1
|
Xu ZH, Tang XL, Huang HX, Lai SM, Li HM, Liao DQ, Du LY, Xiong ZY, Kuang L, Zhang BY, Chen HJ, Han JY, Li ZH. Association between Blood Trihalomethane Concentrations and the Risk of Dyslipidemia: Nationally Representative Cross-Sectional Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:4292-4303. [PMID: 39993058 DOI: 10.1021/acs.est.4c11066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Experimental studies have indicated that exposure to disinfection byproducts (DBPs) may affect the lipid synthesis. However, epidemiological evidence is sparse. This study aimed to investigate the relationship between blood trihalomethane (THM) exposure and the risk of dyslipidemia from the National Health and Nutrition Examination Survey 2005-2018. We included adults (n = 16,159) whose blood concentrations of chloroform (TCM), bromodichloromethane (BDCM), dibromochloromethane (DBCM), and bromoform (TBM) were quantified. Multivariable logistic regression revealed significantly elevated dyslipidemia risks in the highest group of TCM [adjusted odds ratios (ORs) = 1.30, 95% confidence intervals (CIs): 1.15-1.47], BDCM (1.23,1.07-1.42), TBM (1.35, 1.08-1.68), and total THMs (TTHMs; sum of all four THMs) (1.30, 1.14-1.48). In subgroup analysis, these associations were stronger among young/middle-aged individuals. In Weighted Quantile Sum regression and Quantile G-Computation analysis, combined THM exposure was positively associated with dyslipidemia risk. Our study provides new evidence of a positive association between blood THM concentrations and the risk of dyslipidemia, underscoring the need for further research to confirm these findings and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Zi-Hao Xu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xu-Lian Tang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hong-Xuan Huang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shu-Min Lai
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hong-Min Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Dan-Qing Liao
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Li-Ying Du
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhi-Yuan Xiong
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Ling Kuang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Bing-Yun Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hao-Jie Chen
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jia-Yin Han
- Department of Hospital Infection Control, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, Guangdong, China
| | - Zhi-Hao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
2
|
Fang Z, Zhou B, Zheng G, Chen X, Liu M, Zhang H, He F, Chen H, Hao G. Environment-wide association study of cardiovascular and all-cause mortality: Analysis of the National Health and Nutrition Examination Survey, 1999-2018. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125770. [PMID: 39894157 DOI: 10.1016/j.envpol.2025.125770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Environmental toxicants are increasingly suspected to influence cardiovascular (CV) and all-cause mortality, but previous studies mostly focused on one or a few chemicals. We examined the associations of a wide array of environmental chemicals with CV and all-cause mortality using an exposome-wide approach and the potential mediating role of inflammation in these associations. Data from the National Health and Nutrition Examination Survey (1999-2018) were randomly 60:40 split into a training set and a test set. The mortality rates were determined by the National Center for Health Statistics through a process of correlation with the National Death Index records. Based on the 10th revision of (ICD-10) codes, deaths due to heart disease (ICD 100-I09, I11, I13, and I20-I51) or cerebrovascular disease (I60-I69) were defined as CV mortality. Using the NHANES data, with a median 9.42-year follow-up period, we found that higher concentrations of 2-hydroxynaphthalene (2-NAP) and 2-hydroxyfluorene (2-FLU) in the urine, heavy metal cadmium (Cd), and cotinine in the blood were associated with increased risks of both CV and all-cause mortality. We further found 11 chemicals, including polycyclic aromatic hydrocarbons (1-hydroxynaphthalene, 3-hydroxyfluorene, and 9-hydroxyfluorene), 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) in the urine; hydroxycotinine, heavy metals lead, volatile organic compounds (benzene, ethylbenzene, styrene, toluene and 2,5-Dimethylfuran) in the blood were positively associated with all-cause mortality. Furthermore, we found that C-reactive protein levels partially mediate those associations. In summary, exposure to certain environmental chemicals was associated with CV and all-cause mortality, and C-reactive protein plays a mediation role in those associations. Our findings provided more evidence for preventing and controlling important environmental chemicals to improve people's health.
Collapse
Affiliation(s)
- Zhenger Fang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Biying Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Guangjun Zheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xia Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Mingliang Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Haofeng Zhang
- Department of Epidemiology and Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fudong He
- Department of Epidemiology and Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haiyan Chen
- Department of Parasitic Disease and Endemic Disease Control and Prevention, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Guang Hao
- Department of Epidemiology and Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
3
|
Jiang Z, Yang L, Liu Q, Qiu M, Chen Y, Teng M, Zhang Y, Liu X, Zhao Z, Zheng Y, Andersen M, Qu W. Haloacetamides exacerbate non-alcoholic fatty liver disease induced by a high-fat diet in C57BL/6J mice. Toxicol Sci 2025; 204:57-69. [PMID: 39689017 DOI: 10.1093/toxsci/kfae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024] Open
Abstract
Obesity, a significant global health issue, heightens the risk of non-alcoholic fatty liver disease (NAFLD). Its interaction with environmental pollutants might exacerbate NAFLD's severity. Haloacetamides (HAcAms), a group of emerging nitrogenous disinfection byproducts (DBPs) and potent oxidative stressors, are found in chlorinated drinking water. Since oxidative stress is associated with HAcAms-DBP cytotoxicity and a key factor in NAFLD pathogenesis, we hypothesize that HAcAms-DBPs could exacerbate liver injury and NAFLD, particularly with high-fat diets. This study examined HAcAms-DBPs' impact on liver lipid metabolism in mice treated with 1 to 100 times the background drinking water level (13.05 µg/L) for up to 16 weeks of oral administration. Compared to a high-fat-only group, mice co-exposed to a high-fat diet and HAcAms-DBPs for 16 weeks had elevated serum alanine transaminase, aspartate transaminase, triglyceride, hepatic lipid aggregation, and inflammation response. Under high-fat conditions, background drinking water levels of HAcAms significantly upregulated liver Acetyl-CoA carboxylase 1, fatty acid synthase, peroxisome proliferator-activated receptor gamma (PPARγ), PPARγ coactivator-1α, glucose transporter 1 and 4 protein expression in C57BL/6J mice; 10 times background significantly increased expression of inflammatory marker tumor necrosis factor and liver fibrosis marker protein alpha-smooth muscle actin; 100 times further increased both liver damage and markers of early non-alcoholic steatohepatitis phenotypes like steatosis and lobular inflammation. HAcAms-DBPs plus high-fat conditions worsened liver damage. The possible health risks of NAFLD induced by HAcAms in obese individuals deserve further study.
Collapse
Affiliation(s)
- Zhiqiang Jiang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Lili Yang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Qinxin Liu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Meiyue Qiu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yu Chen
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Mengying Teng
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- Key Laboratory of the Public Health Safety, Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xing Liu
- Key Laboratory of the Public Health Safety, Ministry of Education, Department of Epidemiology, School of Public Health, Fudan University, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Melvin Andersen
- ScitoVation LLC, Research Triangle Park, NC 27713, United States
| | - Weidong Qu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Zhao X, Ding H, Qin J, An S, Li S, He H, Zhou L, Gong X, Chu X. Assessing the Co-Exposure Patterns of Volatile Organic Compounds and the Risk of Hyperuricemia: An Analysis of the National Health and Nutrition Examination Survey 2003-2012. TOXICS 2024; 12:772. [PMID: 39590952 PMCID: PMC11598210 DOI: 10.3390/toxics12110772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Co-exposure to multiple volatile organic compounds (VOCs) is common in daily life. However, few studies have evaluated the associations between the patterns of simultaneous exposure to multiple VOCs and the risk of hyperuricemia. METHODS This study included 7490 adults from the National Health and Nutrition Examination Survey conducted between 2003 and 2012. The K-means clustering method was applied to cluster eight kinds of VOCs in the blood into various co-exposure patterns, including benzene, bromodichloromethane, chloroform, dibromochloromethane, 1,4-dichlorobenzene, ethylbenzene, methyl tertiary-butyl ether (MTBE), and o-xylene. Binary logistic regression analysis was utilized to assess the association between single VOCs, the co-exposure patterns of multiple VOCs, and the hyperuricemia risk. Restricted cubic spline functions were utilized to investigate the non-linear relationships. RESULTS Based on eight VOCs, four characteristic co-exposure patterns were generated. Compared with the low-level exposure group, the levels of volatile organic compound (VOC) co-exposure in cluster 2, characterized by relatively high levels of MTBE and moderate levels of bromodichloromethane, chloroform, and dibromochloromethane, were associated with increased hyperuricemia risk, with an odds ratio of 1.32 (1.02, 1.71). Increasing levels of bromodichloromethane and chloroform were significantly associated with an increased risk of hyperuricemia. A strong J-shaped relationship was found between MTBE and hyperuricemia. CONCLUSIONS This study indicated that blood bromodichloromethane and chloroform were positively associated with hyperuricemia risk. Blood MTBE had a J-shaped association with hyperuricemia. In addition, the significant association of the co-exposure patterns of multiple VOCs in the blood with hyperuricemia risk was observed. Changing VOC co-exposure patterns may play a crucial role in the occurrence of hyperuricemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xia Chu
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China; (X.Z.)
| |
Collapse
|
5
|
Jha D, Bakker ENTP, Kumar R. Mechanistic and therapeutic role of NLRP3 inflammasome in the pathogenesis of Alzheimer's disease. J Neurochem 2024; 168:3574-3598. [PMID: 36802053 DOI: 10.1111/jnc.15788] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/20/2023]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, has emerged as the most common form of dementia in the elderly. Several pathological hallmarks have been identified, including neuroinflammation. A comprehensive insight into the underlying mechanisms that can fuel the development of novel therapeutic approaches is necessary because of the alarmingly rapid increase in the frequency of incidence. Recently, NLRP3 inflammasome was identified as a critical mediator of neuroinflammation. Activation of nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome by amyloid, neurofibrillary tangles, impaired autophagy and endoplasmic reticulum stress, triggers the release of pro-inflammatory cytokines such as IL-1β and IL-18. Subsequently, these cytokines can promote neurodegeneration and cognitive impairment. It is well established that genetic or pharmacological ablation of NLRP3 alleviates AD-related pathological features in in vitro and in vivo models. Therefore, several synthetic and natural compounds have been identified that exhibit the potential to inhibit NLRP3 inflammasome and alleviate AD-associated pathology. The current review article will highlight the various mechanisms by which activation of NLRP3 inflammation occurs during Alzheimer's disease, and how it influences neuroinflammation, neurodegeneration and cognitive impairment. Moreover, we will summarise the different small molecules that possess the potential to inhibit NLRP3 and can pave the path for developing novel therapeutic interventions for AD.
Collapse
Affiliation(s)
- Dhanshree Jha
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centre, Location University of Amsterdam, and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| | - Rahul Kumar
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centre, Location University of Amsterdam, and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Jiang Z, Yang L, Liu Q, Qiu M, Chen Y, Qu F, Crabbe MJC, Wang H, Andersen ME, Zheng Y, Qu W. Haloacetamides disinfection by-products, a potential risk factor for nonalcoholic fatty liver disease. WATER RESEARCH 2024; 261:122008. [PMID: 38944971 DOI: 10.1016/j.watres.2024.122008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/21/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic disorder characterized by abnormal lipid deposition, with oxidative stress being a risk factor in its onset and progression. Haloacetamides (HAcAms), as unregulated disinfection by-products in drinking water, may alter the incidence and severity of NAFLD through the production of oxidative stress. We explored whether HAcAms at 1, 10, and 100-fold concentrations in Shanghai drinking water perturbed lipid metabolism in normal human liver LO-2 cells. CRISPR/Cas9 was used to construct a LO-2 line with stable NRF2 knock-down (NRF2-KD) to investigate the mechanism underlying abnormal lipid accumulation and hepatocyte damage caused by mixed exposure to HAcAms. At 100-fold real-world concentration, HAcAms caused lipid deposition and increased triglyceride accumulation in LO-2 cells, consistent with altered de novo lipogenesis. Differences in responses to HAcAms in normal and NRF2-KD LO-2 cells indicated that HAcAms caused hepatocyte lipid deposition and triglyceride accumulation by activation of the NRF2/PPARγ pathway and aggravated liver cell toxicity by inducing ferroptosis. These results indicate that HAcAms are important risk factors for NAFLD. Further observations and verifications of the effect of HAcAms on NAFLD in the population are warranted in the future.
Collapse
Affiliation(s)
- Zhiqiang Jiang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China
| | - Lili Yang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China
| | - Qinxin Liu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China
| | - Meiyue Qiu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China
| | - Yu Chen
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China
| | - Fei Qu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China
| | - M James C Crabbe
- Wolfson College, Oxford University, Oxford OX2 6UD, United Kingdom
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States
| | - Melvin E Andersen
- ScitoVation LLC. 6 Davis Drive, Suite 146, Research Triangle Park, NC 27713, United States
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, No.308 Ningxia Road, Qingdao 266071, China
| | - Weidong Qu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University Shanghai, 200032, China.
| |
Collapse
|
7
|
Yang L, Jiang Z, Yang L, Zheng W, Chen Y, Qu F, Crabbe MJC, Zhang Y, Andersen ME, Zheng Y, Qu W. Disinfection Byproducts of Haloacetaldehydes Disrupt Hepatic Lipid Metabolism and Induce Lipotoxicity in High-Fat Culture Conditions. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:12356-12367. [PMID: 38953388 DOI: 10.1021/acs.est.3c11009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Unhealthy lifestyles, obesity, and environmental pollutants are strongly correlated with the development of nonalcoholic fatty liver disease (NAFLD). Haloacetaldehyde-associated disinfection byproducts (HAL-DBPs) at various multiples of concentrations found in finished drinking water together with high-fat (HF) were examined to gauge their mixed effects on hepatic lipid metabolism. Using new alternative methods (NAMs), studying effects in human cells in vitro for risk assessment, we investigated the combined effects of HF and HAL-DBPs on hepatic lipid metabolism and lipotoxicity in immortalized LO-2 human hepatocytes. Coexposure of HAL-DBPs at various multiples of environmental exposure levels with HF increased the levels of triglycerides, interfered with de novo lipogenesis, enhanced fatty acid oxidation, and inhibited the secretion of very low-density lipoproteins. Lipid accumulation caused by the coexposure of HAL-DBPs and HF also resulted in more severe lipotoxicity in these cells. Our results using an in vitro NAM-based method provide novel insights into metabolic reprogramming in hepatocytes due to coexposure of HF and HAL-DBPs and strongly suggest that the risk of NAFLD in sensitive populations due to HAL-DBPs and poor lifestyle deserves further investigation both with laboratory and epidemiological tools. We also discuss how results from our studies could be used in health risk assessments for HAL-DBPs.
Collapse
Affiliation(s)
- Lili Yang
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Zhiqiang Jiang
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Lan Yang
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Weiwei Zheng
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yu Chen
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Fei Qu
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - M James C Crabbe
- Wolfson College, Oxford University, Oxford OX2 6UD, United Kingdom
- Institute of Biomedical and Environmental Science & Technology, University of Bedfordshire, Luton LU1 3JU, U.K
| | - Yubin Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Melvin E Andersen
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Research Triangle Park, North Carolina 27713, United States
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China
| | - Weidong Qu
- Key Laboratory of Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| |
Collapse
|
8
|
Qiu M, Yang L, Jiang Z, Chen Y, Liu Q, Wang X, Qu W. Mixed exposure to haloacetaldehyde disinfection by-products exacerbates lipid aggregation in the liver of mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 350:123971. [PMID: 38641033 DOI: 10.1016/j.envpol.2024.123971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Haloacetaldehyde disinfection by-products (HAL-DBPs) are among the top three unregulated DBPs found in drinking water. The cytotoxicity and genotoxicity of HALs are much higher than that of the regulated trihalomethanes and haloacetic acids. Previous studies have mainly focused on the toxic effects of single HAL, with few examining the toxic effects of mixed exposures to HALs. The study aimed to observe the effects of mixed exposures of 1∼1000X the realistic level of HALs on the hepatotoxicity and lipid metabolism of C57BL/6J mice, based on the component and concentration of HALs detected in the finished water of Shanghai. Exposure to realistic levels of HALs led to a significant increase in phosphorated acetyl CoA carboxylase 1 (p-ACC1) in the hepatic de novo lipogenesis (DNL) pathway. Additionally, exposure to 100X realistic levels of HALs resulted in significant alterations to key enzymes of DNL pathway, including ACC1, fatty acid synthase (FAS), and diacylglycerol acyltransferase 2 (DGAT2), as well as key proteins of lipid disposal such as carnitine palmitoyltransferase 1 (CPT-1) and peroxisome proliferator activated receptor α (PPARα). Exposure to 1000X realistic levels of HALs significantly increased hepatic and serum triglyceride levels, as well as total cholesterol, low-density lipoprotein, alanine aminotransferase, aspartate transaminase, alkaline phosphatase, and lactate dehydrogenase levels, significantly decreased high-density lipoprotein. Meanwhile, histopathological analysis demonstrated that HALs exacerbated tissue vacuolization and inflammatory cell infiltration in mice livers, which showed the typical phenotypes of non-alcoholic fatty liver disease (NAFLD). These results suggested that the HALs mixture is a critical risk factor for NAFLD and is significantly highly toxic to C57BL/6J mice.
Collapse
Affiliation(s)
- Meiyue Qiu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Lili Yang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Zhiqiang Jiang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Yu Chen
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Qinxin Liu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Xia Wang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Weidong Qu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Deng YL, Lu TT, Hao H, Liu C, Yuan XQ, Miao Y, Zhang M, Zeng JY, Li YF, Lu WQ, Zeng Q. Association between Urinary Haloacetic Acid Concentrations and Liver Injury among Women: Results from the Tongji Reproductive and Environmental (TREE) Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:17006. [PMID: 38261302 PMCID: PMC10805132 DOI: 10.1289/ehp13386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Experimental studies have shown that disinfection byproducts (DBPs) including haloacetic acids (HAAs) can cause liver toxicity, but evidence linking this association in humans is sparse. OBJECTIVES We aimed to explore the associations between HAA exposures and liver injury. METHODS We included 922 women between December 2018 and January 2020 from the Tongji Reproductive and Environmental (TREE) cohort study in Wuhan, China. Urinary HAA concentrations including trichloroacetic acid (TCAA) and dichloroacetic acid (DCAA) and serum indicators of liver function, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyltransferase (GGT) were measured. Liver injury was defined as if any of serum indicator levels were above the 90th percentile. Multivariate logistic and linear regression models were fitted to assess the associations of urinary HAA concentrations with the risk of liver injury and liver function indicators. Stratified analyses by age, body mass index (BMI), alcohol use, and passive smoking were also applied to evaluate the potential effect modifiers. RESULTS There is little evidence of associations of urinary TCAA concentrations with liver injury risk and liver function indicators. However, urinary DCAA concentrations were associated with a higher risk of liver injury [odds ratios (OR) for 1-interquartile range (IQR) increase in natural log (ln) transformed DCAA concentrations: 1.45; 95% confidence interval (CI): 1.07, 1.98]. This association was observed only among nondrinkers (p interaction = 0.058 ). We also found that a 1-IQR increase in ln-transformed DCAA concentrations was positively associated with ALT levels (percentage change = 6.06 % ; 95% CI: 0.48%, 11.95%) and negatively associated with AST/ALT (percentage change = - 4.48 % ; 95% CI: - 7.80 % , - 1.04 % ). In addition, urinary DCAA concentrations in relation to higher GGT levels was observed only among passive smokers (p interaction = 0.040 ). CONCLUSION Our findings suggest that exposure to DCAA but not TCAA is associated with liver injury among women undergoing assisted reproductive technology. https://doi.org/10.1289/EHP13386.
Collapse
Affiliation(s)
- Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Ting-Ting Lu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hua Hao
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Chong Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Qiong Yuan
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Miao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jia-Yue Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu-Feng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen-Qing Lu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
10
|
Roy S, Saha P, Bose D, Trivedi A, More M, Xiao S, Diehl AM, Chatterjee S. Hepatic NLRP3-Derived Hsp70 Binding to TLR4 Mediates MASLD to MASH Progression upon Inhibition of PP2A by Harmful Algal Bloom Toxin Microcystin, a Second Hit. Int J Mol Sci 2023; 24:16354. [PMID: 38003543 PMCID: PMC10671242 DOI: 10.3390/ijms242216354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Harmful algal bloom toxin microcystin has been associated with metabolic dysfunction-associated steatotic liver disease (MASLD) progression and hepatocellular carcinoma, though the mechanisms remain unclear. Using an established mouse model of MASLD, we show that the NLRP3-Hsp70-TLR4 axis drives in part the inflammation of the liver lobule that results in the progression of MASLD to metabolic dysfunction-associated steatohepatitis (MASH). Results showed that mice deficient in NLRP3 exhibited decreased MASH pathology, blocked Hsp70 expression, and co-binding with NLRP3, a crucial protein component of the liver inflammasome. Hsp70, both in the liver lobule and extracellularly released in the liver vasculature, acted as a ligand to TLR4 in the liver, primarily in hepatocytes to activate the NF-κB pathway, ultimately leading to hepatic cell death and necroptosis, a crucial pathology of MASH progression. The above studies show a novel insight into an inflammasome-triggered Hsp70-mediated inflammation that may have broader implications in MASLD pathology. MASLD to MASH progression often requires multiple hits. One of the mediators of progressive MASLD is environmental toxins. In this research report, we show for the first time a novel mechanism where microcystin-LR, an environmental toxin, advances MASLD to MASH by triggering the release of Hsp70 as a DAMP to activate TLR4-induced inflammation in the liver.
Collapse
Affiliation(s)
- Subhajit Roy
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (S.R.); (P.S.); (D.B.); (A.T.)
| | - Punnag Saha
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (S.R.); (P.S.); (D.B.); (A.T.)
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (S.R.); (P.S.); (D.B.); (A.T.)
| | - Ayushi Trivedi
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (S.R.); (P.S.); (D.B.); (A.T.)
| | - Madhura More
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (S.R.); (P.S.); (D.B.); (A.T.)
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, NC 27710, USA;
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (S.R.); (P.S.); (D.B.); (A.T.)
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Liu S, Liu J, Wu Y, Tan L, Luo Y, Ding C, Tang Z, Shi X, Fan W, Song S. Genistein upregulates AHR to protect against environmental toxin-induced NASH by inhibiting NLRP3 inflammasome activation and reconstructing antioxidant defense mechanisms. J Nutr Biochem 2023; 121:109436. [PMID: 37666477 DOI: 10.1016/j.jnutbio.2023.109436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
We have previously proven that the environmental toxin could accelerate the development and progression of nonalcoholic steatohepatitis (NASH). However, the underlying mechanism associated with such excessive inflammation hasn't been fully illustrated. Although Genistein has been well accepted for its capability in anti-inflammation and anti-oxidation, its effect in ameliorating contaminants-induced NASH still needs to be identified. In this study, using chickens and primary chicken hepatocytes as models, we found that NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome were over-activated in bromoacetic acid (BAA, one of the typical environmental toxins)-induced NASH, characterized by the infiltration of inflammatory cell, and the increase of NLRP3, Caspase-1 p20, and cytokines (IL-1β, IL-18) expressions. Interestingly, genistein treatment could recover these changes, with the signs of restored activities of anti-oxidases, decreased expressions of NLRP3 inflammasome components, and increased levels of elements in phase I metabolic system. The detailed mechanism was that, via up-regulating aryl hydrocarbon receptor (AHR), genistein lifted mRNA levels of Cyp1-related genes to reconstruct cytochrome P450 (CYP450) systems, and the raised AHR negatively regulated NLRP3 inflammasome activity to relieve inflammation. More important, the interaction and co-localization between AHR and NLRP3 was first proved, and genistein could promote the levels of AHR that interacted with NLRP3, which thereafter blocked the activation of NLRP3 inflammasome. Conclusively, in this research, we confirmed the AHR-dependent protective role of genistein in environmental toxin-linked NASH, which shed light on the potential precautions for contaminants-induced NASH.
Collapse
Affiliation(s)
- Shuhui Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
| | - Jiwen Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
| | - Yuting Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
| | - Lei Tan
- Administration for Market Regulation of Guangdong Province Key Laboratory of Supervision for Edible Agricultural Products, Shenzhen Centre of Inspection and Testing for Agricultural Products, Shenzhen, 518000, China
| | - Yan Luo
- Administration for Market Regulation of Guangdong Province Key Laboratory of Supervision for Edible Agricultural Products, Shenzhen Centre of Inspection and Testing for Agricultural Products, Shenzhen, 518000, China
| | - Chenchen Ding
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
| | - Zhihui Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
| | - Xizhi Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China.
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China.
| |
Collapse
|
12
|
Chatterjee S, More M. Cyanobacterial Harmful Algal Bloom Toxin Microcystin and Increased Vibrio Occurrence as Climate-Change-Induced Biological Co-Stressors: Exposure and Disease Outcomes via Their Interaction with Gut-Liver-Brain Axis. Toxins (Basel) 2023; 15:289. [PMID: 37104227 PMCID: PMC10144574 DOI: 10.3390/toxins15040289] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
The effects of global warming are not limited to rising global temperatures and have set in motion a complex chain of events contributing to climate change. A consequence of global warming and the resultant climate change is the rise in cyanobacterial harmful algal blooms (cyano-HABs) across the world, which pose a threat to public health, aquatic biodiversity, and the livelihood of communities that depend on these water systems, such as farmers and fishers. An increase in cyano-HABs and their intensity is associated with an increase in the leakage of cyanotoxins. Microcystins (MCs) are hepatotoxins produced by some cyanobacterial species, and their organ toxicology has been extensively studied. Recent mouse studies suggest that MCs can induce gut resistome changes. Opportunistic pathogens such as Vibrios are abundantly found in the same habitat as phytoplankton, such as cyanobacteria. Further, MCs can complicate human disorders such as heat stress, cardiovascular diseases, type II diabetes, and non-alcoholic fatty liver disease. Firstly, this review describes how climate change mediates the rise in cyanobacterial harmful algal blooms in freshwater, causing increased levels of MCs. In the later sections, we aim to untangle the ways in which MCs can impact various public health concerns, either solely or in combination with other factors resulting from climate change. In conclusion, this review helps researchers understand the multiple challenges brought forth by a changing climate and the complex relationships between microcystin, Vibrios, and various environmental factors and their effect on human health and disease.
Collapse
Affiliation(s)
- Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, University of California–Irvine, Irvine, CA 92697, USA
- Toxicology Core, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental and Occupational Health, Program in Public Health, University of California–Irvine, Irvine, CA 92697, USA
- Division of Infectious Disease, Department of Medicine, UCI School of Medicine, University of California–Irvine, Irvine, CA 92697, USA
| | - Madhura More
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, University of California–Irvine, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Urinary trihalomethane concentrations and liver function indicators: a cross-sectional study in China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:39724-39732. [PMID: 36596971 DOI: 10.1007/s11356-022-25072-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023]
Abstract
While it is known that exposure to disinfection by-products (DBPs), including trihalomethanes (THMs), impairs liver function, few epidemiological studies have explored this association. Here, we determined the concentrations of four urinary trihalomethanes (chloroform [TCM], and three Br-THMs, bromodichloromethane [BDCM], dibromochloromethane [DBCM], and bromoform [TBM]), and nine serum liver function indicators in 182 adults ≥ 18 years of age, examined at a medical examination center in Wuxi, China, in 2020 and 2021. Generalized linear model analysis revealed positive associations between urinary DBCM and alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), total protein (TP), and albumin (ALB). Urinary Br-THMs and total THMs (TTHMs) were positively associated with ALT, AST, TBIL, indirect bilirubin (IBIL), TP, and ALB (all P < 0.05). Urinary THMs were not associated with alkaline phosphatase (ALP) or glutamine transaminase (GGT) (all P > 0.05). Generalized additive model-based penalized regression splines were used to confirm these associations. In conclusion, THM exposure was associated with altered serum biomarkers of liver function.
Collapse
|
14
|
Kim HY, Park CH, Park JB, Ko K, Lee MH, Chung J, Yoo YH. Hepatic STAMP2 alleviates polychlorinated biphenyl-induced steatosis and hepatic iron overload in NAFLD models. ENVIRONMENTAL TOXICOLOGY 2022; 37:2223-2234. [PMID: 35616167 DOI: 10.1002/tox.23589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/03/2022] [Accepted: 05/14/2022] [Indexed: 06/15/2023]
Abstract
Polychlorinated biphenyls (PCBs) have been associated with neurotoxicity, hepatoxicity, oncogenicity, and endocrine-disrupting effects. Although the recent studies have demonstrated that PCB exposure leads to nonalcoholic fatty liver disease (NAFLD), the underlying mechanism has remained unsolved. In this study, we examined the hepatic effects of a PCB mixture, Aroclor 1260, whose composition mimics human bioaccumulation patterns, and PCB 126 in C57BL/6 mice. Male C57Bl/6 mice were fed a standard diet or a 60% high-fat diet and exposed to Aroclor 1260 (10 mg/kg or 20 mg/kg) or PCB 126 (1 mg/kg or 5 mg/kg) by intraperitoneal injection for a total of four injections (2, 3, 4, and 5 weeks) for 6 weeks. In mice, both Aroclor 1260 and PCB 126-induced liver damage, hepatic steatosis and inflammation. We also observed that PCB exposure-induced hepatic iron overload (HIO). We previously demonstrated that hepatic six transmembrane protein of prostate 2 (STAMP2) may represent a suitable therapeutic target for NAFLD patients. Thus, we further examined whether hepatic STAMP2 is involved in PCB-induced NAFLD. We observed that hepatic STAMP2 was significantly decreased in PCB-induced NAFLD models in vivo and in vitro. Furthermore, overexpression of hepatic STAMP2 using an adenoviral delivery system resulted in improvement of PCB-induced steatosis and HIO in vivo and in vitro. Our findings indicate that enhancing hepatic STAMP2 expression represents a potential therapeutic avenue for the treatment of PCB exposure-induced NAFLD.
Collapse
Affiliation(s)
- Hye Young Kim
- Department of Anatomy and Cell Biology and BK21 program, Department of Translational Biomedical Science, Dong-A University College of Medicine, Busan, Republic of Korea
- Department of Oral Microbiology and Oral Genomics Research Center, School of Dentistry, Pusan National University, Busan, Republic of Korea
| | - Chul Hee Park
- Department of Anatomy and Cell Biology and BK21 program, Department of Translational Biomedical Science, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Joon Beom Park
- Department of Anatomy and Cell Biology and BK21 program, Department of Translational Biomedical Science, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Kangeun Ko
- Department of Anatomy and Cell Biology and BK21 program, Department of Translational Biomedical Science, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Mi Hwa Lee
- Department of Anatomy and Cell Biology and BK21 program, Department of Translational Biomedical Science, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Jin Chung
- Department of Oral Microbiology and Oral Genomics Research Center, School of Dentistry, Pusan National University, Busan, Republic of Korea
| | - Young Hyun Yoo
- Department of Anatomy and Cell Biology and BK21 program, Department of Translational Biomedical Science, Dong-A University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
15
|
Diurnal Variation in Biomarkers of Exposure to Endocrine-Disrupting Chemicals and Their Association with Oxidative Damage in Norwegian Adults: The EuroMix Study. TOXICS 2022; 10:toxics10040181. [PMID: 35448442 PMCID: PMC9028082 DOI: 10.3390/toxics10040181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/27/2022] [Accepted: 04/02/2022] [Indexed: 11/23/2022]
Abstract
Much evidence on the adverse health effects of endocrine-disrupting chemicals (EDCs) has accumulated during recent decades. EDCs are commonly found in various foods and personal care products (PCP). Data documenting a diurnally varying EDC metabolism in humans is scarce. This study examined (i) the time-of-day effect on the diurnal magnitude and variance of urinary biomarkers of exposure to EDCs, and (ii) the association between EDC exposures and oxidative damage in a Norwegian adult subpopulation. This was a cross-sectional panel study using biobanked samples from the EuroMix project. During a typical weekday, participants were asked to collect all day’s urine voids and record dietary and PCP habitual uses in a diary. Collected time stamps of urine voids were classified into three distinct periods in the day (morning 6 a.m.−12 p.m., mid-day 12 p.m.−6 p.m., evening 6 p.m.−6 a.m.). Questionnaires regarding demographic characteristics, personal care product usage, and dietary habits were completed. Urinary levels of EDCs (phthalates, parabens, and bisphenols) were measured using mass spectrometry and adjusted for urinary volume using specific gravity. Urinary 4-hydroxynonenal (4HNE), a lipid peroxidation marker, was measured using an immunoassay kit. Linear mixed-effect models identified EDCs under the influence of a diurnal variation effect that was adjusted for dietary habits and PCP use and examined associations between EDC and 4HNE. p-values were FDR-adjusted. Most phthalates appeared to be diurnally varying with higher urinary levels towards the evening (q < 0.001) than those measured during mid-day; this strong diurnal variation effect was not present for parabens and bisphenols. Significant (q < 0.001) positive associations were observed between all phthalates, parabens, and bisphenols (except bisphenol S) and 4HNE. This study’s findings highlighted the diurnal variation of excretion for certain EDC, but not for others, in real-life conditions. The degree of EDC chronotoxicity in distinct diurnal windows of the day warrants further investigation with longitudinal human studies.
Collapse
|
16
|
Wang ZX, Wang MY, Yang RX, Zhao ZH, Xin FZ, Li Y, Ren TY, Fan JG. Ammonia Scavenger Restores Liver and Muscle Injury in a Mouse Model of Non-alcoholic Steatohepatitis With Sarcopenic Obesity. Front Nutr 2022; 9:808497. [PMID: 35369074 PMCID: PMC8970101 DOI: 10.3389/fnut.2022.808497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/18/2022] [Indexed: 12/25/2022] Open
Abstract
Recent studies have revealed that sarcopenia is closely associated with obesity and non-alcoholic steatohepatitis (NASH). However, few attempted to explore the cause-and-effect relationship between sarcopenic obesity and NASH. In this study, we investigated muscular alterations in a rodent NASH model to elucidate their intrinsic relations and explore the potential therapeutic target. Forty-six 8-week-old and twenty 42-week-old male C57BL/6 mice (defined as young and middle-aged mice, respectively) were fed with a high-fat diet (HFD) for 12 or 20 weeks. A subset of young mice was subjected to ammonia lowering treatment by L-ornithine L-aspartate (LOLA). We examined body composition and muscle strength by nuclear magnetic resonance and grip strength meter, respectively. At the end of the 12th week, all HFD-fed mice developed typical steatohepatitis. Meanwhile, sarcopenia occurred in HFD-fed middle-aged mice, whereas young mice only demonstrated decreased grip strength. Until the end of week 20, young mice in the HFD group exhibited significant sarcopenia and obesity phenotypes, including decreased lean body mass and grip strength, and increased body fat mass and percentage body fat. Additionally, plasma ammonia level was markedly increased in HFD-fed mice of both ages at week 20. Plasma ammonia level was negatively associated with muscle strength and myofiber diameter in young mice. LOLA can significantly reduce plasma levels of ammonia, alanine aminotransaminase, aspartate aminotransaminase, and cholesterol in mice fed an HFD. Hepatic infiltration of inflammatory cells and collagen deposition area were significantly decreased in HFD group by LOLA treatment. Meanwhile, LOLA significantly increased lean body mass, grip strength, and average muscle fiber diameter of HFD-fed mice. These findings suggest that the occurrence of NASH precedes sarcopenia in HFD mice, and the steatohepatitis-related hyperammonemia might contribute to the pathogenesis of sarcopenia. LOLA might be an effective drug for both steatohepatitis and sarcopenic obesity.
Collapse
Affiliation(s)
- Zi-Xuan Wang
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng-Yu Wang
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Xu Yang
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Hua Zhao
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Feng-Zhi Xin
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Li
- Chinese Academy of Science (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tian-Yi Ren
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
17
|
Chen H, Li G, Chan YL, Zhang HE, Gorrell MD, Pollock CA, Saad S, Oliver BG. Differential Effects of 'Vaping' on Lipid and Glucose Profiles and Liver Metabolic Markers in Obese Versus Non-obese Mice. Front Physiol 2021; 12:755124. [PMID: 34803738 PMCID: PMC8599937 DOI: 10.3389/fphys.2021.755124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Tobacco smoking increases the risk of metabolic disorders due to the combination of harmful chemicals, whereas pure nicotine can improve glucose tolerance. E-cigarette vapour contains nicotine and some of the harmful chemicals found in cigarette smoke at lower levels. To investigate how e-vapour affects metabolic profiles, male Balb/c mice were exposed to a high-fat diet (HFD, 43% fat, 20kJ/g) for 16weeks, and e-vapour in the last 6weeks. HFD alone doubled fat mass and caused dyslipidaemia and glucose intolerance. E-vapour reduced fat mass in HFD-fed mice; only nicotine-containing e-vapour improved glucose tolerance. In chow-fed mice, e-vapour increased lipid content in both blood and liver. Changes in liver metabolic markers may be adaptive responses rather than causal. Future studies can investigate how e-vapour differentially affects metabolic profiles with different diets.
Collapse
Affiliation(s)
- Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Gerard Li
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Yik Lung Chan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Hui Emma Zhang
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mark D. Gorrell
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Carol A. Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Sonia Saad
- Renal Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Brian G. Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Zheng S, Yang Y, Wen C, Liu W, Cao L, Feng X, Chen J, Wang H, Tang Y, Tian L, Wang X, Yang F. Effects of environmental contaminants in water resources on nonalcoholic fatty liver disease. ENVIRONMENT INTERNATIONAL 2021; 154:106555. [PMID: 33857709 DOI: 10.1016/j.envint.2021.106555] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 06/12/2023]
Abstract
The global prevalence of nonalcoholic fatty liver disease (NAFLD) has been increasing rapidly in recent years, which is now estimated to be over 25%. NAFLD is one of the most common chronic liver diseases in the world. At present, with the rapid development of economy and industrialization, many chemicals are released into the environment. These chemical contaminants in the environment might cause harm to human health and result in lipid metabolism disorder during long-term exposure. Moreover, the incentive of many NAFLD cases is unknown, and the environmental risk factors of NAFLD need to be urgently identified. Hence, we focus on the impacts of several popular environmental contaminants in water environment on the development and progression of NAFLD. These contaminants mainly include microcystins (MCs), disinfection by-products (DBPs), heavy metals (HMs), dioxins and polychlorinated biphenyls (PCBs). Through analyzing a great many epidemiological and toxicological studies, we have found positive associations between NAFLD and chronic exposure to these contaminants at the environmental levels. This review may enhance the understanding of liver damage caused by environmental pollutants, which are considered as tangible environmental risk factors for NAFLD.
Collapse
Affiliation(s)
- Shuilin Zheng
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Yue Yang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Cong Wen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Wenya Liu
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Linghui Cao
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Xiangling Feng
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Jihua Chen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Hui Wang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang 421001, China
| | - Yan Tang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang 421001, China
| | - Li Tian
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Fei Yang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China; Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang 421001, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health Southeast University, Nanjing 210009, China.
| |
Collapse
|
19
|
Mondal A, Saha P, Bose D, Chatterjee S, Seth RK, Xiao S, Porter DE, Brooks BW, Scott GI, Nagarkatti M, Nagarkatti P, Chatterjee S. Environmental Microcystin exposure in underlying NAFLD-induced exacerbation of neuroinflammation, blood-brain barrier dysfunction, and neurodegeneration are NLRP3 and S100B dependent. Toxicology 2021; 461:152901. [PMID: 34416350 PMCID: PMC8503918 DOI: 10.1016/j.tox.2021.152901] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been shown to be associated with extrahepatic comorbidities including neuronal inflammation and Alzheimer's-like pathology. Environmental and genetic factors also act as a second hit to modulate severity and are expected to enhance the NAFLD-linked neuropathology. We hypothezied that environmental microcystin-LR (MC-LR), a toxin produced by harmful algal blooms of cyanobacteria, exacerbates the neuroinflammation and degeneration of neurons associated with NAFLD. Using a mouse model of NAFLD, exposed to MC-LR subsequent to the onset of fatty liver, we show that the cyanotoxin could significantly increase proinflammatory cytokine expression in the frontal cortex and cause increased expression of Lcn2 and HMGB1. The above effects were NLRP3 inflammasome activation-dependent since the use of NLRP3 knockout mice abrogated the increase in inflammation. NLRP3 was also responsible for decreased expression of the blood-brain barrier (BBB) tight junction proteins Occludin and Claudin 5 suggesting BBB dysfunction was parallel to neuroinflammation following microcystin exposure. An increased circulatory S100B release, a hallmark of astrocyte activation in MC-LR exposed NAFLD mice also confirmed BBB integrity loss, but the astrocyte activation observed in vivo was NLRP3 independent suggesting an important role of a secondary S100B mediated crosstalk. Mechanistically, conditioned medium from reactive astrocytes and parallel S100B incubation in neuronal cells caused increased inducible NOS, COX-2, and higher BAX/ Bcl2 protein expression suggesting oxidative stress-mediated neuronal cell apoptosis crucial for neurodegeneration. Taken together, MC-LR exacerbated neuronal NAFLD-linked comorbidities leading to cortical inflammation, BBB dysfunction, and neuronal apoptosis.
Collapse
Affiliation(s)
- Ayan Mondal
- Environmental Health and Disease Laboratory, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
| | - Punnag Saha
- Environmental Health and Disease Laboratory, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA; Columbia VA Medical Center, Columbia, SC, 29209, USA
| | - Somdatta Chatterjee
- Environmental Health and Disease Laboratory, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA; Columbia VA Medical Center, Columbia, SC, 29209, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy at Rutgers University, Piscataway, NJ, 08854, USA
| | - Dwayne E Porter
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, 29208, USA
| | - Bryan W Brooks
- Department of Environmental Science, Baylor University, Waco, TX, 76798-7266, USA
| | - Geoff I Scott
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, 29208, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA; Columbia VA Medical Center, Columbia, SC, 29209, USA.
| |
Collapse
|
20
|
Al-Badrani M, Saha P, Mondal A, Seth RK, Sarkar S, Kimono D, Bose D, Porter DE, Scott GI, Brooks B, Raychoudhury S, Nagarkatti M, Nagarkatti P, Chatterjee S. Early microcystin-LR exposure-linked inflammasome activation in mice causes development of fatty liver disease and insulin resistance. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 80:103457. [PMID: 32687983 PMCID: PMC7609636 DOI: 10.1016/j.etap.2020.103457] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/19/2020] [Accepted: 07/14/2020] [Indexed: 05/06/2023]
Abstract
Evidence from pediatric studies show that infants and children are at risk for early exposure to microcystin. The present report tests the hypothesis that early life exposure to microcystin (MC), a principal component of harmful algal blooms followed by a juvenile exposure to high-fat diet feeding potentiate the development of nonalcoholic fatty liver disease phenotype in adulthood. Results showed classical symptoms of early NAFLD linked inflammation. Cytokines and chemokines such as CD68, IL-1β, MCP-1, and TNF-α, as well as α-SMA were increased in the groups that were exposed to MC-LR with the high-fat diet compared to the vehicle group. Also, mechanistically, NLRP3 KO mice showed a significant decrease in the inflammation and NAFLD phenotype and resisted the metabolic changes such as insulin resistance and glucose metabolism in the liver. The data suggested that MC-LR exposure and subsequent NLRP3 inflammasome activation in childhood could impact liver health in juveniles.
Collapse
Affiliation(s)
- Muayad Al-Badrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA; Department of Family and Community Medicine, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Punnag Saha
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Ayan Mondal
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Dwayne E Porter
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Geoff I Scott
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Bryan Brooks
- Department of Environmental Science, Baylor University, Waco, TX 76798-7266, USA
| | - Samir Raychoudhury
- Department of Biology, Chemistry, and Environmental Health Science, Benedict College, Columbia, SC, 29204, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA; NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, USA.
| |
Collapse
|
21
|
Lambert K, Gondeau C, Briolotti P, Scheuermann V, Daujat-Chavanieu M, Aimond F. Biocompatible modified water as a non-pharmaceutical approach to prevent metabolic syndrome features in obesogenic diet-fed mice. Food Chem Toxicol 2020; 141:111403. [PMID: 32387306 DOI: 10.1016/j.fct.2020.111403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 10/24/2022]
Abstract
The prevalence of metabolic syndrome (MetS), elevating cardiovascular risks, is increasing worldwide, with no available global therapeutic options. The intake of plain, mineral or biocompatible modified waters was shown to prevent some MetS features. This study was designed to analyze, in mice fed a high fat and sucrose diet (HFSD), the effects on MetS features of the daily intake of a reverse osmosed, weakly remineralized, water (OW) and of an OW dynamized by a physical processing (ODW), compared to tap water (TW). The HFSD was effective at inducing major features of MetS such as obesity, hepatic steatosis and inflammation, blood dyslipidemia, systemic glucose intolerance and muscle insulin resistance. Compared to TW, OW intake decreased hepatic fibrosis and inflammation, and mitigated hepatic steatosis and dyslipidemia. ODW intake further improved skeletal muscle insulin sensitivity and systemic glucose tolerance. This study highlights the deleterious metabolic impacts of the daily intake of TW, in combination with a high energy diet, and its possible involvement in MetS prevalence increase. In addition, it demonstrates that biocompatible modified water may be promising non-pharmaceutical, cost-effective tools for nutritional approaches in the treatment of MetS.
Collapse
Affiliation(s)
- Karen Lambert
- PhyMedExp, Université Montpellier, INSERM, CNRS, France
| | - Claire Gondeau
- IRMB, Université Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | | | | | | | - Franck Aimond
- PhyMedExp, Université Montpellier, INSERM, CNRS, France.
| |
Collapse
|
22
|
Sarkar S, Alhasson F, Kimono D, Albadrani M, Seth RK, Xiao S, Porter DE, Scott GI, Brooks B, Nagarkatti M, Nagarkatti P, Chatterjee S. Microcystin exposure worsens nonalcoholic fatty liver disease associated ectopic glomerular toxicity via NOX-2-MIR21 axis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 73:103281. [PMID: 31706246 PMCID: PMC7100051 DOI: 10.1016/j.etap.2019.103281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 05/03/2023]
Abstract
NAFLD often results in cardiovascular, intestinal and renal complications. Previous reports from our laboratory highlighted NAFLD induced ectopic inflammatory manifestations in the kidney that gave rise to glomerular inflammation. Extending our studies, we hypothesized that existing inflammatory conditions in NAFLD could make the kidneys more susceptible to environmental toxicity. Our results showed that exposure of Microcystin-LR (MC) in NAFLD mice caused a marked increase in cellular scarring with a concomitant increase in mesangial cell activation as observed by increased α-SMA in the extracellular matrix surrounding the glomeruli. Renal tissue surrounding the glomeruli also showed increased NOX2 activation as shown by greater co-localization of p47 Phox and its membrane component gp91Phox both in the mesangial cell and surrounding tissue. Mechanistically, mesangial cells incubated with apocynin, nitrone spin trap DMPO and miR21 inhibitor showed significantly decreased α-SMA, miR21 levels and proinflammatory cytokine release in the supernatant. In parallel, mice lacking miR21, known to be activated by NOX2, when exposed to MC in NAFLD showed decreased mesangial cell activation. Strikingly, phenyl boronic acid incubated cells that were exposed to MC showed significantly decreased mesangial cell activation showing that peroxynitrite might be the major reactive species involved in mediation of the activation process, release of proinflammatory micro RNAs and cytokines that are crucial for renal toxicity. Thus, in conclusion, MC exposure causes NOX2 activation that leads to mesangial cell activation and toxicity via release of peroxynitrite that also represses PTEN by the upregulation of miR21 thus amplifying the toxicity.
Collapse
Affiliation(s)
- Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, United States; NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, United States; NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Muayad Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, United States; NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, United States; NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Shuo Xiao
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Dwayne E Porter
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Geoff I Scott
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States
| | - Bryan Brooks
- Department of Environmental Science, Baylor University, United States
| | - Mitzi Nagarkatti
- Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, United States
| | - Prakash Nagarkatti
- Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, United States; NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, United States.
| |
Collapse
|
23
|
Albadrani M, Seth RK, Sarkar S, Kimono D, Mondal A, Bose D, Porter DE, Scott GI, Brooks B, Raychoudhury S, Nagarkatti M, Nagarkatti P, Jule Y, Diehl AM, Chatterjee S. Exogenous PP2A inhibitor exacerbates the progression of nonalcoholic fatty liver disease via NOX2-dependent activation of miR21. Am J Physiol Gastrointest Liver Physiol 2019; 317:G408-G428. [PMID: 31393787 PMCID: PMC6842990 DOI: 10.1152/ajpgi.00061.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an emerging global pandemic. Though significant progress has been made in unraveling the pathophysiology of the disease, the role of protein phosphatase 2A (PP2A) and its subsequent inhibition by environmental and genetic factors in NAFLD pathophysiology remains unclear. The present report tests the hypothesis that an exogenous PP2A inhibitor leads to hepatic inflammation and fibrogenesis via an NADPH oxidase 2 (NOX2)-dependent pathway in NAFLD. Results showed that microcystin (MC) administration, a potent PP2A inhibitor found in environmental exposure, led to an exacerbation of NAFLD pathology with increased CD68 immunoreactivity, the release of proinflammatory cytokines, and stellate cell activation, a process that was attenuated in mice that lacked the p47phox gene and miR21 knockout mice. Mechanistically, leptin-primed immortalized Kupffer cells (a mimicked model for an NAFLD condition) treated with apocynin or nitrone spin trap 5,5 dimethyl-1- pyrroline N-oxide (DMPO) had significantly decreased CD68 and decreased miR21 and α-smooth muscle actin levels, suggesting the role of NOX2-dependent reactive oxygen species in miR21-induced Kupffer cell activation and stellate cell pathology. Furthermore, NOX2-dependent peroxynitrite generation was primarily responsible for cellular events observed following MC exposure since incubation with phenylboronic acid attenuated miR21 levels, Kupffer cell activation, and inflammatory cytokine release. Furthermore, blocking of the AKT pathway attenuated PP2A inhibitor-induced NOX2 activation and miR21 upregulation. Taken together, we show that PP2A may have protective roles, and its inhibition exacerbates NAFLD pathology via activating NOX2-dependent peroxynitrite generation, thus increasing miR21-induced pathology.NEW & NOTEWORTHY Protein phosphatase 2A inhibition causes nonalcoholic steatohepatitis (NASH) progression via NADPH oxidase 2. In addition to a novel emchanism of action, we describe a new tool to describe NASH histopathology.
Collapse
Affiliation(s)
- Muayad Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
- Department of Family and Community Medicine, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Ayan Mondal
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Dwayne E Porter
- Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Geoff I Scott
- Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Bryan Brooks
- Department of Environmental Science, Baylor University, Waco, Texas
| | - Samir Raychoudhury
- Departments of Biology, Chemistry, and Environmental Health Science, Benedict College, Columbia, South Carolina
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina
| | | | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, North Carolina
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
24
|
Li G, Chan YL, Sukjamnong S, Anwer AG, Vindin H, Padula M, Zakarya R, George J, Oliver BG, Saad S, Chen H. A Mitochondrial Specific Antioxidant Reverses Metabolic Dysfunction and Fatty Liver Induced by Maternal Cigarette Smoke in Mice. Nutrients 2019; 11:nu11071669. [PMID: 31330878 PMCID: PMC6682890 DOI: 10.3390/nu11071669] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/07/2019] [Accepted: 07/18/2019] [Indexed: 11/16/2022] Open
Abstract
Maternal smoking leads to glucose and lipid metabolic disorders and hepatic damage in the offspring, potentially due to mitochondrial oxidative stress. Mitoquinone mesylate (MitoQ) is a mitochondrial targeted antioxidant with high bioavailability. This study aimed to examine the impact of maternal cigarette smoke exposure (SE) on offspring’s metabolic profile and hepatic damage, and whether maternal MitoQ supplementation during gestation can affect these changes. Female Balb/c mice (eight weeks) were either exposed to air or SE for six weeks prior to mating and throughout gestation and lactation. A subset of the SE dams were supplied with MitoQ in the drinking water (500 µmol/L) during gestation and lactation. Intraperitoneal glucose tolerance test was performed in the male offspring at 12 weeks and the livers and plasma were collected at 13 weeks. Maternal SE induced glucose intolerance, hepatic steatosis, mitochondrial oxidative stress and related damage in the adult offspring. Maternal MitoQ supplementation reduced hepatic mitochondrial oxidative stress and improved markers of mitophagy and mitochondrial biogenesis. This may restore hepatic mitochondrial health and was associated with an amelioration of glucose intolerance, hepatic steatosis and pathological changes induced by maternal SE. MitoQ supplementation may potentially prevent metabolic dysfunction and hepatic pathology induced by intrauterine SE.
Collapse
Affiliation(s)
- Gerard Li
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Yik Lung Chan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Suporn Sukjamnong
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Pathum Wan, Bangkok 10330, Thailand
| | - Ayad G Anwer
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Howard Vindin
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Matthew Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Razia Zakarya
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, Sydney, NSW 2037, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Sonia Saad
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, The University of Sydney, Sydney, NSW 2065, Australia
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia.
- Faculty of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| |
Collapse
|
25
|
Arman T, Lynch KD, Montonye ML, Goedken M, Clarke JD. Sub-Chronic Microcystin-LR Liver Toxicity in Preexisting Diet-Induced Nonalcoholic Steatohepatitis in Rats. Toxins (Basel) 2019; 11:E398. [PMID: 31323923 PMCID: PMC6669744 DOI: 10.3390/toxins11070398] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023] Open
Abstract
Microcystin-LR (MCLR) is a hepatotoxic cyanotoxin reported to cause a phenotype similar to nonalcoholic steatohepatitis (NASH). NASH is a common progressive liver disease that advances in severity due to exogenous stressors such as poor diet and toxicant exposure. Our objective was to determine how sub-chronic MCLR toxicity affects preexisting diet-induced NASH. Sprague-Dawley rats were fed one of three diets for 10 weeks: control, methionine and choline deficient (MCD), or high fat/high cholesterol (HFHC). After six weeks of diet, animals received vehicle, 10 µg/kg, or 30 µg/kg MCLR via intraperitoneal injection every other day for the final 4 weeks. Incidence and severity scoring of histopathology endpoints suggested that MCLR toxicity drove NASH to a less fatty and more fibrotic state. In general, expression of genes involved in de novo lipogenesis and fatty acid esterification were altered in favor of decreased steatosis. The higher MCLR dose increased expression of genes involved in fibrosis and inflammation in the control and HFHC groups. These data suggest MCLR toxicity in the context of preexisting NASH may drive the liver to a more severe phenotype that resembles burnt-out NASH.
Collapse
Affiliation(s)
- Tarana Arman
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Katherine D Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Michelle L Montonye
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Michael Goedken
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08901, USA
| | - John D Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
26
|
Sarkar S, Kimono D, Albadrani M, Seth RK, Busbee P, Alghetaa H, Porter DE, Scott GI, Brooks B, Nagarkatti M, Nagarkatti P, Chatterjee S. Environmental microcystin targets the microbiome and increases the risk of intestinal inflammatory pathology via NOX2 in underlying murine model of Nonalcoholic Fatty Liver Disease. Sci Rep 2019; 9:8742. [PMID: 31217465 PMCID: PMC6584534 DOI: 10.1038/s41598-019-45009-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
With increased climate change pressures likely to influence harmful algal blooms, exposure to microcystin, a known hepatotoxin and a byproduct of cyanobacterial blooms can be a risk factor for NAFLD associated comorbidities. Using both in vivo and in vitro experiments we show that microcystin exposure in NAFLD mice cause rapid alteration of gut microbiome, rise in bacterial genus known for mediating gut inflammation and lactate production. Changes in the microbiome were strongly associated with inflammatory pathology in the intestine, gut leaching, tight junction protein alterations and increased oxidative tyrosyl radicals. Increased lactate producing bacteria from the altered microbiome was associated with increased NOX-2, an NADPH oxidase isoform. Activationof NOX2 caused inflammasome activation as shown by NLRP3/ASCII and NLRP3/Casp-1 colocalizations in these cells while use of mice lacking a crucial NOX2 component attenuated inflammatory pathology and redox changes. Mechanistically, NOX2 mediated peroxynitrite species were primary to inflammasome activation and release of inflammatory mediators. Thus, in conclusion, microcystin exposure in NAFLD could significantly alter intestinal pathology especially by the effects on microbiome and resultant redox status thus advancing our understanding of the co-existence of NAFLD-linked inflammatory bowel disease phenotypes in the clinic.
Collapse
Affiliation(s)
- Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
| | - Muayad Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
| | - Philip Busbee
- Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, USA
| | - Hasan Alghetaa
- Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, USA
| | - Dwayne E Porter
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
| | - Geoff I Scott
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA
| | - Bryan Brooks
- Department of Environmental Science, Baylor University, Waco, USA
| | - Mitzi Nagarkatti
- Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, USA
| | - Prakash Nagarkatti
- Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA.
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, USA.
| |
Collapse
|
27
|
Clarke JD, Dzierlenga A, Arman T, Toth E, Li H, Lynch KD, Tian DD, Goedken M, Paine MF, Cherrington N. Nonalcoholic fatty liver disease alters microcystin-LR toxicokinetics and acute toxicity. Toxicon 2019; 162:1-8. [PMID: 30849452 PMCID: PMC6447445 DOI: 10.1016/j.toxicon.2019.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 02/27/2019] [Accepted: 03/03/2019] [Indexed: 02/08/2023]
Abstract
Microcystin-LR (MCLR) is a cyanotoxin produced by blue-green algae that causes liver and kidney toxicities. MCLR toxicity is dependent on cellular uptake through the organic anion transporting polypeptide (OATP) transporters. Nonalcoholic fatty liver disease (NAFLD) progresses through multiple stages, alters expression of hepatic OATPs, and is associated with chronic kidney disease. The purpose of this study was to determine whether NAFLD increases systemic exposure to MCLR and influences acute liver and kidney toxicities. Rats were fed a control diet or two dietary models of NAFLD; methionine and choline deficient (MCD) or high fat/high cholesterol (HFHC). Two studies were performed in these groups: 1) a single dose intravenous toxicokinetic study (20 μg/kg), and 2) a single dose intraperitoneal toxicity study (60 μg/kg). Compared to control rats, plasma MCLR area under the concentration-time curve (AUC) in MCD rats doubled, whereas biliary clearance (Clbil) was unchanged; in contrast, plasma AUC in HFHC rats was unchanged, whereas Clbil approximately doubled. Less MCLR bound to PP2A was observed in the liver of MCD rats. This shift in exposure decreased the severity of liver pathology only in the MCD rats after a single toxic dose of MCLR (60 μg/kg). In contrast, the single toxic dose of MCLR increased hepatic inflammation, plasma cholesterol, proteinuria, and urinary KIM1 in HFHC rats more than MCLR exposed control rats. In conclusion, rodent models of NAFLD alter MCLR toxicokinetics and acute toxicity and may have implications for liver and kidney pathologies in NAFLD patients.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
| | - Anika Dzierlenga
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Tarana Arman
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Erica Toth
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Hui Li
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Katherine D Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Dan-Dan Tian
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Michael Goedken
- Rutgers Translational Sciences, Rutgers University, Piscataway, NJ, 08901, USA
| | - Mary F Paine
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Nathan Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
28
|
Abdelmegeed MA, Ha SK, Choi Y, Akbar M, Song BJ. Role of CYP2E1 in Mitochondrial Dysfunction and Hepatic Injury by Alcohol and Non-Alcoholic Substances. Curr Mol Pharmacol 2019; 10:207-225. [PMID: 26278393 DOI: 10.2174/1874467208666150817111114] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/17/2022]
Abstract
Alcoholic fatty liver disease (AFLD) and non-alcoholic fatty liver disease (NAFLD) are two pathological conditions that are spreading worldwide. Both conditions are remarkably similar with regard to the pathophysiological mechanism and progression despite different causes. Oxidative stressinduced mitochondrial dysfunction through post-translational protein modifications and/or mitochondrial DNA damage has been a major risk factor in both AFLD and NAFLD development and progression. Cytochrome P450-2E1 (CYP2E1), a known important inducer of oxidative radicals in the cells, has been reported to remarkably increase in both AFLD and NAFLD. Interestingly, CYP2E1 isoforms expressed in both endoplasmic reticulum (ER) and mitochondria, likely lead to the deleterious consequences in response to alcohol or in conditions of NAFLD after exposure to high fat diet (HFD) and in obesity and diabetes. Whether CYP2E1 in both ER and mitochondria work simultaneously or sequentially in various conditions and whether mitochondrial CYP2E1 may exert more pronounced effects on mitochondrial dysfunction in AFLD and NAFLD are unclear. The aims of this review are to briefly describe the role of CYP2E1 and resultant oxidative stress in promoting mitochondrial dysfunction and the development or progression of AFLD and NAFLD, to shed a light on the function of the mitochondrial CYP2E1 as compared with the ER-associated CYP2E1. We finally discuss translational research opportunities related to this field.
Collapse
Affiliation(s)
- Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892. United States
| | - Seung-Kwon Ha
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane, Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD. United States
| | - Youngshim Choi
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane, Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD. United States
| | - Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane, Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD. United States
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane, Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD. United States
| |
Collapse
|
29
|
Dattaroy D, Seth RK, Sarkar S, Kimono D, Albadrani M, Chandrashekaran V, Al Hasson F, Singh UP, Fan D, Nagarkatti M, Nagarkatti P, Diehl AM, Chatterjee S. Sparstolonin B (SsnB) attenuates liver fibrosis via a parallel conjugate pathway involving P53-P21 axis, TGF-beta signaling and focal adhesion that is TLR4 dependent. Eur J Pharmacol 2018; 841:33-48. [PMID: 30194936 PMCID: PMC7193950 DOI: 10.1016/j.ejphar.2018.08.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023]
Abstract
SsnB previously showed a promising role to lessen liver inflammation observed in a mouse model of NAFLD. Since NAFLD can progress to fibrosis, studies were designed to unravel its role in attenuating NAFLD associated fibrosis. Using both in vivo and in vitro approaches, the study probed the possible mechanisms that underlined the role of SsnB in mitigating fibrosis. Mechanistically, SsnB, a TLR4 antagonist, decreased TLR4-PI3k akt signaling by upregulating PTEN protein expression. It also decreased MDM2 protein activation and increased p53 and p21 gene and protein expression. SsnB also downregulated pro-fibrogenic hedgehog signaling pathway, inhibited hepatic stellate cell proliferation and induced apoptosis in hepatic stellate cells, a mechanism that was LPS dependent. Further, SsnB decreased fibrosis by antagonizing TLR4 induced TGFβ signaling pathway. Alternatively, SsnB augmented BAMBI (a TGFβ pseudo-receptor) expression in mice liver by inhibiting TLR4 signaling pathway and thus reduced TGFβ signaling, resulting in decreased hepatic stellate cell activation and extracellular matrix deposition. In vitro experiments on human hepatic stellate cell line showed that SsnB increased gene and protein expression of BAMBI. It also decreased nuclear co-localization of phospho SMAD2/3 and SMAD4 protein and thus attenuated TGFβ signaling in vitro. We also observed a significant decrease in phosphorylation of SMAD2/3 protein, decreased STAT3 activation, alteration of focal adhesion protein and stress fiber disassembly upon SsnB administration in hepatic stellate cells which further confirmed the antagonistic effect of SsnB on TLR4-induced fibrogenesis.
Collapse
Affiliation(s)
- Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Muayad Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Varun Chandrashekaran
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Firas Al Hasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Udai P. Singh
- Department of Pathology Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, USC, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, United States
| | - Prakash Nagarkatti
- Department of Pathology Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, United States
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham 27707, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States.
| |
Collapse
|
30
|
Guo J, Wang C, Guo Z, Zuo Z. Exposure to environmental level phenanthrene induces a NASH-like phenotype in new born rat. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 239:261-271. [PMID: 29656250 DOI: 10.1016/j.envpol.2018.04.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/25/2018] [Accepted: 04/06/2018] [Indexed: 06/08/2023]
Abstract
More and more evidence indicates that persistent organic pollutants (POPs) are a risk factor for non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Phenanthrene (Phe) is a kind of POP which existed extensively in the environment, but whose toxicity on mammals has so far received less focus. Subcutaneously injection of Phe (0.5, 5, 50 μg/kg) for 21 days induced significant NAFLD/NASH symptoms in new born rats. Exposure to environmental levels of Phe decreased body weight and liver-somatic index; impaired histology of liver; influenced the peroxisome proliferator-activated receptor gamma (PPARγ) signaling and lipid metabolism in liver; stimulated oxidative stress in the rats' liver; induced the variation of NFκB pathway and liver inflammatory response; and caused liver fibrosis via transforming growth factor β1 (tgfβ1). We speculated that the subcutaneously injected Phe was transferred to the liver through blood circulation, which may have induced the elevation of PPARγ directly or indirectly, leading to liver steatosis. Excess lipid, acting as the first hit, stimulated the second hit factors - oxidative stress, inflammatory response and lipid peroxidation, and finally resulted in steatohepatitis and liver fibrosis.
Collapse
Affiliation(s)
- Jiaojiao Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361005, China
| | - Zhizhun Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
31
|
Seth RK, Kimono D, Alhasson F, Sarkar S, Albadrani M, Lasley SK, Horner R, Janulewicz P, Nagarkatti M, Nagarkatti P, Sullivan K, Chatterjee S. Increased butyrate priming in the gut stalls microbiome associated-gastrointestinal inflammation and hepatic metabolic reprogramming in a mouse model of Gulf War Illness. Toxicol Appl Pharmacol 2018; 350:64-77. [PMID: 29751049 PMCID: PMC6121708 DOI: 10.1016/j.taap.2018.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 01/07/2023]
Abstract
Most of the associated pathologies in Gulf War Illness (GWI) have been ascribed to chemical and pharmaceutical exposures during the war. Since an increased number of veterans complain of gastrointestinal (GI), neuroinflammatory and metabolic complications as they age and there are limited options for a cure, the present study was focused to assess the role of butyrate, a short chain fatty acid for attenuating GWI-associated GI and metabolic complications. Results in a GWI-mouse model of permethrin and pyridostigmine bromide (PB) exposure showed that oral butyrate restored gut homeostasis and increased GPR109A receptor copies in the small intestine (SI). Claudin-2, a protein shown to be upregulated in conditions of leaky gut was significantly decreased following butyrate administration. Butyrate decreased TLR4 and TLR5 expressions in the liver concomitant to a decrease in TLR4 activation. GW-chemical exposure showed no clinical signs of liver disease but a significant alteration of metabolic markers such as SREBP1c, PPAR-α, and PFK was evident. Liver markers for lipogenesis and carbohydrate metabolism that were significantly upregulated following GW chemical exposure were attenuated by butyrate priming in vivo and in human primary hepatocytes. Further, Glucose transporter Glut-4 that was shown to be elevated following liver complications were significantly decreased in these mice after butyrate administration. Finally, use of TLR4 KO mice completely attenuated the liver metabolic changes suggesting the central role of these receptors in the GWI pathology. In conclusion, we report a butyrate specific mechanistic approach to identify and treat increased metabolic abnormalities in GWI veterans with systemic inflammation, chronic fatigue, GI disturbances, metabolic complications and weight gain.
Collapse
Affiliation(s)
- Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Muayad Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Stephen K Lasley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Ronnie Horner
- Department of Health Services Policy and Management, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Patricia Janulewicz
- Department of Environmental Health, Boston University School of Public Health, Boston University, Boston, MA, USA
| | - Mitzi Nagarkatti
- Department of Pathology Microbiology and Immunology, USC School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology Microbiology and Immunology, USC School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Kimberly Sullivan
- Department of Environmental Health, Boston University School of Public Health, Boston University, Boston, MA, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
32
|
Alhasson F, Seth RK, Sarkar S, Kimono DA, Albadrani MS, Dattaroy D, Chandrashekaran V, Scott GI, Raychoudhury S, Nagarkatti M, Nagarkatti P, Diehl AM, Chatterjee S. High circulatory leptin mediated NOX-2-peroxynitrite-miR21 axis activate mesangial cells and promotes renal inflammatory pathology in nonalcoholic fatty liver disease. Redox Biol 2018; 17:1-15. [PMID: 29660503 PMCID: PMC6006523 DOI: 10.1016/j.redox.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
High circulatory insulin and leptin followed by underlying inflammation are often ascribed to the ectopic manifestations in non-alcoholic fatty liver disease (NAFLD) but the exact molecular pathways remain unclear. We have shown previously that CYP2E1-mediated oxidative stress and circulating leptin in NAFLD is associated with renal disease severity. Extending the studies, we hypothesized that high circulatory leptin in NAFLD causes renal mesangial cell activation and tubular inflammation via a NOX2 dependent pathway that upregulates proinflammatory miR21. High-fat diet (60% kcal) was used to induce fatty liver phenotype with parallel insulin and leptin resistance. The kidneys were probed for mesangial cell activation and tubular inflammation that showed accelerated NASH phenotype and oxidative stress in the liver. Results showed that NAFLD kidneys had significant increases in α-SMA, a marker of mesangial cell activation, miR21 levels, tyrosine nitration and renal inflammation while they were significantly decreased in leptin and p47 phox knockout mice. Micro RNA21 knockout mice showed decreased tubular immunotoxicity and proinflammatory mediator release. Mechanistically, use of NOX2 siRNA or apocynin,phenyl boronic acid (FBA), DMPO or miR21 antagomir inhibited leptin primed-miR21-mediated mesangial cell activation in vitro suggesting a direct role of leptin-mediated NOX-2 in miR21-mediated mesangial cell activation. Finally, JAK-STAT inhibitor completely abrogated the mesangial cell activation in leptin-primed cells suggesting that leptin signaling in the mesangial cells depended on the JAK-STAT pathway. Taken together the study reports a novel mechanistic pathway of leptin-mediated renal inflammation that is dependent on NOX-2-miR21 axis in ectopic manifestations underlying NAFLD-induced co-morbidities.
Collapse
Affiliation(s)
- Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Diana A Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Muayad S Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Varun Chandrashekaran
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Geoffrey I Scott
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Samir Raychoudhury
- Department of Biology, Chemistry and Environmental Health Science, Benedict College, Columbia, SC 29204, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, NC 27707, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
33
|
Gängler S, Charisiadis P, Seth R, Chatterjee S, Makris KC. Time of the day dictates the variability of biomarkers of exposure to disinfection byproducts. ENVIRONMENT INTERNATIONAL 2018; 112:33-40. [PMID: 29247841 DOI: 10.1016/j.envint.2017.12.013] [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: 09/19/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 06/07/2023]
Abstract
Non-persistent environmental chemicals (NOPEC) are xenobiotics with short half-lives of elimination (<7h). Similar to chronopharmacokinetics, NOPEC metabolism may follow diurnal patterns of cytochrome P450 activity. The role of circadian liver clock in shaping NOPEC metabolism and their concomitant measurements of biomarkers of exposure and effect remains poorly understood in real-life human settings. Metabolic activation (toxication) by CYP2E1 converts trihalomethanes (THM) to harmful metabolites. We investigated the diurnal variation of urinary THM exposures and their metabolism patterns as catalyzed by CYP2E1 redox activity, using the surrogate marker of 4-hydroxynonenal (4HNE). We implemented three time-series trials with adult volunteers conducting specific household cleaning activities at predefined times of a day. Circadia variation of 4HNE was assessed with a cosinor model and its mesor levels increased with THM exposure. The time of exposure within the day dictated the magnitude of urinary THM levels and their toxication effect; in all three trials and relative to urinary THM levels before the activity, lower and higher median THM were measured right after the activity in morning and afternoon/night, respectively. This is consistent with higher reported CYP2E1 redox activity in light/active phase. Population health studies should incorporate time-stamped biomarker data to improve the understanding of chronic disease processes.
Collapse
Affiliation(s)
- Stephanie Gängler
- Water and Health Laboratory, Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol 3041, Cyprus
| | - Pantelis Charisiadis
- Water and Health Laboratory, Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol 3041, Cyprus
| | - Ratanesh Seth
- Environmental Health Sciences Dept., University of South Carolina, United States
| | - Saurabh Chatterjee
- Environmental Health Sciences Dept., University of South Carolina, United States
| | - Konstantinos C Makris
- Water and Health Laboratory, Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol 3041, Cyprus.
| |
Collapse
|
34
|
Narsale AA, Puppa MJ, Hardee JP, VanderVeen BN, Enos RT, Murphy EA, Carson JA. Short-term pyrrolidine dithiocarbamate administration attenuates cachexia-induced alterations to muscle and liver in ApcMin/+ mice. Oncotarget 2018; 7:59482-59502. [PMID: 27449092 PMCID: PMC5312326 DOI: 10.18632/oncotarget.10699] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/09/2016] [Indexed: 12/19/2022] Open
Abstract
Cancer cachexia is a complex wasting condition characterized by chronic inflammation, disrupted energy metabolism, and severe muscle wasting. While evidence in pre-clinical cancer cachexia models have determined that different systemic inflammatory inhibitors can attenuate several characteristics of cachexia, there is a limited understanding of their effects after cachexia has developed, and whether short-term administration is sufficient to reverse cachexia-induced signaling in distinctive target tissues. Pyrrolidine dithiocarbamate (PDTC) is a thiol compound having anti-inflammatory and antioxidant properties which can inhibit STAT3 and nuclear factor κB (NF-κB) signaling in mice. This study examined the effect of short-term PDTC administration to ApcMin/+ mice on cachexia-induced disruption of skeletal muscle protein turnover and liver metabolic function. At 16 weeks of age ApcMin/+ mice initiating cachexia (7% BW loss) were administered PDTC (10mg/kg bw/d) for 2 weeks. Control ApcMin/+ mice continued to lose body weight during the treatment period, while mice receiving PDTC had no further body weight decrease. PDTC had no effect on either intestinal tumor burden or circulating IL-6. In muscle, PDTC rescued signaling disrupting protein turnover regulation. PDTC suppressed the cachexia induction of STAT3, increased mTORC1 signaling and protein synthesis, and suppressed the induction of Atrogin-1 protein expression. Related to cachectic liver metabolic function, PDTC treatment attenuated glycogen and lipid content depletion independent to the activation of STAT3 and mTORC1 signaling. Overall, these results demonstrate short-term PDTC treatment to cachectic mice attenuated cancer-induced disruptions to muscle and liver signaling, and these changes were independent to altered tumor burden and circulating IL-6.
Collapse
Affiliation(s)
- Aditi A Narsale
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA
| | - Melissa J Puppa
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA
| | - Justin P Hardee
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA
| | - Brandon N VanderVeen
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology & Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | - E Angela Murphy
- Department of Pathology, Microbiology & Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | - James A Carson
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
35
|
Altered gut microbiome in a mouse model of Gulf War Illness causes neuroinflammation and intestinal injury via leaky gut and TLR4 activation. PLoS One 2017; 12:e0172914. [PMID: 28328972 PMCID: PMC5362211 DOI: 10.1371/journal.pone.0172914] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/10/2017] [Indexed: 02/06/2023] Open
Abstract
Many of the symptoms of Gulf War Illness (GWI) that include neurological abnormalities, neuroinflammation, chronic fatigue and gastrointestinal disturbances have been traced to Gulf War chemical exposure. Though the association and subsequent evidences are strong, the mechanisms that connect exposure to intestinal and neurological abnormalities remain unclear. Using an established rodent model of Gulf War Illness, we show that chemical exposure caused significant dysbiosis in the gut that included increased abundance of phylum Firmicutes and Tenericutes, and decreased abundance of Bacteroidetes. Several gram negative bacterial genera were enriched in the GWI-model that included Allobaculum sp. Altered microbiome caused significant decrease in tight junction protein Occludin with a concomitant increase in Claudin-2, a signature of a leaky gut. Resultant leaching of gut caused portal endotoxemia that led to upregulation of toll like receptor 4 (TLR4) activation in the small intestine and the brain. TLR4 knock out mice and mice that had gut decontamination showed significant decrease in tyrosine nitration and inflammatory mediators IL1β and MCP-1 in both the small intestine and frontal cortex. These events signified that gut dysbiosis with simultaneous leaky gut and systemic endotoxemia-induced TLR4 activation contributes to GW chemical-induced neuroinflammation and gastrointestinal disturbances.
Collapse
|
36
|
Cytochrome P450-2E1 promotes fast food-mediated hepatic fibrosis. Sci Rep 2017; 7:39764. [PMID: 28051126 PMCID: PMC5209674 DOI: 10.1038/srep39764] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023] Open
Abstract
Cytochrome P450-2E1 (CYP2E1) increases oxidative stress. High hepatic cholesterol causes non-alcoholic steatohepatitis (NASH) and fibrosis. Thus, we aimed to study the role of CYP2E1 in promoting liver fibrosis by high cholesterol-containing fast-food (FF). Male wild-type (WT) and Cyp2e1-null mice were fed standard chow or FF for 2, 12, and 24 weeks. Various parameters of liver fibrosis and potential mechanisms such as oxidative and endoplasmic reticulum (ER) stress, inflammation, and insulin resistance (IR) were studied. Indirect calorimetry was also used to determine metabolic parameters. Liver histology showed that only WT fed FF (WT-FF) developed NASH and fibrosis. Hepatic levels of fibrosis protein markers were significantly increased in WT-FF. The nitroxidative stress marker iNOS, but not CYP2E1, was significantly elevated only in FF-fed WT. Serum endotoxin, TLR-4 levels, and inflammatory markers were highest in WT-FF. FAS, PPAR-α, PPAR-γ, and CB1-R were markedly altered in WT-FF. Electron microscopy and immunoblot analyses showed significantly higher levels of ER stress in FF-fed WT. Indirect calorimetry showed that Cyp2e1-null-mice fed FF exhibited consistently higher total energy expenditure (TEE) than their corresponding WT. These results demonstrate that CYP2E1 is important in fast food-mediated liver fibrosis by promoting nitroxidative and ER stress, endotoxemia, inflammation, IR, and low TEE.
Collapse
|
37
|
Seth RK, Das S, Dattaroy D, Chandrashekaran V, Alhasson F, Michelotti G, Nagarkatti M, Nagarkatti P, Diehl AM, Bell PD, Liedtke W, Chatterjee S. TRPV4 activation of endothelial nitric oxide synthase resists nonalcoholic fatty liver disease by blocking CYP2E1-mediated redox toxicity. Free Radic Biol Med 2017; 102:260-273. [PMID: 27913210 PMCID: PMC5989309 DOI: 10.1016/j.freeradbiomed.2016.11.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 01/11/2023]
Abstract
NAFLD is a clinically progressive disease with steatosis, inflammation, endothelial dysfunction and fibrosis being the stages where clinical intervention becomes necessary. Lack of early biomarkers and absence of a FDA approved drug obstructs efforts for effective treatment. NAFLD progression is strongly linked to a balance between liver injury, tissue regeneration and the functioning of endogenous defense mechanisms. The failure of the defense pathways to resist the tissue damage arising from redox stress, one of the "multiple hits" in disease progression, give rise to heightened inflammation and occasional fibrosis. We introduce an endogenous defense mechanism in the liver that is mediated by TRPV4, a transient receptor potential calcium-permeable ion channel that responds to the cytotoxic liver environment and negatively regulates CYP2E1, a cytochrome p450 enzyme. Using Trpv4-/- mice and cultured primary cells, we show that TRPV4 is activated both by damage associated molecular pattern HMGB1 and collagen in diseased Kupffer cells that in turn activate the endothelial NOS (NOS3) to release nitric oxide (NO). The diffusible NO acts in a paracrine fashion in neighboring hepatocytes to deactivate the redox toxicity induced by CYP2E1. We also find that CYP2E1-mediated TRPV4 repression in late stages causes an unrestricted progression of disease. Thus, TRPV4 functions as a sensor of cell stress in the diseased fatty liver and constitutes an endogenous defense molecule, a novel concept with potential for therapeutic approaches against NAFLD, perhaps also against hepatic drug toxicity in general.
Collapse
Affiliation(s)
- Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Varun Chandrashekaran
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | | | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, NC 27707, USA
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Wolfgang Liedtke
- Department of Neurology, Duke University School of Medicine, Durham, NC 27707, USA.
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
38
|
Faustino-Rocha AI, Rodrigues D, da Costa RG, Diniz C, Aragão S, Talhada D, Botelho M, Colaço A, Pires MJ, Peixoto F, Oliveira PA. Trihalomethanes in liver pathology: Mitochondrial dysfunction and oxidative stress in the mouse. ENVIRONMENTAL TOXICOLOGY 2016; 31:1009-1016. [PMID: 25640707 DOI: 10.1002/tox.22110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 12/18/2014] [Accepted: 12/21/2014] [Indexed: 06/04/2023]
Abstract
Trihalomethanes (THMs) are disinfection byproducts found in chlorinated water, and are associated with several different kinds of cancer in human populations and experimental animal models. Metabolism of THMs proceeds through enzymes such as GSTT1 and CYP2E1 and gives rise to reactive intermediates, which form the basis for their toxic activities. The aim of this study was to assess the mitochondrial dysfunction caused by THMs at low levels, and the resulting hepatic histological and biochemical changes in the mouse. Male ICR mice were administered with two THMs: dibromochloromethane (DBCM) and bromodichloromethane (BDCM); once daily, by gavage, to a total of four administrations. Animals were sacrificed four weeks after DBCM and BDCM administrations. Blood biochemistry was performed for alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), total bilirubin (TB), albumin (Alb), total protein (TP), creatinine, and urea. Animals exposed to DBCM and BDCM showed elevated ALT and TB levels (p < 0.05) as compared with controls. Histological analysis confirmed the presence of vacuolar degenerescence and a multifocal necrotizing hepatitis in 33% of animals (n = 2). Mitochondrial analysis showed that THMs reduced mitochondrial bioenergetic activity (succinate dehydrogenase (SQR), cytochrome c oxidase (COX), and ATP synthase) and increased oxidative stress (glutathione S-transferase (GST)) in hepatic tissues (p < 0.05). These results add detail to the current understanding of the mechanisms underlying THM-induced toxicity, supporting the role of mitochondrial dysfunction and oxidative stress in liver toxicity caused by DBCM and BDCM. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1009-1016, 2016.
Collapse
Affiliation(s)
- Ana I Faustino-Rocha
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - D Rodrigues
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - R Gil da Costa
- Faculty of Engineering, Laboratory for Process, Environment and Energy Engineering (LEPAE), University of Porto, UP, 4099-002, Porto, Portugal
| | - C Diniz
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - S Aragão
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - D Talhada
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior, 6201-001, Covilhã, Portugal
| | - M Botelho
- Department of Health Promotion, National Institute of Health (INSA), 4000-055, Porto, Portugal
| | - A Colaço
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
- Animal and Veterinary Research Center (CECAV), School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - M J Pires
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - F Peixoto
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
- Department of Chemistry, School of Life Sciences and Environment, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| | - P A Oliveira
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, UTAD, 5001-911, Vila Real, Portugal
| |
Collapse
|
39
|
Makris KC, Andrianou XD, Charisiadis P, Burch JB, Seth RK, Ioannou A, Picolos M, Christophi CA, Chatterjee S. Association between exposures to brominated trihalomethanes, hepatic injury and type II diabetes mellitus. ENVIRONMENT INTERNATIONAL 2016; 92-93:486-493. [PMID: 27173514 DOI: 10.1016/j.envint.2016.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 06/05/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder in the Western world, commonly diagnosed in the majority of obese patients with type 2 diabetes mellitus (T2DM). Metabolic disrupting chemicals with short half-lives, such as those of halogenated structure (trihalomethanes, THM) have been linked with hepatic insulin resistance phenomena in animal studies. However, human studies evaluating the role of THM exposure on liver pathogenesis and T2DM disease process are scarce. The objectives of this study were to: i) determine the association of urinary brominated THM (BrTHM) levels and T2DM disease status, and ii) investigate the association between urinary BrTHM levels and serum alanine aminotransferase (ALT) concentrations, often used as surrogate markers of NAFLD. A pilot case-control study was conducted in Nicosia, Cyprus (n=95). Cases were physician-diagnosed T2DM patients and controls were healthy individuals. Liver enzymes, leptin and TNF-α were measured in sera, while urinary THM levels were measured using tandem mass spectrometry. Diabetics had higher levels of serum leptin, body mass index and ALT than the controls. Among all study participants those with serum ALT levels above the median (17IU/L) had higher mean tribromomethane (TBM) concentrations compared to those with serum ALT below 17IU/L. A significant increase in the odds of having above the median serum ALT levels [OR 6.38, 95% CI: 1.11, 42.84 (p=0.044)] was observed for each unit increase in creatinine-unadjusted urinary TBM levels, along with BMI and past smoking, after adjusting for possible confounders, such as urinary creatinine, age, sex, and leptin; no other THM compound showed a significant association with serum ALT. Logistic regression models for T2DM using the urinary BrTHM as exposure variables did not reach the predetermined level of significance. The interplay between exposures to BrTHM and the initiation of key pathophysiological events relating to hepatic injury (ALT) and inflammation (leptin) was recognized via the use of selected biomarkers of effect. Our evidence that THM could act as hepatic toxins with a further initiation of diabetogenic effects call for additional studies to help us better understand the disease process of the two co-morbidities (NAFLD and T2DM).
Collapse
Affiliation(s)
- Konstantinos C Makris
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus.
| | - Xanthi D Andrianou
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - Pantelis Charisiadis
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - James B Burch
- South Carolina Statewide Cancer Prevention & Control Program, University of South Carolina, Columbia, SC, USA; Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA; Dorn Department of Veterans Affairs Medical Center, Columbia, SC, USA
| | - Ratanesh K Seth
- Environmental Health & Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Androniki Ioannou
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | | | - Costas A Christophi
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - Saurabh Chatterjee
- Environmental Health & Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
40
|
Dattaroy D, Seth RK, Das S, Alhasson F, Chandrashekaran V, Michelotti G, Fan D, Nagarkatti M, Nagarkatti P, Diehl AM, Chatterjee S. Sparstolonin B attenuates early liver inflammation in experimental NASH by modulating TLR4 trafficking in lipid rafts via NADPH oxidase activation. Am J Physiol Gastrointest Liver Physiol 2016; 310:G510-25. [PMID: 26718771 PMCID: PMC4824178 DOI: 10.1152/ajpgi.00259.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/28/2015] [Indexed: 01/31/2023]
Abstract
Although significant research data exist on the pathophysiology of nonalcoholic steatohepatitis (NASH), finding an efficient treatment regimen for it remains elusive. The present study used sparstolonin B (SsnB), a novel TLR4 antagonist derived from the Chinese herb Sparganium stoloniferum, as a possible drug to mitigate early inflammation in NASH. This study used an early steatohepatitic injury model in high-fat-fed mice with CYP2E1-mediated oxidative stress as a second hit. SsnB was administered for 1 wk along with bromodichloromethane (BDCM), an inducer of CYP2E1-mediated oxidative stress. Results showed that SsnB administration attenuated inflammatory morphology and decreased elevation of the liver enzyme alanine aminotransferase (ALT). Mice administered SsnB also showed decreased mRNA expression of proinflammatory cytokines TNF-α, IFN-γ, IL-1β, and IL-23, while protein levels of both TNF-α and IL-1β were significantly decreased. SsnB significantly decreased Kupffer cell activation as evidenced by reduction in CD68 and monocyte chemoattractant protein-1 (MCP1) mRNA and protein levels with concomitant inhibition of macrophage infiltration in the injured liver. Mechanistically, SsnB decreased TLR4 trafficking to the lipid rafts, a phenomenon described by the colocalization of TLR4 and lipid raft marker flotillin in tissues and immortalized Kupffer cells. Since we have shown previously that NADPH oxidase drives TLR4 trafficking in NASH, we studied the role of SsnB in modulating this pathway. SsnB prevented NADPH oxidase activation in vivo and in vitro as indicated by decreased peroxynitrite formation. In summary, the present study reports a novel use of the TLR4 antagonist SsnB in mitigating inflammation in NASH and in parallel shows a unique molecular mechanism of decreasing nitrative stress.
Collapse
Affiliation(s)
- Diptadip Dattaroy
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Ratanesh Kumar Seth
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Suvarthi Das
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Firas Alhasson
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Varun Chandrashekaran
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | | | - Daping Fan
- 3Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina; and
| | - Mitzi Nagarkatti
- 4Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Prakash Nagarkatti
- 4Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Anna Mae Diehl
- 2Division of Gastroenterology, Duke University, Durham, North Carolina;
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| |
Collapse
|
41
|
He J, Li G, Chen J, Lin J, Zeng C, Chen J, Deng J, Xie P. Prolonged exposure to low-dose microcystin induces nonalcoholic steatohepatitis in mice: a systems toxicology study. Arch Toxicol 2016; 91:465-480. [PMID: 26984711 DOI: 10.1007/s00204-016-1681-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/09/2016] [Indexed: 12/20/2022]
Abstract
Microcystin-LR (MCLR), a cyanotoxin widely present in freshwater, has been shown to have potent acute hepatotoxicity. However, the chronic toxicity of low-dose MCLR remains confusing by traditional measurements of toxicity. This has impeded understanding of the chronic liver damage of low-dose MCLR and corresponding safety risks of the human exposure guideline value. Here, iTRAQ-based proteomics and NMR-based metabonomics were used to decipher the molecular toxicological signatures of low doses of MCLR in mice exposed to this agent for 90 days. Low levels of MCLR, even under the reported no observed adverse effect level, significantly altered hepatic protein expression, especially of proteins associated with lipid metabolism, transport, immune and proteolysis. Coherently, MCLR induced marked perturbations in lipid metabolites in both liver and serum. Integrated analysis of proteomic, metabolic, histological and cytokine profiles revealed that MCLR significantly inhibited fatty acid β-oxidation and hepatic lipoprotein secretion and promoted hepatic inflammation, resulting in nonalcoholic steatohepatitis disease (NASH). These findings for the first time provide compelling evidence that chronic exposure to low-level MCLR can induce NASH. These results also indicate that current guidelines for MCs in drinking water may be inadequate and associated with risks to human health.
Collapse
Affiliation(s)
- Jun He
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7, Wuhan, 430072, People's Republic of China
| | - Guangyu Li
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jun Chen
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7, Wuhan, 430072, People's Republic of China
| | - Juan Lin
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7, Wuhan, 430072, People's Republic of China
| | - Cheng Zeng
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jing Chen
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7, Wuhan, 430072, People's Republic of China
| | - Junliang Deng
- Shanghai Biotree Biotech Co., Ltd, Shanghai, 200433, People's Republic of China
| | - Ping Xie
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7, Wuhan, 430072, People's Republic of China.
| |
Collapse
|
42
|
Villagarcía HG, Sabugo V, Castro MC, Schinella G, Castrogiovanni D, Spinedi E, Massa ML, Francini F. Chronic Glucocorticoid-Rich Milieu and Liver Dysfunction. Int J Endocrinol 2016; 2016:7838290. [PMID: 27597864 PMCID: PMC4997070 DOI: 10.1155/2016/7838290] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/30/2016] [Accepted: 07/12/2016] [Indexed: 01/08/2023] Open
Abstract
We investigated the impact of chronic hypercorticosteronemia (due to neonatal monosodium L-glutamate, MSG, and treatment) on liver oxidative stress (OS), inflammation, and carbohydrate/lipid metabolism in adult male rats. We evaluated the peripheral concentrations of several metabolic and OS markers and insulin resistance indexes. In liver we assessed (a) OS (GSH and protein carbonyl groups) and inflammatory (IL-1b, TNFa, and PAI-1) biomarkers and (b) carbohydrate and lipid metabolisms. MSG rats displayed degenerated optic nerves, hypophagia, low body and liver weights, and enlarged adipose tissue mass; higher peripheral levels of glucose, triglycerides, insulin, uric acid, leptin, corticosterone, transaminases and TBARS, and peripheral and liver insulin resistance; elevated liver OS, inflammation markers, and glucokinase (mRNA/activity) and fructokinase (mRNA). Additionally, MSG liver phosphofructokinase-2, glucose-6-phosphatase (mRNA and activity) and glucose-6-phosphate dehydrogenase, Chrebp, Srebp1c, fatty acid synthase, and glycerol-3-phosphate (mRNAs) were increased. In conclusion adult MSG rats developed an insulin-resistant state and increased OS and serious hepatic dysfunction characterized by inflammation and metabolic signs suggesting increased lipogenesis. These features, shared by both metabolic and Cushing's syndrome human phenotypes, support that a chronic glucocorticoid-rich endogenous environment mainly impacts on hepatic glucose cycle, displacing local metabolism to lipogenesis. Whether correcting the glucocorticoid-rich environment ameliorates such dysfunctions requires further investigation.
Collapse
Affiliation(s)
| | - Vanesa Sabugo
- Centro de Endocrinología Experimental y Aplicada (CENEXA), UNLP-CONICET-FCM, 1900 La Plata, Argentina
| | - María Cecilia Castro
- Centro de Endocrinología Experimental y Aplicada (CENEXA), UNLP-CONICET-FCM, 1900 La Plata, Argentina
| | - Guillermo Schinella
- Cátedra de Farmacología Básica, Facultad de Ciencias Médicas, UNLP and CICPBA, 1900 La Plata, Argentina
| | - Daniel Castrogiovanni
- Instituto Multidisciplinario de Biología Celular (IMBICE), CONICET-CICPBA-UNLP, 1900 La Plata, Argentina
| | - Eduardo Spinedi
- Centro de Endocrinología Experimental y Aplicada (CENEXA), UNLP-CONICET-FCM, 1900 La Plata, Argentina
| | - María Laura Massa
- Centro de Endocrinología Experimental y Aplicada (CENEXA), UNLP-CONICET-FCM, 1900 La Plata, Argentina
| | - Flavio Francini
- Centro de Endocrinología Experimental y Aplicada (CENEXA), UNLP-CONICET-FCM, 1900 La Plata, Argentina
- *Flavio Francini:
| |
Collapse
|
43
|
Alhasson F, Dattaroy D, Das S, Chandrashekaran V, Seth RK, Schnellmann RG, Chatterjee S. NKT cell modulates NAFLD potentiation of metabolic oxidative stress-induced mesangial cell activation and proximal tubular toxicity. Am J Physiol Renal Physiol 2015; 310:F85-F101. [PMID: 26447219 DOI: 10.1152/ajprenal.00243.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022] Open
Abstract
Obesity and nonalcoholic fatty liver disease (NAFLD) are associated with the development and progression of chronic kidney disease. We recently showed that NAFLD induces liver-specific cytochrome P-450 (CYP)2E1-mediated metabolic oxidative stress after administration of the CYP2E1 substrate bromodichloromethane (BDCM) (Seth RK, Das S, Kumar A, Chanda A, Kadiiska MB, Michelotti G, Manautou J, Diehl AM, Chatterjee S. Toxicol Appl Pharmacol 274: 42-54, 2014; Seth RK, Kumar A, Das S, Kadiiska MB, Michelotti G, Diehl AM, Chatterjee S. Toxicol Sci 134:291-303, 2013). The present study examined the effects of CYP2E1-mediated oxidative stress in NAFLD leading to kidney toxicity. Mice were fed a high-fat diet for 12 wk to induce NAFLD. NAFLD mice were exposed to BDCM, a CYP2E1 substrate, for 4 wk. NAFLD + BDCM increased CYP2E1-mediated lipid peroxidation in proximal tubular cells compared with mice with NAFLD alone or BDCM-treated lean mice, thus ruling out the exclusive role of BDCM. Lipid peroxidation increased IL-1β, TNF-α, and interferon-γ. In parallel, mesangial cell activation was observed by increased α-smooth muscle actin and transforming growth factor-β, which was blocked by the CYP2E1 inhibitor diallyl sulphide both in vivo and in vitro. Mice lacking natural killer T cells (CD1d knockout mice) showed elevated (>4-fold) proinflammatory mediator release, increased Toll-like receptor (TLR)4 and PDGF2 mRNA, and mesangial cell activation in the kidney. Finally, NAFLD CD1D knockout mice treated with BDCM exhibited increased high mobility group box 1 and Fas ligand levels and TUNEL-positive nuclei, indicating that higher cell death was attenuated in TLR4 knockout mice. Tubular cells showed increased cell death and cytokine release when incubated with activated mesangial cells. In summary, an underlying condition of progressive NAFLD causes renal immunotoxicity and aberrant glomerular function possibly through high mobility group box 1-dependent TLR4 signaling and mesangial cell activation, which, in turn, is modulated by intrinsic CD1D-dependent natural killer T cells.
Collapse
Affiliation(s)
- Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; and
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; and
| | - Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; and
| | - Varun Chandrashekaran
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; and
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; and
| | - Rick G Schnellmann
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; and
| |
Collapse
|
44
|
Burch JB, Everson TM, Seth RK, Wirth MD, Chatterjee S. Trihalomethane exposure and biomonitoring for the liver injury indicator, alanine aminotransferase, in the United States population (NHANES 1999-2006). THE SCIENCE OF THE TOTAL ENVIRONMENT 2015; 521-522:226-34. [PMID: 25847167 PMCID: PMC4462191 DOI: 10.1016/j.scitotenv.2015.03.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/12/2015] [Accepted: 03/13/2015] [Indexed: 05/08/2023]
Abstract
Exposure to trihalomethanes (or THMs: chloroform, bromoform, bromodichloromethane, and dibromochloromethane [DBCM]) formed via drinking water disinfection has been associated with adverse reproductive outcomes and cancers of the digestive or genitourinary organs. However, few studies have examined potential associations between THMs and liver injury in humans, even though experimental studies suggest that these agents exert hepatotoxic effects, particularly among obese individuals. This study examined participants in the National Health and Nutrition Examination Survey (1999-2006, N=2781) to test the hypothesis that THMs are associated with liver injury as assessed by alanine aminotransferase (ALT) activity in circulation. Effect modification by body mass index (BMI) or alcohol consumption also was examined. Associations between blood THM concentrations and ALT activity were assessed using unconditional multiple logistic regression to calculate prevalence odds ratios (ORs) with 95% confidence intervals (CIs) for exposure among cases with elevated ALT activity (men: >40IU/L, women: >30IU/L) relative to those with normal ALT, after adjustment for variables that may confound the relationship between ALT and THMs. Compared to controls, cases were 1.35 times more likely (95% CI: 1.02, 1.79) to have circulating DBCM concentrations exceeding median values in the study population. There was little evidence for effect modification by BMI, although the association varied by alcohol consumption. Among non-drinkers, cases were more likely than controls to be exposed to DBCM (OR: 3.30, 95% CI: 1.37, 7.90), bromoform (OR: 2.88, 95% CI: 1.21, 6.81), or brominated THMs (OR: 4.00, 95% CI: 1.31, 12.1), but no association was observed among participants with low, or moderate to heavy alcohol consumption. Total THM levels exceeding benchmark exposure limits continue to be reported both in the United States and globally. Results from this study suggest a need for further characterization of ALT activity and possibly other hepatic or metabolic diseases in populations with elevated drinking water THM concentrations.
Collapse
Affiliation(s)
- James B Burch
- South Carolina Statewide Cancer Prevention & Control Program, University of South Carolina, Columbia, SC, United States; Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States; Dorn Department of Veterans Affairs Medical Center, Columbia, SC, United States.
| | - Todd M Everson
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Ratanesh K Seth
- Environmental Health & Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Michael D Wirth
- South Carolina Statewide Cancer Prevention & Control Program, University of South Carolina, Columbia, SC, United States; Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Saurabh Chatterjee
- Environmental Health & Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
45
|
Song BJ, Akbar M, Jo I, Hardwick JP, Abdelmegeed MA. Translational Implications of the Alcohol-Metabolizing Enzymes, Including Cytochrome P450-2E1, in Alcoholic and Nonalcoholic Liver Disease. ADVANCES IN PHARMACOLOGY 2015; 74:303-72. [PMID: 26233911 DOI: 10.1016/bs.apha.2015.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fat accumulation (hepatic steatosis) in alcoholic and nonalcoholic fatty liver disease is a potentially pathologic condition which can progress to steatohepatitis (inflammation), fibrosis, cirrhosis, and carcinogenesis. Many clinically used drugs or some alternative medicine compounds are also known to cause drug-induced liver injury, which can further lead to fulminant liver failure and acute deaths in extreme cases. During liver disease process, certain cytochromes P450 such as the ethanol-inducible cytochrome P450-2E1 (CYP2E1) and CYP4A isozymes can be induced and/or activated by alcohol and/or high-fat diets and pathophysiological conditions such as fasting, obesity, and diabetes. Activation of these P450 isozymes, involved in the metabolism of ethanol, fatty acids, and various drugs, can produce reactive oxygen/nitrogen species directly and/or indirectly, contributing to oxidative modifications of DNA/RNA, proteins and lipids. In addition, aldehyde dehydrogenases including the mitochondrial low Km aldehyde dehydrogenase-2 (ALDH2), responsible for the metabolism of acetaldehyde and lipid aldehydes, can be inactivated by various hepatotoxic agents. These highly reactive acetaldehyde and lipid peroxides, accumulated due to ALDH2 suppression, can interact with cellular macromolecules DNA/RNA, lipids, and proteins, leading to suppression of their normal function, contributing to DNA mutations, endoplasmic reticulum stress, mitochondrial dysfunction, steatosis, and cell death. In this chapter, we specifically review the roles of the alcohol-metabolizing enzymes including the alcohol dehydrogenase, ALDH2, CYP2E1, and other enzymes in promoting liver disease. We also discuss translational research opportunities with natural and/or synthetic antioxidants, which can prevent or delay the onset of inflammation and liver disease.
Collapse
Affiliation(s)
- Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.
| | - Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Inho Jo
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, South Korea
| | - James P Hardwick
- Biochemistry and Molecular Pathology in Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Joshi-Barve S, Kirpich I, Cave MC, Marsano LS, McClain CJ. Alcoholic, Nonalcoholic, and Toxicant-Associated Steatohepatitis: Mechanistic Similarities and Differences. Cell Mol Gastroenterol Hepatol 2015; 1:356-367. [PMID: 28210688 PMCID: PMC5301292 DOI: 10.1016/j.jcmgh.2015.05.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022]
Abstract
Hepatic steatosis and steatohepatitis are common histologic findings that can be caused by multiple etiologies. The three most frequent causes for steatosis/steatohepatitis are alcohol (alcoholic steatohepatitis, ASH), obesity/metabolic syndrome (nonalcoholic steatohepatitis, NASH), and environmental toxicants (toxicant-associated steatohepatitis, TASH). Hepatic steatosis is an early occurrence in all three forms of liver disease, and they often share common pathways to disease progression/severity. Disease progression is a result of both direct effects on the liver as well as indirect alterations in other organs/tissues such as intestine, adipose tissue, and the immune system. Although the three liver diseases (ASH, NASH, and TASH) share many common pathogenic mechanisms, they also exhibit distinct differences. Both shared and divergent mechanisms can be potential therapeutic targets. This review provides an overview of selected important mechanistic similarities and differences in ASH, NASH, and TASH.
Collapse
Key Words
- ALD, alcoholic liver disease
- ALT, alanine aminotransferase
- ASH, alcoholic steatohepatitis
- AST, aspartate transaminase
- Alcoholic Steatohepatitis
- BMI, body mass index
- CYP2E1, cytochrome P450 isoform 2E1
- ECM, extracellular matrix
- ER, endoplasmic reticulum
- HCC, hepatocellular carcinoma
- HDAC, histone deacetylase
- HSC, hepatic stellate cell
- IL, interleukin
- LA, linoleic acid
- LPS, lipopolysaccharide
- Mechanisms
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NK, natural killer
- NKT, natural killer T
- Nonalcoholic Steatohepatitis
- OXLAM, oxidized linoleic acid metabolite
- PAI-1, plasminogen activator inhibitor-1
- PCB153, 2,2′,4,4′,5,5′-hexachlorobiphenyl
- PPAR, peroxisome proliferator-activated receptor
- RNS, reactive nitrogen species
- SNP, single-nucleotide polymorphism
- TAFLD, toxicant-associated fatty liver disease
- TASH, toxicant-associated steatohepatitis
- TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin
- TH, helper T cell
- TLR, Toll-like receptor
- TNF, tumor necrosis factor
- Toxicant-Associated Steatohepatitis
- VA, U.S. Department of Veterans Affairs/Veterans Administration
- miR, microRNA
Collapse
Affiliation(s)
- Swati Joshi-Barve
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Irina Kirpich
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Matthew C. Cave
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Luis S. Marsano
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky,Correspondence Address correspondence to: Craig J. McClain, MD, University of Louisville, 505 South Hancock Street, Louisville, Kentucky 40292. fax: (502) 852-8927.
| |
Collapse
|
47
|
Das S, Alhasson F, Dattaroy D, Pourhoseini S, Seth RK, Nagarkatti M, Nagarkatti PS, Michelotti GA, Diehl AM, Kalyanaraman B, Chatterjee S. NADPH Oxidase-Derived Peroxynitrite Drives Inflammation in Mice and Human Nonalcoholic Steatohepatitis via TLR4-Lipid Raft Recruitment. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1944-57. [PMID: 25989356 DOI: 10.1016/j.ajpath.2015.03.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/04/2015] [Accepted: 03/26/2015] [Indexed: 12/12/2022]
Abstract
The molecular events that link NADPH oxidase activation and the induction of Toll-like receptor (TLR)-4 recruitment into hepatic lipid rafts in nonalcoholic steatohepatitis (NASH) are unclear. We hypothesized that in liver, NADPH oxidase activation is key in TLR4 recruitment into lipid rafts, which in turn up-regulates NF-κB translocation to the nucleus and subsequent DNA binding, leading to NASH progression. Results from confocal microscopy showed that liver from murine and human NASH had NADPH oxidase activation, which led to the formation of highly reactive peroxynitrite, as shown by 3-nitrotyrosine formation in diseased liver. Expression and recruitment of TLR4 into the lipid rafts were significantly greater in rodent and human NASH. The described phenomenon was NADPH oxidase, p47phox, and peroxynitrite dependent, as liver from p47phox-deficient mice and from mice treated with a peroxynitrite decomposition catalyst [iron(III) tetrakis(p-sulfonatophenyl)porphyrin] or a peroxynitrite scavenger (phenylboronic acid) had markedly less Tlr4 recruitment into lipid rafts. Mechanistically, peroxynitrite-induced TLR4 recruitment was linked to increased IL-1β, sinusoidal injury, and Kupffer cell activation while blocking peroxynitrite-attenuated NASH symptoms. The results strongly suggest that NADPH oxidase-mediated peroxynitrite drove TLR4 recruitment into hepatic lipid rafts and inflammation, whereas the in vivo use of the peroxynitrite scavenger phenylboronic acid, a novel synthetic molecule having high reactivity with peroxynitrite, attenuates inflammatory pathogenesis in NASH.
Collapse
Affiliation(s)
- Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Sahar Pourhoseini
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, South Carolina
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, South Carolina
| | - Gregory A Michelotti
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Balaraman Kalyanaraman
- Department of Biophysics, Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina.
| |
Collapse
|
48
|
Narsale AA, Enos RT, Puppa MJ, Chatterjee S, Murphy EA, Fayad R, Pena MO, Durstine JL, Carson JA. Liver inflammation and metabolic signaling in ApcMin/+ mice: the role of cachexia progression. PLoS One 2015; 10:e0119888. [PMID: 25789991 PMCID: PMC4366213 DOI: 10.1371/journal.pone.0119888] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/21/2015] [Indexed: 02/04/2023] Open
Abstract
The ApcMin/+ mouse exhibits an intestinal tumor associated loss of muscle and fat that is accompanied by chronic inflammation, insulin resistance and hyperlipidemia. Since the liver governs systemic energy demands through regulation of glucose and lipid metabolism, it is likely that the liver is a pathological target of cachexia progression in the ApcMin/+ mouse. The purpose of this study was to determine if cancer and the progression of cachexia affected liver endoplasmic reticulum (ER)-stress, inflammation, metabolism, and protein synthesis signaling. The effect of cancer (without cachexia) was examined in wild-type and weight-stable ApcMin/+ mice. Cachexia progression was examined in weight-stable, pre-cachectic, and severely-cachectic ApcMin/+ mice. Livers were analyzed for morphology, glycogen content, ER-stress, inflammation, and metabolic changes. Cancer induced hepatic expression of ER-stress markers BiP (binding immunoglobulin protein), IRE-1α (endoplasmic reticulum to nucleus signaling 1), and inflammatory intermediate STAT-3 (signal transducer and activator of transcription 3). While gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) mRNA expression was suppressed by cancer, glycogen content or protein synthesis signaling remained unaffected. Cachexia progression depleted liver glycogen content and increased mRNA expression of glycolytic enzyme PFK (phosphofrucktokinase) and gluconeogenic enzyme PEPCK. Cachexia progression further increased pSTAT-3 but suppressed p-65 and JNK (c-Jun NH2-terminal kinase) activation. Interestingly, progression of cachexia suppressed upstream ER-stress markers BiP and IRE-1α, while inducing its downstream target CHOP (DNA-damage inducible transcript 3). Cachectic mice exhibited a dysregulation of protein synthesis signaling, with an induction of p-mTOR (mechanistic target of rapamycin), despite a suppression of Akt (thymoma viral proto-oncogene 1) and S6 (ribosomal protein S6) phosphorylation. Thus, cancer induced ER-stress markers in the liver, however cachexia progression further deteriorated liver ER-stress, disrupted protein synthesis regulation and caused a differential inflammatory response related to STAT-3 and NF-κB (Nuclear factor—κB) signaling.
Collapse
Affiliation(s)
- Aditi A. Narsale
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
| | - Reilly T. Enos
- Department of Pathology, Microbiology & Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States of America
| | - Melissa J. Puppa
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, South Carolina, United States of America
| | - E. Angela Murphy
- Department of Pathology, Microbiology & Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States of America
| | - Raja Fayad
- Center for Colon Cancer Research, Columbia, South Carolina, United States of America
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
| | - Majorette O’ Pena
- Center for Colon Cancer Research, Columbia, South Carolina, United States of America
| | - J. Larry Durstine
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
| | - James A. Carson
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
- Center for Colon Cancer Research, Columbia, South Carolina, United States of America
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
49
|
Dattaroy D, Pourhoseini S, Das S, Alhasson F, Seth RK, Nagarkatti M, Michelotti GA, Diehl AM, Chatterjee S. Micro-RNA 21 inhibition of SMAD7 enhances fibrogenesis via leptin-mediated NADPH oxidase in experimental and human nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2015; 308:G298-312. [PMID: 25501551 PMCID: PMC4329476 DOI: 10.1152/ajpgi.00346.2014] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic fibrosis in nonalcoholic steatohepatitis (NASH) is the common pathophysiological process resulting from chronic liver inflammation and oxidative stress. Although significant research has been carried out on the role of leptin-induced NADPH oxidase in fibrogenesis, the molecular mechanisms that connect the leptin-NADPH oxidase axis in upregulation of transforming growth factor (TGF)-β signaling have been unclear. We aimed to investigate the role of leptin-mediated upregulation of NADPH oxidase and its subsequent induction of micro-RNA 21 (miR21) in fibrogenesis. Human NASH livers and a high-fat (60% kcal) diet-fed chronic mouse model, where hepatotoxin bromodichloromethane was used to induce NASH, were used for this study. To prove the role of the leptin-NADPH oxidase-miR21 axis, mice deficient in genes for leptin, p47phox, and miR21 were used. Results showed that wild-type mice and human livers with NASH had increased oxidative stress, increased p47phox expression, augmented NF-κB activation, and increased miR21 levels. These mice and human livers showed increased TGF-β, SMAD2/3-SMAD4 colocalizations in the nucleus, increased immunoreactivity against Col1α, and α-SMA with a concomitant decrease in protein levels of SMAD7. Mice that were deficient in leptin or p47phox had decreased activated NF-κB and miR21 levels, suggesting the role of leptin and NADPH oxidase in inducing NF-κB-mediated miR21 expression. Further miR21 knockout mice had decreased colocalization events of SMAD2/3-SMAD4 in the nucleus, increased SMAD7 levels, and decreased fibrogenesis. Taken together, the studies show the novel role of leptin-NADPH oxidase induction of miR21 as a key regulator of TGF-β signaling and fibrogenesis in experimental and human NASH.
Collapse
Affiliation(s)
- Diptadip Dattaroy
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina;
| | - Sahar Pourhoseini
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina;
| | - Suvarthi Das
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina;
| | - Firas Alhasson
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina;
| | - Ratanesh Kumar Seth
- 1Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina;
| | - Mitzi Nagarkatti
- 2Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina;
| | | | - Anna Mae Diehl
- 3Division of Gastroenterology, Duke University, Durham, North Carolina
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina;
| |
Collapse
|
50
|
Seth RK, Das S, Pourhoseini S, Dattaroy D, Igwe S, Ray JB, Fan D, Michelotti GA, Diehl AM, Chatterjee S. M1 polarization bias and subsequent nonalcoholic steatohepatitis progression is attenuated by nitric oxide donor DETA NONOate via inhibition of CYP2E1-induced oxidative stress in obese mice. J Pharmacol Exp Ther 2015; 352:77-89. [PMID: 25347994 PMCID: PMC4279102 DOI: 10.1124/jpet.114.218131] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/23/2014] [Indexed: 12/15/2022] Open
Abstract
Activation of M1 macrophages in nonalcoholic steatohepatitis (NASH) is produced by several external or endogenous factors: inflammatory stimuli, oxidative stress, and cytokines are known. However, any direct role of oxidative stress in causing M1 polarization in NASH has been unclear. We hypothesized that CYP2E1-mediated oxidative stress causes M1 polarization in experimental NASH, and that nitric oxide (NO) donor administration inhibits CYP2E1-mediated inflammation with concomitant attenuation of M1 polarization. Because CYP2E1 takes center stage in these studies, we used a toxin model of NASH that uses a ligand and a substrate of CYP2E1 for inducing NASH. Subsequently, we used a methionine and choline-deficient diet-induced rodent NASH model where the role of CYP2E1 in disease progression has been shown. Our results show that CYP2E1 causes M1 polarization bias, which includes a significant increase in interleukin-1β (IL-1β) and IL-12 in both models of NASH, whereas CYP2E1-null mice or diallyl sulfide administration prevented it. Administration of gadolinium chloride (GdCl3), a macrophage toxin, attenuated both the initial M1 response and the subsequent M2 response, showing that the observed increase in cytokine levels is primarily from macrophages. Based on the evidence of an adaptive NO increase, the NO donor administration in vivo that mechanistically inhibited CYP2E1 catalyzed the oxidative stress during the entire study in NASH-abrogated M1 polarization and NASH progression. The results obtained show the association of CYP2E1 in M1 polarization, and that inhibition of CYP2E1 catalyzed oxidative stress by an NO donor (DETA NONOate [(Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate]) can be a promising therapeutic strategy in NASH.
Collapse
Affiliation(s)
- Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Sahar Pourhoseini
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Stephen Igwe
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Julie Basu Ray
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Daping Fan
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Gregory A Michelotti
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Anna Mae Diehl
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina (R.K.S., S.D., S.P., D.D., S.C.); School of Science, Technology, Engineering and Mathematics (STEM), Dillard University, New Orleans, Louisiana (S.I., J.B.R.); Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina (D.F.); and Division of Gastroenterology, Duke University, Durham, North Carolina (G.A.M., A.M.D.)
| |
Collapse
|