTd1 were up-regulated by 1.731, 1.221, 1.366, 1.376 and 6.591-fold, respectively. The enzymatic activity of CYP450, CarE and GST were increased over time. These results provided possible insights into the injury of silk gland and the transcriptional response to detoxifying enzymes-related genes in silkworm after phoxim exposure.
Collapse
Affiliation(s)
- Xiaoyu Cheng
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jiahuan Hu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jinxin Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jian Chen
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Hui Wang
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Tingting Mao
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bin Xue
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bing Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu 215123, PR China; National Engineering Laboratory for Modern Silk, Soochow University, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|
10
|
Firdaus F, Zafeer MF, Ahmad M, Afzal M. Anxiolytic and anti-inflammatory role of thymoquinone in arsenic-induced hippocampal toxicity in Wistar rats. Heliyon 2018; 4:e00650. [PMID: 29984327 PMCID: PMC6024171 DOI: 10.1016/j.heliyon.2018.e00650] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/20/2018] [Accepted: 06/05/2018] [Indexed: 12/05/2022] Open
Abstract
Arsenic (As) is a widely existing metalloid in the biosphere. Drinking water contamination by arsenic is a major route of human exposure, either by natural means or through industrial pollution. Numerous evidence form earlier reports suggest that arsenic exposure causes cerebral neurodegeneration which initiates behavioral disturbances concomitant to psychiatric disorders. Also, mood disorders in humans as well as in animals correlate with arsenic exposure; the present study is carried out to implore the neuroprotective potential of thymoquinone (TQ) in arsenic-stressed rats. TQ is an active component of Nigella sativa (Kalonji) seed oil. Arsenic exposure in the form of sodium arsenate (10 mg/kg/day; p.o) caused neurobehavioral deficits as evidenced by changes in locomotion and exploratory behavior in open-field and elevated plus maze tasks. Alongside this, arsenate also elevated hippocampal oxidative stress parameters like lipid peroxidation (TBARS) and protein carbonyl formation with a decrease in superoxide dismutase (SOD) and reduced glutathione (GSH) content. Genotoxicity assessment by Comet assay also showed prominent levels of DNA damage. Furthermore, arsenic also elevated hippocampal cytokine levels, TNF-α and INF-γ. However, TQ supplementation (2.5 and 5 mg/kg/day, p.o) preceded three days before arsenic administration, significantly attenuated arsenic-associated anxiogenic changes which majorly attributed to its antioxidant and anxiolytic potential. Also, TQ pre-treated rats expressed positive shifts in the hippocampal oxidative stress and cytokine levels with decreased DNA fragmentation. Thus, this study concludes that TQ might serve as a strong therapeutic agent for management of anxiety and depressive outcomes of arsenic intoxication.
Collapse
Affiliation(s)
- Fakiha Firdaus
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohd Faraz Zafeer
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Masood Ahmad
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohammad Afzal
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
11
|
Bambino K, Zhang C, Austin C, Amarasiriwardena C, Arora M, Chu J, Sadler KC. Inorganic arsenic causes fatty liver and interacts with ethanol to cause alcoholic liver disease in zebrafish. Dis Model Mech 2018; 11:dmm.031575. [PMID: 29361514 PMCID: PMC5894941 DOI: 10.1242/dmm.031575] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
The rapid increase in fatty liver disease (FLD) incidence is attributed largely to genetic and lifestyle factors; however, environmental toxicants are a frequently overlooked factor that can modify the effects of more common causes of FLD. Chronic exposure to inorganic arsenic (iAs) is associated with liver disease in humans and animal models, but neither the mechanism of action nor the combinatorial interaction with other disease-causing factors has been fully investigated. Here, we examined the contribution of iAs to FLD using zebrafish and tested the interaction with ethanol to cause alcoholic liver disease (ALD). We report that zebrafish exposed to iAs throughout development developed specific phenotypes beginning at 4 days post-fertilization (dpf), including the development of FLD in over 50% of larvae by 5 dpf. Comparative transcriptomic analysis of livers from larvae exposed to either iAs or ethanol revealed the oxidative stress response and the unfolded protein response (UPR) caused by endoplasmic reticulum (ER) stress as common pathways in both these models of FLD, suggesting that they target similar cellular processes. This was confirmed by our finding that arsenic is synthetically lethal with both ethanol and a well-characterized ER-stress-inducing agent (tunicamycin), suggesting that these exposures work together through UPR activation to cause iAs toxicity. Most significantly, combined exposure to sub-toxic concentrations of iAs and ethanol potentiated the expression of UPR-associated genes, cooperated to induce FLD, reduced the expression of as3mt, which encodes an arsenic-metabolizing enzyme, and significantly increased the concentration of iAs in the liver. This demonstrates that iAs exposure is sufficient to cause FLD and that low doses of iAs can potentiate the effects of ethanol to cause liver disease. This article has an associated First Person interview with the first author of the paper. Summary: Using zebrafish, the authors show that exposure to a common environmental contaminant, inorganic arsenic, increases the risk of alcoholic liver disease.
Collapse
Affiliation(s)
- Kathryn Bambino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chi Zhang
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| | - Christine Austin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jaime Chu
- Department of Pediatrics, Division of Pediatric Hepatology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| |
Collapse
|
12
|
Oyagbemi AA, Omobowale TO, Ola-Davies OE, Adejumobi OA, Asenuga ER, Adeniji FK, Adedapo AA, Yakubu MA. Protective Effect of Azadirachta indica and Vitamin E Against Arsenic Acid-Induced Genotoxicity and Apoptosis in Rats. J Diet Suppl 2017; 15:251-268. [PMID: 28777671 DOI: 10.1080/19390211.2017.1336147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Sodium arsenite (NaAsO2) is one of the major environmental toxicants with severe toxicological consequences in some developing and developed countries. Rats in Group A received normal saline. Genotoxicity and apoptosis were induced by single intraperitoneal injection of 10 mg/kg sodium arsenite to rats in Groups B-F. Rats in Groups C and D had earlier been pretreated with Azadirachta indica (100 and 200 mg/kg) or E and F with vitamin E (50 and 100 mg/kg), respectively. Markers of oxidative stress, inflammation, hepatic damage, genotoxicity, and apoptosis were assessed. Pretreatment of rats with either Azadirachta indica or vitamin E led to a significant (p <.05) increase in the activities of glutathione-S-transferase (GST), catalase (CAT), superoxide dismutase (SOD), and reduced glutathione (GSH) in the liver compared to the group that received NaAsO2 alone. Markers of oxidative stress and inflammation, malondialdehyde (MDA), hydrogen peroxide (H2O2) generation, nitric oxide (NO), and myeloperoxidase (MPO), were significantly (p <.05) lowered in rats pretreated with Azadirachta indica or vitamin E. The frequency of micronucleated polychromatic erythrocytes (MNPCEs) and expression of caspase-3 were significantly (p <.05) reduced in rats pretreated with either Azadirachta indica or vitamin E compared to rats intoxicated with arsenite. Histopathology of the liver showed areas of infiltration of inflammatory cells with deaths of numerous hepatocytes in NaAsO2-intoxicated rats, and these were reversed by Azadirachta indica. Together, we report for the first time the genoprotective and antiapoptotic effect of Azadirachta indica by a significant reduction in the frequency of micronuclei-induced apoptosis and oxidative stress by arsenic intoxication.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- a Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine , University of Ibadan , Ibadan , Nigeria
| | - Temidayo Olutayo Omobowale
- b Department of Veterinary Medicine, Faculty of Veterinary Medicine , University of Ibadan , Ibadan , Nigeria
| | - Olufunke Eunice Ola-Davies
- a Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine , University of Ibadan , Ibadan , Nigeria
| | - Olumuyiwa Abiola Adejumobi
- b Department of Veterinary Medicine, Faculty of Veterinary Medicine , University of Ibadan , Ibadan , Nigeria
| | - Ebunoluwa Rachael Asenuga
- c Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine , University of Benin , Benin City , Nigeria
| | - Funmilola Kehinde Adeniji
- a Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine , University of Ibadan , Ibadan , Nigeria
| | - Adeolu Alex Adedapo
- d Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine , University of Ibadan , Ibadan , Nigeria
| | - Momoh Audu Yakubu
- e Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology , NSB303, Vascular Biology Unit, Center for Cardiovascular Diseases, COPHS, Texas Southern University , Houston , TX , USA
| |
Collapse
|
13
|
Chen C, Jiang X, Gu S, Lai Y, Liu Y, Zhang Z. Protection of Nrf2 against arsenite-induced oxidative damage is regulated by the cyclic guanosine monophosphate-protein kinase G signaling pathway. ENVIRONMENTAL TOXICOLOGY 2017; 32:2004-2020. [PMID: 27774770 PMCID: PMC5403658 DOI: 10.1002/tox.22374] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 10/03/2016] [Accepted: 10/05/2016] [Indexed: 05/05/2023]
Abstract
Arsenite has been shown to induce a variety of oxidative damage in mammalian cells. However, the mechanisms underlying cellular responses to its adverse effects remain unknown. We previously showed that the level of Nrf2, a nuclear transcription factor significantly increased in arsenite-treated human bronchial epithelial (HBE) cells suggesting that Nrf2 is involved in responding to arsenite-induced oxidative damage. To explore how Nrf2 can impact arsenite-induced oxidative damage, in this study, we examined Nrf2 activation and its regulation upon cellular arsenite exposure as well as its effects on arsenite-induced oxidative damage in HBE cells. We found that Nrf2 mRNA and protein levels were significantly increased by arsenite in a dose- and time-dependent manner. Furthermore, we showed that over-expression of Nrf2 significantly reduced the level of arsenite-induced oxidative damage in HBE cells including DNA damage, chromosomal breakage, lipid peroxidation and depletion of antioxidants. This indicates a protective role of Nrf2 against arsenite toxicity. This was further supported by the fact that activation of Nrf2 by its agonists, tertiary butylhydroquinone (t-BHQ) and sulforaphane (SFN) resulted in the same protective effects against arsenite toxicity. Moreover, we demonstrated that arsenite-induced activation of Nrf2 was mediated by the cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) signaling pathway. This is the first evidence showing that Nrf2 protects against arsenite-induced oxidative damage through the cGMP-PKG pathway. Our study suggests that activation of Nrf2 through the cGMP-PKG signaling pathway in HBE cells may be developed as a new strategy for prevention of arsenite toxicity. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 2004-2020, 2017.
Collapse
Affiliation(s)
- Chengzhi Chen
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing, People’s Republic of China
| | - Xuejun Jiang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing, People’s Republic of China
| | - Shiyan Gu
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biochemistry Ph.D. Program, Florida International University, Miami, Florida, USA
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, USA
- Corresponding authors: Zunzhen Zhang, Ph.D., Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu 610041, People’s Republic of China. ; Tel: +86 028 85501298; Fax: +86 028 85501295, Yuan Liu, Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8 Street, Miami, FL, 33199, USA ; Tel: 305-348-3628; Fax: 305-348-3772
| | - Zunzhen Zhang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Corresponding authors: Zunzhen Zhang, Ph.D., Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu 610041, People’s Republic of China. ; Tel: +86 028 85501298; Fax: +86 028 85501295, Yuan Liu, Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8 Street, Miami, FL, 33199, USA ; Tel: 305-348-3628; Fax: 305-348-3772
| |
Collapse
|
14
|
Chatterjee A, Chatterji U. All-Trans Retinoic Acid Ameliorates Arsenic-Induced Oxidative Stress and Apoptosis in the Rat Uterus by Modulating MAPK Signaling Proteins. J Cell Biochem 2017; 118:3796-3809. [PMID: 28374919 DOI: 10.1002/jcb.26029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 04/03/2017] [Indexed: 12/19/2022]
Abstract
Exposure to arsenic leads to inhibition of the anti-oxidant defense mechanism of the body. Reactive oxygen species generated in response to arsenic causes reproductive failures in exposed females and also acts as an inducer of apoptosis. As a prospective remedial agent, all-trans retinoic acid (ATRA) was assessed for reversing arsenic-induced oxidative stress and apoptosis. Rats exposed to arsenic for 28 days were allowed to recover naturally or were treated simultaneously with ATRA for 28 days or up to 56 days. Production of H2 O2 was detected using 2',7'-dichlorfluorescein diacetate (DCFCA) by flow cytometry. Catalase, superoxide dismutase, glutathione, ALT, and AST were estimated by biochemical assays and Western blot analyses. Detection of apoptosis was performed using annexin V-FITC/propidium iodide. Expressions of p53, p21, cleaved caspase 3, JNK/pJNK, and ERK/pERK levels were estimated using Western blot analysis. Elemental arsenic deposition in the rat uterus and liver was estimated by atomic absorption spectrophotometry. Our results confirmed that ATRA ameliorated sodium arsenite-induced ROS generation, restored redox balance, and prevented apoptosis. Concomitant recovery was observed to be more prominent for ATRA-treated rats as compared to the rats that were allowed to recover naturally for 56 days. Tissue arsenic deposition was significantly reduced in the uterus upon continuous ATRA treatment. The results revealed that ATRA reversed arsenic-induced free radical generation, activated the anti-oxidant defence system, and subsequently repressed p53-dependent apoptosis through inhibition of the MAPK signaling components. J. Cell. Biochem. 118: 3796-3809, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aniruddha Chatterjee
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Urmi Chatterji
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| |
Collapse
|
15
|
Martin EM, Stýblo M, Fry RC. Genetic and epigenetic mechanisms underlying arsenic-associated diabetes mellitus: a perspective of the current evidence. Epigenomics 2017; 9:701-710. [PMID: 28470093 PMCID: PMC5480787 DOI: 10.2217/epi-2016-0097] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/12/2016] [Indexed: 12/18/2022] Open
Abstract
Chronic exposure to arsenic has been associated with the development of diabetes mellitus (DM), a disease characterized by hyperglycemia resulting from dysregulation of glucose homeostasis. This review summarizes four major mechanisms by which arsenic induces diabetes, namely inhibition of insulin-dependent glucose uptake, pancreatic β-cell damage, pancreatic β-cell dysfunction and stimulation of liver gluconeogenesis that are supported by both in vivo and in vitro studies. Additionally, the role of polymorphic variants associated with arsenic toxicity and disease susceptibility, as well as epigenetic modifications associated with arsenic exposure, are considered in the context of arsenic-associated DM. Taken together, in vitro, in vivo and human genetic/epigenetic studies support that arsenic has the potential to induce DM phenotypes and impair key pathways involved in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Elizabeth M. Martin
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Miroslav Stýblo
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum of Toxicology, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Curriculum of Toxicology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Ameer SS, Engström K, Hossain MB, Concha G, Vahter M, Broberg K. Arsenic exposure from drinking water is associated with decreased gene expression and increased DNA methylation in peripheral blood. Toxicol Appl Pharmacol 2017; 321:57-66. [PMID: 28242323 DOI: 10.1016/j.taap.2017.02.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/03/2017] [Accepted: 02/22/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Exposure to inorganic arsenic increases the risk of cancer and non-malignant diseases. Inefficient arsenic metabolism is a marker for susceptibility to arsenic toxicity. Arsenic may alter gene expression, possibly by altering DNA methylation. OBJECTIVES To elucidate the associations between arsenic exposure, gene expression, and DNA methylation in peripheral blood, and the modifying effects of arsenic metabolism. METHODS The study participants, women from the Andes, Argentina, were exposed to arsenic via drinking water. Arsenic exposure was assessed as the sum of arsenic metabolites in urine (U-As), using high performance liquid-chromatography hydride-generation inductively-coupled-plasma-mass-spectrometry, and arsenic metabolism efficiency was assessed by the urinary fractions (%) of the individual metabolites. Genome-wide gene expression (N=80 women) and DNA methylation (N=93; 80 overlapping with gene expression) in peripheral blood were measured using Illumina DirectHyb HumanHT-12 v4.0 and Infinium Human-Methylation 450K BeadChip, respectively. RESULTS U-As concentrations, ranging 10-1251μg/L, was associated with decreased gene expression: 64% of the top 1000 differentially expressed genes were down-regulated with increasing U-As. U-As was also associated with hypermethylation: 87% of the top 1000CpGs were hypermethylated with increasing U-As. The expression of six genes and six individual CpG sites were significantly associated with increased U-As concentration. Pathway analyses revealed enrichment of genes related to cell death and cancer. The pathways differed somewhat depending on arsenic metabolism efficiency. We found no overlap between arsenic-related gene expression and DNA methylation for individual genes. CONCLUSIONS Increased arsenic exposure was associated with lower gene expression and hypermethylation in peripheral blood, but with no evident overlap.
Collapse
Affiliation(s)
- Syeda Shegufta Ameer
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Karin Engström
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden; Institute of Environmental Medicine, Unit of Metals & Health, Karolinska Institutet, Stockholm, Sweden
| | - Mohammad Bakhtiar Hossain
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Gabriela Concha
- Science Department, Risk Benefit Assessment Unit, National Food Agency, Uppsala, Sweden
| | - Marie Vahter
- Institute of Environmental Medicine, Unit of Metals & Health, Karolinska Institutet, Stockholm, Sweden
| | - Karin Broberg
- Institute of Environmental Medicine, Unit of Metals & Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|