1
|
Layman AJ, Alsbrook SM, Koturbash IK, McGill MR. Natural Products That Protect Against Acetaminophen Hepatotoxicity: A Call for Increased Rigor in Preclinical Studies of Dietary Supplements. J Diet Suppl 2024; 22:105-122. [PMID: 38562009 PMCID: PMC11442681 DOI: 10.1080/19390211.2024.2335573] [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: 04/04/2024]
Abstract
Acetaminophen (APAP) overdose is one of the most common causes of acute liver injury. The current standard-of-care treatment for APAP hepatotoxicity, N-acetyl-l-cysteine, is highly effective when administered early after overdose, but loses efficacy in later-presenting patients. As a result, there is interest in the identification of new treatments for APAP overdose patients. Natural products are a promising source of new treatments because many are purported to have hepatoprotective effects. In fact, a great deal of research has been done to identify natural products that can protect against APAP-induced liver injury. However, serious concerns have been raised about the rigor and human relevance of these studies. Here, we systematically reviewed the APAP-natural product literature from 2013 to 2023 to determine the veracity of these concerns and the scope of the potential problem. The results substantiate the concerns that have been previously raised and point to concrete steps that can be taken to improve APAP-natural product research.
Collapse
Affiliation(s)
- Alexander J. Layman
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Scott M. Alsbrook
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Igor K. Koturbash
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Mitchell R. McGill
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Dept. of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Dept. of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR USA
| |
Collapse
|
2
|
Masubuchi Y, Ihara A. Protection of mice against carbon tetrachloride-induced acute liver injury by endogenous and exogenous estrogens. Drug Metab Pharmacokinet 2022; 46:100460. [DOI: 10.1016/j.dmpk.2022.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022]
|
3
|
Atia A, Alrawaiq NS, Abdullah A. Tocotrienols Activate Nrf2 Nuclear Translocation and Increase the Antioxidant- Related Hepatoprotective Mechanism in Mice Liver. Curr Pharm Biotechnol 2021; 22:1085-1098. [PMID: 32988349 DOI: 10.2174/1389201021666200928095950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/26/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The most common preparation of tocotrienols is the Tocotrienol-Rich Fraction (TRF). This study aimed to investigate whether TRF induced liver Nrf2 nuclear translocation and influenced the expression of Nrf2-regulated genes. METHODS In the Nrf2 induction study, mice were divided into control, 2000 mg/kg TRF and diethyl maleate treated groups. After acute treatment, mice were sacrificed at specific time points. Liver nuclear extracts were prepared and Nrf2 nuclear translocation was detected through Western blotting. To determine the effect of increasing doses of TRF on the extent of liver nuclear Nrf2 translocation and its implication on the expression levels of several Nrf2-regulated genes, mice were divided into 5 groups (control, 200, 500 and 1000 mg/kg TRF, and butylated hydroxyanisole-treated groups). After 14 days, mice were sacrificed and liver RNA was extracted for qPCR assay. RESULTS 2000 mg/kg TRF administration initiated Nrf2 nuclear translocation within 30 min, reached a maximum level of around 1 h and dropped to half-maximal levels by 24 h. Incremental doses of TRF resulted in dose-dependent increases in liver Nrf2 nuclear levels, along with concomitant dosedependent increases in the expressions of Nrf2-regulated genes. CONCLUSION TRF activated the liver Nrf2 pathway resulting in increased expression of Nrf2-regulated cytoprotective genes.
Collapse
Affiliation(s)
- Ahmed Atia
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Nadia S Alrawaiq
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Azman Abdullah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Ke Q, Yang J, Liu H, Huang Z, Bu L, Jin D, Liu C. Dose- and time-effects responses of Nonylphenol on oxidative stress in rat through the Keap1-Nrf2 signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 216:112185. [PMID: 33836420 DOI: 10.1016/j.ecoenv.2021.112185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
Nonylphenol (NP) is a representative environmental endocrine-disrupting compound that can induce oxidative stress in organisms. The Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway acts an important role in preventing oxidative stress. The aim of this study was to investigate the influence of oxidative stress caused by NP on Keap1-Nrf2 pathway in rats. Rats were treated with NP (30, 90, 270 mg/kg) for different exposure time (7, 14 and 28 days). The levels of reactive oxygen species (ROS) in serum and glutathione S-transferase (GST), UDP-Glucuronosyl Transferase (UGT) in liver were detected by ELISA kits. Western blot was used to detect Keap1, Nrf2 protein expression in liver and cerebral cortex. The results showed that 28 days of NP exposure significantly increased ROS levels in NPH group. And 14 days exposure to NP significantly enhanced the levels of GST and UGT, while 28 days of exposure showed a suppressive effect. In liver, Keap1 levels was upregulated at 7, 14 and 28 days of NP exposure, while nuclear Nrf2 levels decreased at 7 and 28 days but increased at 14 days. In cerebral cortex, Keap1 and Nrf2 expression increased at 14 days but decreased at 28 days. Besides, with the prolongation of NP exposure time, the GST and UGT levels in NPM and NPH groups were increased firstly and then decreased, while Keap1 and Nrf2 protein levels were constantly decreased in liver and cerebral cortex. In conclusion, the lower dose and shorter exposure time of NP activated the Keap1-Nrf2 pathway that may reduce the damage of oxidative stress, but when further exposed to NP at higher dose and time, the pathway could be inhibited.
Collapse
Affiliation(s)
- Qianhua Ke
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Jiao Yang
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Huan Liu
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Zhuoquan Huang
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Lingling Bu
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Dengpeng Jin
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Chunhong Liu
- The Key Laboratory of Food Quality and Safety of Guangdong Province, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
5
|
Lee SE, Park YS. The Emerging Roles of Antioxidant Enzymes by Dietary Phytochemicals in Vascular Diseases. Life (Basel) 2021; 11:life11030199. [PMID: 33806594 PMCID: PMC8001043 DOI: 10.3390/life11030199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
Vascular diseases are major causes of death worldwide, causing pathologies including diabetes, atherosclerosis, and chronic obstructive pulmonary disease (COPD). Exposure of the vascular system to a variety of stressors and inducers has been implicated in the development of various human diseases, including chronic inflammatory diseases. In the vascular wall, antioxidant enzymes form the first line of defense against oxidative stress. Recently, extensive research into the beneficial effects of phytochemicals has been conducted; phytochemicals are found in commonly used spices, fruits, and herbs, and are used to prevent various pathologic conditions, including vascular diseases. The present review aims to highlight the effects of dietary phytochemicals role on antioxidant enzymes in vascular diseases.
Collapse
|
6
|
Kopacz A, Werner E, Kloska D, Hajduk K, Fichna J, Jozkowicz A, Piechota-Polanczyk A. Nrf2 transcriptional activity in the mouse affects the physiological response to tribromoethanol. Biomed Pharmacother 2020; 128:110317. [PMID: 32485566 DOI: 10.1016/j.biopha.2020.110317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022] Open
Abstract
Up to date, there is no information on the influence of 2,2,2-tribromoethanol (TBE; Avertin), a commonly used anaesthetic, on mice with impaired antioxidant capacity. We aimed to analyse the effect of a single dose of Avertin on anaesthesia duration time, inflammatory response, oxidative stress and collagen deposition in the large intestine of Nrf2 transcriptional knockout mice (tNrf2-/-). The studies were performed on six-month-old female mice Nrf2+/+ and tNrf2-/- randomly assigned to Avertin (250 mg/kg b.w. single i.p. injection) or vehicle group. We observed a 2-fold increase in anaesthesia time and longer recovery time (p = 0.015) in tNrf2-/- in comparison to Nrf2+/+. However, no hepato- or nephrotoxicity was detected. Interestingly, we found severe changes in colon morphology of untreated tNrf2-/- mice associated with colon shortening (p = 0.02) and thickening (p = 0.015). Avertin treatment caused colon damage manifested with epithelial layer damage and goblet depletion in Nrf2+/+ mice but not in tNrf2-/- individuals. Additionally, Avertin did not induce oxidative stress in colon tissue, but it increased leukocyte infiltration in Nrf2+/+ mice (p = 0.02). Immunofluorescent staining also revealed enhanced deposition of collagen I and collagen III in the colon of untreated tNrf2-/- mice. Avertin contributed to increased deposition of collagen I in Nrf2+/+ mice but reduced deposition of collagen I and III in tNrf2-/- individuals. In conclusion, tNrf2-/- respond to Avertin with prolonged anaesthesia that is not associated with acute toxicity, inflammatory reaction or enhanced oxidative stress. Avertin does not impair intestine morphology in tNrf2-/- mice but can normalise the enhanced fibrosis.
Collapse
Affiliation(s)
- A Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - E Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Animal Reproduction and Anatomy, Faculty of Animal Science, University of Agriculture, Krakow, Poland
| | - D Kloska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - K Hajduk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - J Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - A Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - A Piechota-Polanczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
7
|
Taguchi K, Kensler TW. Nrf2 in liver toxicology. Arch Pharm Res 2019; 43:337-349. [PMID: 31782059 DOI: 10.1007/s12272-019-01192-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022]
Abstract
Liver plays essential roles in the metabolism of many endogenous chemicals and exogenous toxicants. Mechanistic studies in liver have been at the forefront of efforts to probe the roles of bioactivation and detoxication of environmental toxins and toxicants in hepatotoxicity. Moreover, idiosyncratic hepatoxicity remains a key barrier in the clinical development of drugs. The now vast Nrf2 field emerged in part from biochemical and molecular studies on chemical inducers of hepatic detoxication enzymes and subsequent characterization of the modulation of drug/toxicant induced hepatotoxicities in mice through disruption of either Nrf2 or Keap1 genes. In general, loss of Nrf2 increases the sensitivity to such toxic chemicals, highlighting a central role of this transcription factor and its downstream target genes as a modifier to chemical stress. In this review, we summarize the impact of Nrf2 on the toxicology of multiple hepatotoxicants, and discuss efforts to utilize the Nrf2 response in predictive toxicology.
Collapse
Affiliation(s)
- Keiko Taguchi
- Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba, Sendai, 980-8575, Japan.
| | - Thomas W Kensler
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, Seattle, WA, 98109, USA
| |
Collapse
|
8
|
Li S, Zhou J, Xu S, Li J, Liu J, Lu Y, Shi J, Zhou S, Wu Q. Induction of Nrf2 pathway by Dendrobium nobile Lindl. alkaloids protects against carbon tetrachloride induced acute liver injury. Biomed Pharmacother 2019; 117:109073. [PMID: 31212129 DOI: 10.1016/j.biopha.2019.109073] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/25/2019] [Accepted: 06/02/2019] [Indexed: 12/18/2022] Open
Abstract
Dendrobium nobile Lindl. alkaloids (DNLA), the active ingredients of Dendrobium, has been shown to possess anti-oxidative effects. The nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant signaling pathway plays a critical role in the cellular response to oxidative stress. Oxidative damage has been implicated in the mechanism of various hepatotoxins induced liver injury. The present study aimed to examine the protective effects of DNLA on CCl4-induced acute liver injury, and to explore the role of the Nrf2 pathway in the protective action of DNLA. Wild-type (WT) and Nrf2-knockout (Nrf2-/-) mice were administrated with DNLA (20 mg/kg/day, ig) for 7 days, and then challenged with CCl4 (20 μL/kg, ip). In WT mice, DNLA reduced CCl4 induced liver injury, as evidenced by the reduction in the serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), attenuation of malondialdehyde (MDA) production, and improved ultrastructural morphology in hepatocytes. However, the protective effect was diminished in Nrf2-/- mice, indicating an essential role of Nrf2 in DNLA-mediated protection over CCl4 liver injury. Furthermore, it was found that DNLA enhanced Nrf2 expression and nuclear accumulation and increased the expression of Nrf2 regulated downstream proteins. These results demonstrate that DNLA protects mice from CCl4 induced liver injury, probably through the activation of the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Shiyue Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Jinxin Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Shangfu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Jin Li
- Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Yuanfu Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China.
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563003, China.
| |
Collapse
|
9
|
Mukhammadiyeva GF, Karimov DO, Kutlina TG, Valova YV, Khusnutdinova NY, Repina EF, Bakirov AB. Expression of Cell Cycle, Oxidative Stress, and Apoptosis Related Genes Chek1, Hmox1, Casp7 in Rat Liver Exposed to Carbon Tetrachloride. Mol Biol 2019. [DOI: 10.1134/s0026893319010102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Yamashita A, Deguchi J, Honda Y, Yamada T, Miyawaki I, Nishimura Y, Tanaka T. Increased susceptibility to oxidative stress-induced toxicological evaluation by genetically modified nrf2a-deficient zebrafish. J Pharmacol Toxicol Methods 2018; 96:34-45. [PMID: 30594530 DOI: 10.1016/j.vascn.2018.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/10/2018] [Accepted: 12/26/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Oxidative stress plays an important role in drug-induced toxicity. Oxidative stress-mediated toxicities can be detected using conventional animal models but their sensitivity is insufficient, and novel models to improve susceptibility to oxidative stress have been researched. In recent years, gene targeting methods in zebrafish have been developed, making it possible to generate homozygous null mutants. In this study, we established zebrafish deficient in the nuclear factor erythroid 2-related factor 2a (nrf2a), a key antioxidant-responsive gene, and its potential to detect oxidative stress-mediated toxicity was examined. METHODS Nrf2a-deficient zebrafish were generated using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 technique. The loss of nrf2a function was confirmed by the tolerability to hydrogen peroxide and hydrogen peroxide-induced gene expression profiles being related to antioxidant response element (ARE)-dependent signaling. Subsequently, vulnerability of nrf2a-deficient zebrafish to acetaminophen (APAP)- or doxorubicin (DOX)-induced toxicity was investigated. RESULTS Nrf2a-deficient zebrafish showed higher mortality than wild type accompanied by less induction of ARE-dependent genes with hydrogen peroxide treatment. Subsequently, this model showed increased severity and incidence of APAP-induced hepatotoxicity or DOX-induced cardiotoxicity than wild type. DISCUSSION Our results demonstrated that anti-oxidative response might not fully function in this model, and resulted in higher sensitivity to drug-induced oxidative stress. Our data support the usefulness of nrf2a-deficient model as a tool for evaluation of oxidative stress-related toxicity in drug discovery research.
Collapse
Affiliation(s)
- Akihito Yamashita
- Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan; Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan.
| | - Jiro Deguchi
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Yayoi Honda
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Toru Yamada
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Izuru Miyawaki
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Mie, Japan
| | - Toshio Tanaka
- Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan; Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan; Mie University Medical Zebrafish Research Center, Mie, Japan; Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan; Department of Omics Medicine, Mie University Industrial Technology Innovation Institute, Mie, Japan
| |
Collapse
|
11
|
McGill MR, Jaeschke H. Animal models of drug-induced liver injury. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1031-1039. [PMID: 31007174 DOI: 10.1016/j.bbadis.2018.08.037] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/18/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023]
Abstract
Drug-induced liver injury (DILI) presents unique challenges for consumers, clinicians, and regulators. It is the most common cause of acute liver failure in the US. It is also one of the most common reasons for termination of new drugs during pre-clinical testing and withdrawal of new drugs post-marketing. DILI is generally divided into two forms: intrinsic and idiosyncratic. Many of the challenges with DILI are due in large part to poor understanding of the mechanisms of toxicity. Although useful models of intrinsic DILI are available, they are frequently misused. Modeling idiosyncratic DILI presents greater challenges, but promising new models have recently been developed. The purpose of this manuscript is to provide a critical review of the most popular animal models of DILI, and to discuss the future of DILI research.
Collapse
Affiliation(s)
- Mitchell R McGill
- Dept. of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Dept. of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Hartmut Jaeschke
- Dept. of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
12
|
Morais SB, Figueiredo BC, Assis NRG, Alvarenga DM, de Magalhães MTQ, Ferreira RS, Vieira AT, Menezes GB, Oliveira SC. Schistosoma mansoni SmKI-1 serine protease inhibitor binds to elastase and impairs neutrophil function and inflammation. PLoS Pathog 2018; 14:e1006870. [PMID: 29425229 PMCID: PMC5823468 DOI: 10.1371/journal.ppat.1006870] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 02/22/2018] [Accepted: 01/09/2018] [Indexed: 12/27/2022] Open
Abstract
Protease inhibitors have important function during homeostasis, inflammation and tissue injury. In this study, we described the role of Schistosoma mansoni SmKI-1 serine protease inhibitor in parasite development and as a molecule capable of regulating different models of inflammatory diseases. First, we determine that recombinant (r) SmKI-1 and its Kunitz domain but not the C-terminal region possess inhibitory activity against trypsin and neutrophil elastase (NE). To better understand the molecular basis of NE inhibition by SmKI-1, molecular docking studies were also conducted. Docking results suggest a complete blockage of NE active site by SmKI-1 Kunitz domain. Additionally, rSmKI-1 markedly inhibited the capacity of NE to kill schistosomes. In order to further investigate the role of SmKI-1 in the parasite, we designed specific siRNA to knockdown SmKI-1 in S. mansoni. SmKI-1 gene suppression in larval stage of S. mansoni robustly impact in parasite development in vitro and in vivo. To determine the ability of SmKI-1 to interfere with neutrophil migration and function, we tested SmKI-1 anti-inflammatory potential in different murine models of inflammatory diseases. Treatment with SmKI-1 rescued acetaminophen (APAP)-mediated liver damage, with a significant reduction in both neutrophil recruitment and elastase activity. In the model of gout arthritis, this protein reduced neutrophil accumulation, IL-1β secretion, hypernociception, and overall pathological score. Finally, we demonstrated the ability of SmKI-1 to inhibit early events that trigger neutrophil recruitment in pleural cavities of mice in response to carrageenan. In conclusion, SmKI-1 is a key protein in S. mansoni survival and it has the ability to inhibit neutrophil function as a promising therapeutic molecule against inflammatory diseases. Schistosoma mansoni is one of the main agents of schistosomiasis, which is the most important human helminthic infection in terms of global morbidity and mortality. Although schistosomiasis represents a major public health problem in endemic countries, evidences show that S. mansoni downregulates inflammatory responses in many diseases. Fortunately, the control of inflammatory responses is extended to pathogen-derived antigens, leading us to study one S. mansoni Kunitz type protease inhibitor (SmKI-1), found in larval and adult phases of the parasite. We demonstrate that SmKI-1 inhibits trypsin and neutrophil elastase (NE). Additionally, live parasites that SmKI-1 gene has been suppressed using siRNA displayed an impaired schistosome development both in vitro and in vivo. Further, we demonstrate that SmKI-1 possesses an anti-inflammatory potential in three different murine models of inflammatory diseases: acetaminophen (APAP)-mediated liver damage, gout arthritis, and pleural inflammation in response to carrageenan. In these inflammatory disease models, we evaluated SmKI-1 effect on neutrophil and our results demonstrate this molecule is able to inhibit neutrophil migration and function, regulating inflammation. Thus, our data suggest that SmKI-1 is a promising therapeutic molecule against inflammatory diseases.
Collapse
Affiliation(s)
- Suellen B. Morais
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Barbara C. Figueiredo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Natan R. G. Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Debora M. Alvarenga
- Centro de Biologia Gastrointestinal, Departamento de Morfologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana T. Q. de Magalhães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rafaela S. Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angélica T. Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo B. Menezes
- Centro de Biologia Gastrointestinal, Departamento de Morfologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C. Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
- * E-mail:
| |
Collapse
|
13
|
Luo L, Chen Y, Wang H, Wang S, Liu K, Li X, Wang XJ, Tang X. Mkp-1 protects mice against toxin-induced liver damage by promoting the Nrf2 cytoprotective response. Free Radic Biol Med 2018; 115:361-370. [PMID: 29241671 DOI: 10.1016/j.freeradbiomed.2017.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 12/30/2022]
Abstract
The present study was undertaken to investigate the possible protective effect of mitogen-activated protein kinase phosphatase 1 (Mkp-1) on toxin-induced hepatic injury. Here, we uncovered a positive feedback loop between Mkp-1, a dual threonine/tyrosine phosphatase, and nuclear factor erythroid 2-related factor 2 (Nrf2), a crucial regulator of the defense system in the liver. Mkp-1-/- mice exhibited decreased protein levels of Nrf2, phase II gene products, and reduced glutathione (GSH) in the liver. Induction of detoxifying enzymes by the Nrf2 activator butylated hydroxyanisole (BHA) or sulforaphane, was attenuated in the liver and small intestines of Mkp-1-/- mice, indicating that the Nrf2 signaling pathway is impaired as a result of Mkp-1 deficiency. Mkp-1-/- mice suffered more severe liver injury after a single exposure to hepatotoxin carbon tetrachloride (CCl4) than their wild-type (WT) counterparts. BHA partially rescued the CCl4-induced liver damage in WT mice, but not in Mkp-1-/- mice, suggesting the requirement of Mkp-1 in the activation of Nrf2 signaling against the liver injury. Mechanistically, Mkp-1 upregulated Nrf2 through a direct interaction with the Neh2 domain in the transcription factor, while Nrf2 enhanced the expression of Mkp-1 mRNA by binding to the ARE site at -1719 to -1710bp in the Mkp-1 promoter. Our results reveal novel role of Mkp-1 in the maintenance of redox homeostasis in the liver. Thus, strategies aimed at augmenting Mkp-1 expression may be beneficial in protecting the liver and may provide novel therapeutic approaches to toxin-induced liver injury.
Collapse
Affiliation(s)
- Lin Luo
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Yeru Chen
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Hongyan Wang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China; Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Shengcun Wang
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Kaihua Liu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xin Li
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xiu Jun Wang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China.
| | - Xiuwen Tang
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China.
| |
Collapse
|
14
|
Peng X, Dai C, Liu Q, Li J, Qiu J. Curcumin Attenuates on Carbon Tetrachloride-Induced Acute Liver Injury in Mice via Modulation of the Nrf2/HO-1 and TGF-β1/Smad3 Pathway. Molecules 2018; 23:E215. [PMID: 29351226 PMCID: PMC6017508 DOI: 10.3390/molecules23010215] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/12/2018] [Accepted: 01/18/2018] [Indexed: 01/14/2023] Open
Abstract
This study aimed to investigate the protective effect of curcumin against carbon tetrachloride (CCl₄)-induced acute liver injury in a mouse model, and to explain the underlying mechanism. Curcumin at doses of 50, 100 and 200 mg/kg/day were administered orally once daily for seven days prior to CCl₄ exposure. At 24 h, curcumin-attenuated CCl₄ induced elevated serum transaminase activities and histopathological damage in the mouse's liver. Curcumin pre-treatment at 50, 100 and 200 mg/kg significantly ameliorated CCl₄-induced oxidative stress, characterized by decreased malondialdehyde (MDA) formations, and increased superoxide dismutase (SOD), catalase (CAT) activities and glutathione (GSH) content, followed by a decrease in caspase-9 and -3 activities. Curcumin pre-treatment significantly decreased CCl₄-induced inflammation. Furthermore, curcumin pre-treatment significantly down-regulated the expression of TGF-β1 and Smad3 mRNAs (both p < 0.01), and up-regulated the expression of nuclear-factor erythroid 2-related factor 2 (Nrf2) and HO-1 mRNA (both p < 0.01) in the liver. Inhibition of HO-1 attenuated the protective effect of curcumin on CCl₄-induced acute liver injury. Given these outcomes, curcumin could protect against CCl₄-induced acute liver injury by inhibiting oxidative stress and inflammation, which may partly involve the activation of Nrf2/HO-1 and inhibition of TGF-β1/Smad3 pathways.
Collapse
Affiliation(s)
- Xinyan Peng
- College of Food Engineering, Ludong University, 186 Middle Hongqi Road, Yantai 264025, China.
| | - Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China.
| | - Quanwen Liu
- College of Food Engineering, Ludong University, 186 Middle Hongqi Road, Yantai 264025, China.
| | - Junke Li
- College of Food Engineering, Ludong University, 186 Middle Hongqi Road, Yantai 264025, China.
| | - Jingru Qiu
- College of Food Engineering, Ludong University, 186 Middle Hongqi Road, Yantai 264025, China.
| |
Collapse
|
15
|
Fukaya S, Yoshioka H, Okano T, Nagatsu A, Miura N, Nonogaki T, Onosaka S. Non-toxic Level of Acetaminophen Potentiates Carbon Tetrachloride-Induced Hepatotoxicity in Mice. Biol Pharm Bull 2017; 40:1590-1594. [PMID: 28867745 DOI: 10.1248/bpb.b17-00344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A wide range of medications are routinely used to maintain and improve human health. Hence, it is essential that we understand and predict adverse effects caused by the combined use of multiple medications. In the present study, we investigated whether the combination of carbon tetrachloride (CCl4) and acetaminophen (APAP) had a detrimental effect on the liver. Mice injected with APAP (100 mg/kg) showed no significant changes in hepatic injury markers (alanine aminotransferase and aspartate aminotransferase), histopathological findings, pro-inflammatory cytokine levels, or hepatic oxidative stress. In contrast, a single injection of CCl4 (15 mg/kg) led to a significant increase in hepatic injury, in addition to an increase in pro-inflammatory cytokine levels and oxidative stress. Co-administration of APAP and CCl4 resulted in exacerbation of these hepatic injuries. Our results suggest that a non-toxic dose of APAP has the potential to increase CCl4-induced liver damage and oxidative stress.
Collapse
Affiliation(s)
- Shiori Fukaya
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University
| | - Hiroki Yoshioka
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University
| | | | - Akito Nagatsu
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University
| | - Nobuhiko Miura
- Division of Health Effects Research, Japan National Institute of Occupational Safety and Health
| | | | | |
Collapse
|
16
|
Naturally Occurring Nrf2 Activators: Potential in Treatment of Liver Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3453926. [PMID: 28101296 PMCID: PMC5215260 DOI: 10.1155/2016/3453926] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/08/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022]
Abstract
Oxidative stress plays a major role in acute and chronic liver injury. In hepatocytes, oxidative stress frequently triggers antioxidant response by activating nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor, which upregulates various cytoprotective genes. Thus, Nrf2 is considered a potential therapeutic target to halt liver injury. Several studies indicate that activation of Nrf2 signaling pathway ameliorates liver injury. The hepatoprotective potential of naturally occurring compounds has been investigated in various models of liver injuries. In this review, we comprehensively appraise various phytochemicals that have been assessed for their potential to halt acute and chronic liver injury by enhancing the activation of Nrf2 and have the potential for use in humans.
Collapse
|
17
|
Effects of Eicosapentaenoic Acid on the Cytoprotection Through Nrf2-Mediated Heme Oxygenase-1 in Human Endothelial Cells. J Cardiovasc Pharmacol 2016; 66:108-17. [PMID: 25815672 DOI: 10.1097/fjc.0000000000000251] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Consumption of omega-3 polyunsaturated fatty acid, particularly eicosapentaenoic acid (EPA), is associated with a significant reduction in the risk of developing cardiovascular disease. The aim of this study was to investigate whether heme oxygenase-1 (HO-1) induction contributes to the cytoprotective effects of EPA in endothelial cells threatened with oxidative damage. In this study, we investigated the effect of EPA on the induction of HO-1 by NF-E2-related factor 2 (Nrf2) in human umbilical vein endothelial cells. In cells treated with low concentrations of EPA (10-25 μM), HO-1 expression increased in a time- and concentration-dependent manner. Additionally, EPA treatment increased Nrf2 nuclear translocation and antioxidant response element activity, leading to the upregulation of HO-1 expression. Furthermore, treatment with EPA reduced hydrogen peroxide (H(2)O(2))-induced cell death. The reduction in cell death was reversed by treatment with zinc protoporphyrin, an inhibitor of HO-1, indicating that HO-1 contributed to the protective effect of EPA. These data suggest that EPA protects against H(2)O(2)-induced oxidative stress in endothelial cells by activating Nrf2 and inducting HO-1 expression.
Collapse
|
18
|
Wojdyla K, Wrzesinski K, Williamson J, Fey SJ, Rogowska-Wrzesinska A. Acetaminophen-induced S-nitrosylation and S-sulfenylation signalling in 3D cultured hepatocarcinoma cell spheroids. Toxicol Res (Camb) 2016; 5:905-920. [PMID: 30090399 PMCID: PMC6072433 DOI: 10.1039/c5tx00469a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/26/2016] [Indexed: 12/12/2022] Open
Abstract
Acetaminophen (APAP) is possibly the most widely used medication globally and yet little is known of its molecular effects at therapeutic doses. Using a novel approach, we have analysed the redox proteome of the hepatocellular cell line HepG2/C3A treated with therapeutic doses of APAP and quantitated both individual protein abundance and their reversible S-nitrosylation (SNO) and S-sulfenylation (SOH) modifications by mass spectrometry. APAP treatment results in a late, transient increase in ATP production and a multiplicity of alterations in protein abundance and modifications. The majority of the differentially SNO or SOH modified proteins are found in the endoplasmic reticulum and cytosol, suggesting that the source of reactive species is there. The cellular response indicates: constraint of fatty acid metabolism; reduction in ribosome construction and protein synthesis (to conserve ATP); maintenance of glutathione levels (by increased synthetic capacity); and an increased NADPH production (via the pentose phosphate pathway). This response appears to be coordinated, directly or indirectly, by the canonical Wnt and Nrf2 signalling pathways. Combined with the known role of NAPQI, these studies suggest that the physiological and toxicological responses form a continuum: therapeutic doses of APAP produce reactive species and NAPQI in the cytoplasm but result in little permanent damage. The cell mounts a multifaceted response which minimises disruption and repairs are effected within a day or two. Higher doses of APAP lead to intensified reactive species production, which increasingly disturbs mitochondrial function and eventually leads to cell death.
Collapse
Affiliation(s)
- Katarzyna Wojdyla
- Protein Research Group , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark .
| | - Krzysztof Wrzesinski
- Tissue Culture Engineering Laboratory , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark
| | - James Williamson
- Protein Research Group , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark .
| | - Stephen J Fey
- Tissue Culture Engineering Laboratory , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark
| | - Adelina Rogowska-Wrzesinska
- Protein Research Group , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark .
| |
Collapse
|
19
|
Value of monitoring Nrf2 activity for the detection of chemical and oxidative stress. Biochem Soc Trans 2015; 43:657-62. [PMID: 26551708 PMCID: PMC4613517 DOI: 10.1042/bst20150044] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Indexed: 02/08/2023]
Abstract
Beyond specific limits of exposure, chemical entities can provoke deleterious effects in mammalian cells via direct interaction with critical macromolecules or by stimulating the accumulation of reactive oxygen species (ROS). In particular, these chemical and oxidative stresses can underpin adverse reactions to therapeutic drugs, which pose an unnecessary burden in the clinic and pharmaceutical industry. Novel pre-clinical testing strategies are required to identify, at an earlier stage in the development pathway, chemicals and drugs that are likely to provoke toxicity in humans. Mammalian cells can adapt to chemical and oxidative stress via the action of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which up-regulates the expression of numerous cell defence genes and has been shown to protect against a variety of chemical toxicities. Here, we provide a brief overview of the Nrf2 pathway and summarize novel experimental models that can be used to monitor changes in Nrf2 pathway activity and thus understand the functional consequences of such perturbations in the context of chemical and drug toxicity. We also provide an outlook on the potential value of monitoring Nrf2 activity for improving the pre-clinical identification of chemicals and drugs with toxic liability in humans.
Collapse
|
20
|
McGill MR, Du K, Xie Y, Bajt ML, Ding WX, Jaeschke H. The role of the c-Jun N-terminal kinases 1/2 and receptor-interacting protein kinase 3 in furosemide-induced liver injury. Xenobiotica 2015; 45:442-9. [PMID: 25423287 PMCID: PMC4442771 DOI: 10.3109/00498254.2014.986250] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
1. The mechanisms of furosemide (FS) hepatotoxicity were explored in mice. Specifically, C57Bl/6 J mice were treated with 500 mg FS/kg bodyweight, and c-Jun N-terminal kinase (JNK) activation and receptor-interacting protein kinase 3 (RIP3) expression were measured by western blotting. Co-treatment with FS and the JNK inhibitor SP600125 was also performed, and FS-induced liver injury was compared in wild-type and RIP3 knockout (KO) mice. 2. JNK phosphorylation and RIP3 expression were increased in livers from the FS-treated mice as early as 6 h after treatment and persisted until at least 24 h. JNK phosphorylation was also observed in primary mouse hepatocytes and human HepaRG cells treated with FS. 3. Phosphorylated JNK translocated into mitochondria in livers, but no evidence of mitochondrial damage was observed. 4. SP600125-treated mice, SP600125 co-treated primary mouse hepatocytes and RIP3 KO mice were not protected against FS hepatotoxicity. These data show that, although JNK activation and RIP3 expression are induced by FS, neither contributes to the liver injury.
Collapse
Affiliation(s)
- Mitchell R McGill
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center , Kansas City, KS , USA
| | | | | | | | | | | |
Collapse
|
21
|
Pérez-Vargas JE, Zarco N, Vergara P, Shibayama M, Segovia J, Tsutsumi V, Muriel P. l-Theanine prevents carbon tetrachloride-induced liver fibrosis via inhibition of nuclear factor κB and down-regulation of transforming growth factor β and connective tissue growth factor. Hum Exp Toxicol 2015; 35:135-46. [PMID: 25852135 DOI: 10.1177/0960327115578864] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Here we evaluated the ability of L-theanine in preventing experimental hepatic cirrhosis and investigated the roles of nuclear factor-κB (NF-κB) activation as well as transforming growth factor β (TGF-β) and connective tissue growth factor (CTGF) regulation. Experimental hepatic cirrhosis was established by the administration of carbon tetrachloride (CCl4) to rats (0.4 g/kg, intraperitoneally, three times per week, for 8 weeks), and at the same time, adding L-theanine (8.0 mg/kg) to the drinking water. Rats had ad libitum access to water and food throughout the treatment period. CCl4 treatment promoted NF-κB activation and increased the expression of both TGF-β and CTGF. CCl4 increased the serum activities of alanine aminotransferase and γ-glutamyl transpeptidase and the degree of lipid peroxidation, and it also induced a decrease in the glutathione and glutathione disulfide ratio. L-Theanine prevented increased expression of NF-κB and down-regulated the pro-inflammatory (interleukin (IL)-1β and IL-6) and profibrotic (TGF-β and CTGF) cytokines. Furthermore, the levels of messenger RNA encoding these proteins decreased in agreement with the expression levels. L-Theanine promoted the expression of the anti-inflammatory cytokine IL-10 and the fibrolytic enzyme metalloproteinase-13. Liver hydroxyproline contents and histopathological analysis demonstrated the anti-fibrotic effect of l-theanine. In conclusion, L-theanine prevents CCl4-induced experimental hepatic cirrhosis in rats by blocking the main pro-inflammatory and pro-fibrogenic signals.
Collapse
Affiliation(s)
| | - N Zarco
- Department of Physiology, Biophysics and Neurosciences, Cinvestav-IPN Apartado, D.F. México
| | - P Vergara
- Department of Physiology, Biophysics and Neurosciences, Cinvestav-IPN Apartado, D.F. México
| | - M Shibayama
- Department of Infectomics and Molecular Pathogenesis, Cinvestav-IPN Apartado, D.F. México
| | - J Segovia
- Department of Physiology, Biophysics and Neurosciences, Cinvestav-IPN Apartado, D.F. México
| | - V Tsutsumi
- Department of Infectomics and Molecular Pathogenesis, Cinvestav-IPN Apartado, D.F. México
| | - P Muriel
- Department of Pharmacology, Cinvestav-IPN Apartado, D.F. México
| |
Collapse
|
22
|
Ma D, Fang Q, Wang P, Gao R, Wu W, Lu T, Cao L, Hu X, Wang J. Induction of heme oxygenase-1 by Na+-H+ exchanger 1 protein plays a crucial role in imatinib-resistant chronic myeloid leukemia cells. J Biol Chem 2015; 290:12558-71. [PMID: 25802333 DOI: 10.1074/jbc.m114.626960] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Indexed: 12/18/2022] Open
Abstract
Resistance toward imatinib (IM) and other BCR/ABL tyrosine kinase inhibitors remains troublesome in the treatment of advanced stage chronic myeloid leukemia (CML). The aim of this study was to estimate the reversal effects of down-regulation of Na(+)/H(+) exchanger 1 (NHE1) on the chemoresistance of BCR-ABL-positive leukemia patients' cells and cell lines. After treatment with the specific NHE1 inhibitor cariporide to decrease intracellular pH (pHi), the heme oxygenase-1 (HO-1) levels of the K562R cell line and cells from IM-insensitive CML patients decreased. HO-1, as a Bcr/Abl-dependent survival molecule in CML cells, is important for the resistance to tyrosine kinase inhibitors in patients with newly diagnosed CML or IM-resistant CML. Silencing PKC-β and Nrf-2 or treatment with inhibitors of p38 pathways obviously blocked NHE1-induced HO-1 expression. Furthermore, treatment with HO-1 or p38 inhibitor plus IM increased the apoptosis of the K562R cell line and IM-insensitive CML patients' cells. Inhibiting HO-1 enhanced the activation of caspase-3 and poly(ADP-ribose) polymerase-1. Hence, the results support the anti-apoptotic role of HO-1 induced by NHE1 in the K562R cell line and IM-insensitive CML patients and provide a mechanism by which inducing HO-1 expression via the PKC-β/p38-MAPK pathway may promote tumor resistance to oxidative stress.
Collapse
Affiliation(s)
- Dan Ma
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Department of Pharmacy, Affiliated BaiYun Hospital of Guiyang Medical University, Guiyang 550014, China
| | - Qin Fang
- Department of Pharmacy, Affiliated BaiYun Hospital of Guiyang Medical University, Guiyang 550014, China, Department of Pharmacy, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, and
| | - Ping Wang
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Rui Gao
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Weibing Wu
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Tangsheng Lu
- School of Pharmacy, Guiyang Medical University, Guiyang 550004, China
| | - Lu Cao
- School of Pharmacy, Guiyang Medical University, Guiyang 550004, China
| | - Xiuying Hu
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Jishi Wang
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China,
| |
Collapse
|
23
|
Rudraiah S, Moscovitz JE, Donepudi AC, Campion SN, Slitt AL, Aleksunes LM, Manautou JE. Differential Fmo3 gene expression in various liver injury models involving hepatic oxidative stress in mice. Toxicology 2014; 325:85-95. [PMID: 25193093 PMCID: PMC4428328 DOI: 10.1016/j.tox.2014.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/29/2014] [Accepted: 08/31/2014] [Indexed: 12/12/2022]
Abstract
Flavin-containing monooxygenase-3 (FMO3) catalyzes metabolic reactions similar to cytochrome P450 monooxygenase, however, most metabolites of FMO3 are considered non-toxic. Recent findings in our laboratory demonstrated Fmo3 gene induction following toxic acetaminophen (APAP) treatment in mice. The goal of this study was to evaluate Fmo3 gene expression in other diverse mouse models of hepatic oxidative stress and injury. Fmo3 gene regulation by Nrf2 was also investigated using Nrf2 knockout (Nrf2 KO) mice. In our studies, male C57BL/6J mice were treated with toxic doses of hepatotoxicants or underwent bile duct ligation (BDL, 10 days). Hepatotoxicants included APAP (400 mg/kg, 24-72 h), alpha-naphthyl isothiocyanate (ANIT; 50 mg/kg, 2-48 h), carbon tetrachloride (CCl4; 10 or 30 μL/kg, 24 and 48 h) and allyl alcohol (AlOH; 30 or 60 mg/kg, 6 and 24 h). Because oxidative stress activates nuclear factor (erythroid-derived 2)-like 2 (Nrf2), additional studies investigated Fmo3 gene regulation by Nrf2 using Nrf2 knockout (Nrf2 KO) mice. At appropriate time-points, blood and liver samples were collected for assessment of plasma alanine aminotransferase (ALT) activity, plasma and hepatic bile acid levels, as well as liver Fmo3 mRNA and protein expression. Fmo3 mRNA expression increased significantly by 43-fold at 12 h after ANIT treatment, and this increase translates to a 4-fold change in protein levels. BDL also increased Fmo3 mRNA expression by 1899-fold, but with no change in protein levels. Treatment of mice with CCl4 decreased liver Fmo3 gene expression, while no change in expression was detected with AlOH treatment. Nrf2 KO mice are more susceptible to APAP (400mg/kg, 72 h) treatment compared to their wild-type (WT) counterparts, which is evidenced by greater plasma ALT activity. The Fmo3 mRNA and protein expression increased in Nrf2 KO mice after APAP treatment. Collectively, not all hepatotoxicants that produce oxidative stress alter Fmo3 gene expression. Along with APAP, toxic ANIT treatment in mice markedly increased Fmo3 gene expression. While BDL increased the Fmo3 mRNA expression, the protein level did not change. The discrepancy with Fmo3 induction in cholestatic models, ANIT and BDL, is not entirely clear. Results from Nrf2 KO mice with APAP suggest that the transcriptional regulation of Fmo3 during liver injury may not involve Nrf2.
Collapse
Affiliation(s)
- Swetha Rudraiah
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA.
| | - Jamie E Moscovitz
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA.
| | - Ajay C Donepudi
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.
| | - Sarah N Campion
- Drug Safety Research and Development, Pfizer Inc., Groton, USA.
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA.
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
24
|
Wu Q, Zhang D, Tao N, Zhu QN, Jin T, Shi JS, Liu J. Induction of Nrf2 and metallothionein as a common mechanism of hepatoprotective medicinal herbs. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:207-21. [PMID: 24467545 DOI: 10.1142/s0192415x14500141] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many Chinese medicines have the potential to be hepatoprotective and therefore can be used to treat acute and chronic liver diseases. The challenge is to identify the molecular target for their protective mechanism. This study investigated the induction of nuclear factor-erythroid 2(NF-E2)-related factor 2 (Nrf2) antioxidant genes and metallothionein as a common mechanism of hepatoprotective effects of Chinese medicines such as Piper puberulum. Mice were pretreated with Piper puberulum extract (PPE, 500 mg/kg, po) or vehicles for seven days, followed by intoxication with CCl 4 (25 μl/kg, ip for 16 h), D-galactosamine (800 mg/kg, ip for 8 h), or acetaminophen (400 mg/kg, ip for 8 h). Hepatotoprotection was evaluated by serum enzyme activities and histopathology. To determine the mechanism of protection, mice were given PPE (250-1000 mg/kg, po for seven days) and livers were collected to quantify the expression of Nrf2-targeted genes and metallothionein. Nrf2-null mice were also used to determine the role of Nrf2 in PPE-mediated hepatoprotection.PPE pretreatment protected against the hepatotoxicity produced by CCl 4, D-galactosamine, and acetaminophen, as evidenced by decreased serum enzyme activities and ameliorated liver lesions. PPE treatment increased the expression of hepatic Nrf2, NAD(P)H:quinone oxidoreductase1 (Nqo1), heme oxygenase-1 (Ho-1), glutamate-cysteine ligases (Gclc), and metallothionein (MT), at both transcripts and protein levels. PPE protected wild-type mice from CCl 4 and acetaminophen hepatotoxicity, but not Nrf2-null mice, fortifying the Nrf2-dependent protection. In conclusion, induction of the Nrf2 antioxidant pathways and metallothionein appears to be a common mechanism for hepatoprotective herbs such as PPE.
Collapse
Affiliation(s)
- Qin Wu
- Department of Pharmacology and Key Lab for Basic Pharmacology of Ministry of Education, Zunyi Medical College, Zunyi 563003, P. R. China
| | | | | | | | | | | | | |
Collapse
|
25
|
Qu Q, Liu J, Zhou HH, Klaassen CD. Nrf2 protects against furosemide-induced hepatotoxicity. Toxicology 2014; 324:35-42. [PMID: 24813929 DOI: 10.1016/j.tox.2014.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/17/2014] [Accepted: 02/17/2014] [Indexed: 10/25/2022]
Abstract
Furosemide is a diuretic drug, but its reactive intermediates lead to acute liver injury in mice. Given the essential role of Nrf2 as a cellular defense regulator, we investigated whether Nrf2 would protect against furosemide-induced liver injury using the Nrf2 "gene-dose response" mouse model (Nrf2-null with Nrf2 knock-out, wild-type with normal expression of Nrf2, Keap1-KD with enhanced Nrf2 activation and Keap1-HKO mice with maximum Nrf2 activation). Twenty-four hours after furosemide administration (250mg/kg, i.p.), serum ALT activities and histopathological analysis indicated severe hepatotoxicity in Nrf2-null and WT mice, but significantly less in the Nrf2-overexpressing Keap1-KD and Keap1-HKO mice. Furosemide increased the mRNA of genes involved in the acute phase response (hemeoxygenase-1 and metallothionein-1), ER stress (C/Ebp-homologous protein and Growth arrest and DNA-damage-inducible protein), inflammatory cytokine (interleukin 1 beta), chemokines (macrophage inflammatory protein 2 and mouse keratinocyte-derived chemokine), as well as apoptosis (early growth response factor and BCL2-associated X protein) in livers of Nrf2-null and wild-type mice, but these genes increased less in mice with more Nrf2. The two genotypes of over-expressed Nrf2 mice had increased expression of the Nrf2 target genes Gclm, Gclc and Nqo1 prior to furosemide administration, and the expressions of these genes were increased further after furosemide administration. Thus, our findings provide strong evidence that over-expression of Nrf2 in Keap1-KD and Keap1-HKO mice and the increases in mRNA of a number of genes involved in anti-oxidative stress, anti-inflammation, anti-ER stress and anti-apoptosis protect against furosemide-induced hepatotoxicity.
Collapse
Affiliation(s)
- Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; University of Kansas Medical Center, Kansas City, KS 66101, USA; Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- University of Kansas Medical Center, Kansas City, KS 66101, USA
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | | |
Collapse
|
26
|
Wei Q, Huang H, Yang L, Yuan J, Yang X, Liu Y, Zhuang Z. Hydrogen peroxide induces adaptive response and differential gene expression in human embryo lung fibroblast cells. ENVIRONMENTAL TOXICOLOGY 2014; 29:478-485. [PMID: 22489041 DOI: 10.1002/tox.21775] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 05/31/2023]
Abstract
Hydrogen peroxide (H2 O2 ), a substance involved in cellular oxidative stress, has been observed to induce an adaptive response, which is characterized by a protection against the toxic effect of H2 O2 at higher concentrations. However, the molecular mechanism for the adaptive response remains unclear. In particular, the existing reports on H2 O2 -induced adaptive response are limited to animal cells and human tumor cells, and relatively normal human cells have never been observed for an adaptive response to H2 O2 . In this study, a human embryo lung fibroblast (MRC-5) cell line was used to model an adaptive response to H2 O2 , and the relevant differential gene expressions by using fluoro mRNA differential display RT-PCR. The results showed significant suppression of cytotoxicity of H2 O2 (1100 μM, 1 h) after pretreatment of the cells with H2 O2 at lower concentrations (0.088-8.8 μM, 24 h), as indicated by cell survival, lactate dehydrogenase release, and the rate of apoptotic cells. Totally 60 mRNA components were differentially expressed compared to untreated cells, and five of them (sizing 400-600 bp) which demonstrated the greatest increase in expression were cloned and sequenced. They showed identity with known genes, such as BCL-2, eIF3S5, NDUFS4, and RPS10. Real time RT-PCR analysis of the five genes displayed a pattern of differential expression consistent with that by the last method. These five genes may be involved in the induction of adaptive response by H2 O2 in human cells, at least in this particular cell type.
Collapse
Affiliation(s)
- Qinzhi Wei
- Department of Toxicology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China; Department of Toxicology, Faculty of Preventive Medicine, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China; Toxicology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, 21 TianBei 1st Road, Shenzhen 518020, Guangdong Province, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
27
|
Kaneko M, Nagamine T, Nakazato K, Mori M. The anti-apoptotic effect of fucoxanthin on carbon tetrachloride-induced hepatotoxicity. J Toxicol Sci 2013; 38:115-26. [PMID: 23358145 DOI: 10.2131/jts.38.115] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
This study evaluated the anti-apoptotic activity of fucoxanthin in carbon tetrachloride (CCl(4))-induced hepatotoxicity. An in vitro study using the 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide (MTT) assay clearly demonstrated an attenuation of CCl(4)-induced hepatotoxicity with fucoxanthin. This effect was dose-dependent; 25 µM was more effective than 10 µM of fucoxanthin for attenuating the hepatotoxicity induced by 5 mM of CCl(4). Acute CCl(4)-hepatotoxicity in rats, with numerous cells positive for the terminal deoxynucleotidyl - transferase (TdT) -mediated deoxyuridine triphosphate-digoxigenin (dUTP) nick-end labeling (TUNEL) stain were seen in the pericentral area of the hepatic lobule. Oral pretreatment of CCl(4)- injected rats with fucoxanthin significantly reduced hepatocyte apoptosis. Fucoxanthin was immunohistochemically shown to increase heme oxygenase-1 expression in the cultured liver cells of Hc cells and TRL1215 cells. By oral pretreatment of CCl(4)-injected rats with fucoxanthin, the hepatic heme oxygenase-1 protein levels were significantly increased compared to those not pretreated with fucoxanthin. Heme oxygenase-1 mRNA expression after CCl(4 )injection was higher in the CCl(4)+fucoxanthin group than in the CCl(4 )group, although the difference was not significant. The findings suggest that fucoxanthin attenuates hepatocyte apoptosis through heme oxygenase-1 induction in CCl(4)-induced acute liver injury.
Collapse
Affiliation(s)
- Mieko Kaneko
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | | | | | | |
Collapse
|
28
|
Lee SE, Park YS. The role of antioxidant enzymes in adaptive responses to environmental toxicants in vascular disease. Mol Cell Toxicol 2013. [DOI: 10.1007/s13273-013-0013-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
29
|
Role of the Nrf2-ARE pathway in liver diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:763257. [PMID: 23766860 PMCID: PMC3665261 DOI: 10.1155/2013/763257] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 04/12/2013] [Indexed: 12/14/2022]
Abstract
The liver is a central organ that performs a wide range of functions such as detoxification and metabolic homeostasis. Since it is a metabolically active organ, liver is particularly susceptible to oxidative stress. It is well documented that liver diseases including hepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma are highly associated with antioxidant capacity. NF-E2-related factor-2 (Nrf2) is an essential transcription factor that regulates an array of detoxifying and antioxidant defense genes expression in the liver. It is activated in response to electrophiles and induces its target genes by binding to the antioxidant response element (ARE). Therefore, the roles of the Nrf2-ARE pathway in liver diseases have been extensively investigated. Studies from several animal models suggest that the Nrf2-ARE pathway collectively exhibits diverse biological functions against viral hepatitis, alcoholic and nonalcoholic liver disease, fibrosis, and cancer via target gene expression. In this review, we will discuss the role of the Nrf2-ARE pathway in liver pathophysiology and the potential application of Nrf2 as a therapeutic target to prevent and treat liver diseases.
Collapse
|
30
|
Stachulski AV, Baillie TA, Kevin Park B, Scott Obach R, Dalvie DK, Williams DP, Srivastava A, Regan SL, Antoine DJ, Goldring CEP, Chia AJL, Kitteringham NR, Randle LE, Callan H, Castrejon JL, Farrell J, Naisbitt DJ, Lennard MS. The Generation, Detection, and Effects of Reactive Drug Metabolites. Med Res Rev 2012; 33:985-1080. [DOI: 10.1002/med.21273] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories; University of Liverpool; Liverpool; L69 7ZD; UK
| | - Thomas A. Baillie
- School of Pharmacy; University of Washington; Box 357631; Seattle; Washington; 98195-7631
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; Groton; Connecticut 06340
| | - Deepak K. Dalvie
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; La Jolla; California 94121
| | - Dominic P. Williams
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Abhishek Srivastava
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Sophie L. Regan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Daniel J. Antoine
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Christopher E. P. Goldring
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Alvin J. L. Chia
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Neil R. Kitteringham
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Laura E. Randle
- School of Pharmacy and Biomolecular Sciences, Faculty of Science; Liverpool John Moores University; James Parsons Building, Byrom Street; Liverpool L3 3AF; UK
| | - Hayley Callan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - J. Luis Castrejon
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - John Farrell
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Martin S. Lennard
- Academic Unit of Medical Education; University of Sheffield; 85 Wilkinson Street; Sheffield S10 2GJ; UK
| |
Collapse
|
31
|
Interactions between drugs and sulforaphane modulate the drug metabolism enzymatic system. Pharmacol Rep 2012; 64:1243-52. [DOI: 10.1016/s1734-1140(12)70920-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 06/08/2012] [Indexed: 11/18/2022]
|
32
|
Lee SE, Yang H, Jeong SI, Jin YH, Park CS, Park YS. Induction of heme oxygenase-1 inhibits cell death in crotonaldehyde-stimulated HepG2 cells via the PKC-δ-p38-Nrf2 pathway. PLoS One 2012; 7:e41676. [PMID: 22848562 PMCID: PMC3405012 DOI: 10.1371/journal.pone.0041676] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 06/24/2012] [Indexed: 12/22/2022] Open
Abstract
Background Crotonaldehyde, an alpha, beta-unsaturated aldehyde present in cigarette smoke, is an environmental pollutant and a product of lipid peroxidation. It also produces adverse effects to humans and is considered as a risk factor for various diseases. Heme oxygenase-1 (HO-1) plays important roles in protecting cells against oxidative stress as a prime cellular defense mechanism. However, HO-1 may be associated with cell proliferation and resistance to apoptosis in cancer cells. The aim of this study was to examine the effects of HO-1 induction on cell survival in crotonaldehyde-stimulated human hepatocellular carcinoma (HepG2) cells. Methods To investigate the signaling pathway involved in crotonaldehyde-induced HO-1 expression, we compared levels of inhibition efficiency of specific inhibitors and specific small interfering RNAs (siRNAs) of several kinases. The cell-cycle and cell death was measured by FACS and terminal dUTP nick-end labeling (TUNEL) staining. Results Treatment with crotonaldehyde caused a significant increase in nuclear translocation of NF-E2 related factor (Nrf2). Treatment with inhibitors of the protein kinase C-δ (PKC-δ) and p38 pathways resulted in obvious blockage of crotonaldehyde-induced HO-1 expression. Furthermore, treatment with HO-1 siRNA and the specific HO-1 inhibitor zinc-protoporphyrin produced an increase in the G0/G1 phase of the cell cycle in crotonaldehyde-stimulated HepG2 cells. Conclusions Taken together, the results support an anti-apoptotic role for HO-1 in crotonaldehyde-stimulated human hepatocellular carcinoma cells and provide a mechanism by which induction of HO-1 expression via PKC-δ–p38 MAPK–Nrf2 pathway may promote tumor resistance to oxidative stress.
Collapse
Affiliation(s)
- Seung Eun Lee
- Department of Microbiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hana Yang
- Department of Microbiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seong Il Jeong
- Department of Microbiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Young-Ho Jin
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Cheung-Seog Park
- Department of Microbiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Yong Seek Park
- Department of Microbiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
33
|
Yoshikawa Y, Miyashita T, Higuchi S, Tsuneyama K, Endo S, Tsukui T, Toyoda Y, Fukami T, Nakajima M, Yokoi T. Mechanisms of the hepatoprotective effects of tamoxifen against drug-induced and chemical-induced acute liver injuries. Toxicol Appl Pharmacol 2012; 264:42-50. [PMID: 22841776 DOI: 10.1016/j.taap.2012.06.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 06/09/2012] [Accepted: 06/29/2012] [Indexed: 02/07/2023]
Abstract
Although estrogen receptor (ER)α agonists, such as estradiol and ethinylestradiol (EE2), cause cholestasis in mice, they also reduce the degree of liver injury caused by hepatotoxicants as well as ischemia-reperfusion. The functional mechanisms of ERα have yet to be elucidated in drug-induced or chemical-induced liver injury. The present study investigated the effects of an ERα agonist, selective ER modulators (SERMs) and an ER antagonist on drug-induced and chemical-induced liver injuries caused by acetaminophen, bromobenzene, diclofenac, and thioacetamide (TA). We observed hepatoprotective effects of EE2, tamoxifen (TAM) and raloxifene pretreatment in female mice that were exposed to a variety of hepatotoxic compounds. In contrast, the ER antagonist did not show any hepatoprotective effects. DNA microarray analyses suggested that monocyte to macrophage differentiation-associated 2 (Mmd2) protein, which has an unknown function, is commonly increased by TAM and RAL pretreatment, but not by pretreatment with the ER antagonist. In ERα-knockout mice, the hepatoprotective effects of TAM and the increased expression of Mmd2 mRNA were not observed in TA-induced liver injury. To investigate the function of Mmd2, the expression level of Mmd2 mRNA was significantly knocked down to approximately 30% in mice by injection of siRNA for Mmd2 (siMmd2). Mmd2 knockdown resulted in a reduction of the protective effects of TAM on TA-induced liver injury in mice. This is the first report of the involvement of ERα in drug-induced or chemical-induced liver injury. Upregulation of Mmd2 protein in the liver was suggested as the mechanism of the hepatoprotective effects of EE2 and SERMs.
Collapse
Affiliation(s)
- Yukitaka Yoshikawa
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Masubuchi Y, Nakayama J, Sadakata Y. Protective effects of exogenous glutathione and related thiol compounds against drug-induced liver injury. Biol Pharm Bull 2011; 34:366-70. [PMID: 21372386 DOI: 10.1248/bpb.34.366] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An overdose of acetaminophen (APAP) causes liver injury both in experimental animals and humans. N-acetylcysteine (NAC) is clinically used as an antidote for APAP intoxication, and it is thought to act by providing cysteine as a precursor of glutathione, which traps a reactive metabolite of APAP. Other hepatoprotective mechanisms of NAC have also been suggested. Here, we examined the effects of thiol compounds with different abilities to restore hepatic glutathione, on hepatotoxicity of APAP and furosemide in mice. Overnight-fasted male CD-1 mice were given APAP or furosemide intraperitoneally. NAC, cysteine, glutathione, or glutathione-monoethyl ester was administered concomitantly with APAP or furosemide. All thiol compounds used in this study effectively protected mice against APAP-induced liver injury. Only glutathione-monoethyl ester completely prevented APAP-induced early hepatic glutathione depletion. Cysteine also significantly restored hepatic glutathione levels. NAC partially restored glutathione levels. Exogenous glutathione had no effect on hepatic glutathione loss. NAC and glutathione highly stimulated the hepatic expression of cytokines, particularly interleukin-6, which might be involved in the alleviation of APAP hepatotoxicity. Furosemide-induced liver injury, which does not accompany hepatic glutathione depletion, was also attenuated by NAC and exogenous glutathione, supporting their protective mechanisms other than replenishment of glutathione. In conclusion, exogenous thiols could alleviate drug-induced liver injury. NAC and glutathione might exert their effects, at least partially, via mechanisms that are independent of increasing hepatic glutathione, but probably act through cytokine-mediated and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Yasuhiro Masubuchi
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Chiba Institute of Science, Choshi, Chiba 288–0025, Japan.
| | | | | |
Collapse
|
35
|
Choi JH, Kim DW, Yun N, Choi JS, Islam MN, Kim YS, Lee SM. Protective effects of hyperoside against carbon tetrachloride-induced liver damage in mice. JOURNAL OF NATURAL PRODUCTS 2011; 74:1055-1060. [PMID: 21428416 DOI: 10.1021/np200001x] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
In this study, the hepatoprotective effects of hyperoside (1), a flavonoid glycoside isolated from Artemisia capillaris, have been examined against carbon tetrachloride (CCl4)-induced liver injury. Mice were treated intraperitoneally with vehicle or 1 (50, 100, and 200 mg·kg(-1)) 30 min before and 2 h after CCl4 (20 μL·kg(-1)) injection. Levels of serum aminotransferases were increased 24 h after CCl4 injection, and these increases were attenuated by 1. Histological analysis showed that 1 prevented portal inflammation, centrizonal necrosis, and Kupffer cell hyperplasia. Lipid peroxidation was increased and hepatic glutathione content was decreased significantly after CCl4 treatment, and these changes were reduced by administration of 1. Protein and mRNA expression of tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and heme oxygenase-1 (HO-1) and nuclear protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2) significantly increased after CCl4 injection. Compound 1 suppressed TNF-α, iNOS, and COX-2 protein and mRNA expression and augmented HO-1 protein and mRNA expression and Nrf2 nuclear protein expression. These results suggest that 1 has protective effects against CCl4-induced acute liver injury, and this protection is likely due to enhancement of the antioxidative defense system and suppression of the inflammatory response.
Collapse
Affiliation(s)
- Jun-Ho Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeongi-do, 440-746, Korea
| | | | | | | | | | | | | |
Collapse
|
36
|
Li HY, Wu SY, Ma Q, Shi N. The pesticide deltamethrin increases free radical production and promotes nuclear translocation of the stress response transcription factor Nrf2 in rat brain. Toxicol Ind Health 2011; 27:579-90. [PMID: 21398409 DOI: 10.1177/0748233710393400] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The transcription factor NF-E2-related factor 2 (Nrf2) plays a critical role in the mammalian response to chemical and oxidative stress through induction of phase II detoxification enzymes and oxidative stress response proteins. We reported that Nrf2 expression was activated by deltamethrin (DM), a prototype of the widely used Parathyroid pesticides, in PC12 cells. However, no study has examined Nrf2 nuclear translocation and free radical production, two hallmarks of oxidative stress, in the mammalian brain in vivo. To this end, we examined translocation of Nrf2 and production of free radicals in rat brain exposed to DM. Indeed, DM initiated nuclear translocation of Nrf2 in a dose-dependent manner. Furthermore, Nrf2 translocation was accompanied by the expression of heme oxygenase-1 gene, an Nrf2-regulated gene linked to free radical production. Deltamethrin exposure promoted free radical formation in rat brain and reactive oxygen species generation in PC12 cells. Translocation of Nrf2 may be a response to DM-dependent induction of free radicals and DM may act as a mammalian neurotoxin by initiating oxidative stress.
Collapse
Affiliation(s)
- H Y Li
- Department of Occupational and Environmental Health, Institution of Environmental and Health, Major Subject of Environment and Health of Fujian Key Universities, School of Public Health, Fujian Medical University, Fuzhou, China
| | | | | | | |
Collapse
|
37
|
Mondal SC, Tripathi DN, Vikram A, Ramarao P, Jena GB. Furosemide-induced genotoxicity and cytotoxicity in the hepatocytes, but weak genotoxicity in the bone marrow cells of mice. Fundam Clin Pharmacol 2011; 26:383-92. [DOI: 10.1111/j.1472-8206.2011.00927.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Gao SS, Choi BM, Chen XY, Zhu RZ, Kim Y, So H, Park R, Sung M, Kim BR. Kaempferol suppresses cisplatin-induced apoptosis via inductions of heme oxygenase-1 and glutamate-cysteine ligase catalytic subunit in HEI-OC1 cell. Pharm Res 2010; 27:235-45. [PMID: 19937094 DOI: 10.1007/s11095-009-0003-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 10/26/2009] [Indexed: 01/22/2023]
Abstract
PURPOSE The present study was undertaken to elucidate the chemoprotective mechanism of kaempferol, which possesses anti-oxidative and anti-apoptotic properties. METHODS House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were treated with kaempferol in the presence or absence of cisplatin. Cisplatin-induced oxidative stress was assessed by analysis of Comet assay, DNA-laddering assay and activation of caspases. Heme oxygenase-1 (HO-1), mitogen-activated protein kinase (MAPK) pathway and nuclear factor-E2-related factor 2 (Nrf2) were measured by Western blot analysis. Transfection of small interfering RNAs (siRNA), glutathione (GSH) assay and RT-PCR were performed in this study. RESULTS Kaempferol protected cells against cisplatin-induced apoptosis in a dose-dependent manner in HEI-OC1 cells. Kaempferol-induced HO-1 expression protected against cell death though the c-Jun N-terminal kinase (JNK) pathway and by the aid of Nrf2 translocation. Kaempferol increased the cellular level of GSH and the expression of GCLC time-dependently. siRNA GCLC blocked the increase of GSH level by kaempferol and the protective effect of kaempferol against cisplatin-induced cell death. CONCLUSION The expression of HO-1 by kaempferol inhibits cisplatin-induced apoptosis in HEI-OC1 cells, and the mechanism of protective effect is also associated with its inductive effect of GCLC expression.
Collapse
Affiliation(s)
- Shang Shang Gao
- Vestibulocochlear Research Center and Department of Biochemistry, School of Medicine, Wonkwang University, Chonbuk, 570-749, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Copple IM, Goldring CE, Kitteringham NR, Park BK. The keap1-nrf2 cellular defense pathway: mechanisms of regulation and role in protection against drug-induced toxicity. Handb Exp Pharmacol 2010:233-66. [PMID: 20020265 DOI: 10.1007/978-3-642-00663-0_9] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adverse drug reactions pose a significant public health problem. In some cases, the process of drug metabolism can contribute to the onset of toxicity through the bioactivation of a parent molecule to a chemically reactive intermediate. In order to maintain a favorable balance between bioactivation and detoxification, mammalian cells have evolved an inducible cell defense system known as the antioxidant response pathway. The activity of this cytoprotective pathway is largely regulated by the transcription factor Nrf2, which governs the expression of many phase II detoxification and antioxidant enzymes. In turn, the activity of Nrf2 is regulated by the cysteine-rich cytosolic inhibitor Keap1, which acts as a "sensor" for chemical/oxidative stress. This article summarizes our current understanding of the molecular mechanisms that regulate the function of the Keap1-Nrf2 pathway and highlights the importance of Nrf2 in the protection against drug-induced toxicity.
Collapse
Affiliation(s)
- Ian M Copple
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, Merseyside L69 3GE, UK
| | | | | | | |
Collapse
|
40
|
Liu J, Zhou ZX, Zhang W, Bell MW, Waalkes MP. Changes in hepatic gene expression in response to hepatoprotective levels of zinc. Liver Int 2009; 29:1222-9. [PMID: 19490425 PMCID: PMC3500762 DOI: 10.1111/j.1478-3231.2009.02007.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Zinc (Zn) administration at non-toxic doses protects against the hepatotoxicity produced by many agents, but the underlying mechanisms remain elusive. AIM To examine the basis of Zn-induced generalised hepatoprotective effects. METHODS Rats and mice were given Zn at known hepatoprotective levels (100 mumol ZnCl2/kg/day, s.c., for 4 days) and molecular responses were assessed. RESULTS Zn treatment produced changes in 5% of the genes on custom-designed mouse liver array and Rat Toxicology-II array. Changes in gene expression were further confirmed and extended by real-time reverse transcriptase-polymerase chain reaction. Zn treatment dramatically increased the expression of the metallothionein (Mt), and modestly increased the expression of acute-phase protein genes (ceruloplasmin, Stat3, egr1, Cxc chemokines and heat-shock proteins). For genes encoding for antioxidant enzymes, some were increased (Nrf2 and Nqo1), while others remained unaltered (Cu, Zn SOD and glutathione S-transferases). Expressions of cytokine and pro-inflammatory genes were not affected, while genes related to cell proliferation (cyclin D1) were modestly upregulated. Some metabolic enzyme genes, including cytochrome P450s and UDP-glucuronosyltransferase, were modestly suppressed, perhaps to switch cellular metabolic energy to acute-phase responses. Liver Zn content was increased between 1.6- and 2.1-fold, while hepatic MT protein was increased between 50 and 200-fold. Mice typically showed greater responses than rats. CONCLUSION Such gene expression changes, particularly the dramatic induction of MT and Nrf2 antioxidant pathway, occur in the absence of overt liver injury, and are probably important in the hepatoprotective effects of Zn against toxic insults.
Collapse
Affiliation(s)
- Jie Liu
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at NIEHS, Research Triangle Park, NC
| | - Zhan-Xiang Zhou
- Department of Medicine, University of Louisville, Louisville, KY
| | - Wei Zhang
- Laboratory of Pharmacology, NIEHS, Research Triangle Park, NC
| | - Matthew W. Bell
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at NIEHS, Research Triangle Park, NC
| | - Michael P. Waalkes
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at NIEHS, Research Triangle Park, NC
| |
Collapse
|
41
|
The Nrf2 transcription factor protects from toxin-induced liver injury and fibrosis. J Transl Med 2008; 88:1068-78. [PMID: 18679376 DOI: 10.1038/labinvest.2008.75] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The liver is frequently exposed to insults, including toxic chemicals and alcohol, viral infection or metabolic overload. Although it can fully regenerate after acute injury, chronic liver damage causes liver fibrosis and cirrhosis, which can result in complete liver failure. In this study, we demonstrate that the NF-E2-related factor 2 (Nrf2) transcription factor protects the liver from acute and chronic toxin-mediated damage. Repair of the liver injury that occurs after a single treatment with the hepatotoxin carbon tetrachloride (CCl(4)) was severely delayed in Nrf2-deficient mice. The defect in repair was accompanied by an enhanced and prolonged inflammatory and profibrotic response. After long-term CCl(4) treatment, liver fibrosis was strongly aggravated in the Nrf2 knockout mice and inflammation was enhanced. We demonstrate that these abnormalities are at least in part due to the reduced expression of known and novel Nrf2 target genes in hepatocytes, which encode enzymes involved in the detoxification of CCl(4) and its metabolites. These results suggest that activation of Nrf2 may be a novel strategy to prevent or ameliorate toxin-induced liver injury and fibrosis.
Collapse
|
42
|
Liu J, Wu Q, Lu YF, Pi J. New insights into generalized hepatoprotective effects of oleanolic acid: key roles of metallothionein and Nrf2 induction. Biochem Pharmacol 2008; 76:922-8. [PMID: 18706400 DOI: 10.1016/j.bcp.2008.07.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 07/14/2008] [Accepted: 07/15/2008] [Indexed: 12/15/2022]
Abstract
Oleanolic acid (OA) is a natural triperpenoid that protects against a variety of hepatotoxicants such as carbon tetrachloride, cadmium, acetaminophen, and bromobenzene. To gain insight into the molecular mechanisms of this generalized hepatoprotection, genomic analysis was performed on mouse and rat livers after OA treatment. Mice and rats were given OA at a hepatoprotective dose (50 micromol/kg, s.c., daily for 4 days) and hepatic RNA was isolated, purified, and subjected to gene expression analysis. OA treatment produced changes in 5% of the genes on custom-designed mouse liver array and rat toxicology-II array. Changes in key gene expressions were further analyzed by real-time RT-PCR. OA treatment dramatically increased expression of hepatic metallothionein (Mt), and increased the expression of the nuclear factor E2-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase 1 (Nqo1), heme oxygenase-1 (Hmox1), and glutamate-cysteine ligases (Gclc and Gclm). OA treatment also increased the expression of genes related to cell proliferation and suppressed the expression of several cytochrome P450 genes possibly to switch cellular metabolic energy to an acute-phase response. Hepatic MT protein was increased 60- and 15-fold in mice and rats, respectively, together with a 30% increase in mouse liver zinc. These gene expression changes, particularly the dramatic induction of MT and the Nrf2 signaling, occur with hepatoprotection doses of OA, and likely are important in the generalized protective effects of OA against hepatotoxicants.
Collapse
Affiliation(s)
- Jie Liu
- Zunyi Medical College, Key Laboratory of Pharmacology, 201 Dalian Road, Zunyi 563000, China.
| | | | | | | |
Collapse
|
43
|
Yamaji K, Ochiai Y, Ohnishi KI, Yawata A, Chikuma T, Hojo H. Up-regulation of hepatic heme oxygenase-1 expression by locally induced interleukin-6 in rats administered carbon tetrachloride intraperitoneally. Toxicol Lett 2008; 179:124-9. [PMID: 18547752 DOI: 10.1016/j.toxlet.2008.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 04/23/2008] [Accepted: 04/23/2008] [Indexed: 12/12/2022]
Abstract
We previously reported that interleukin-6 (IL-6) was locally produced in the early period after intraperitoneal (i.p.) or subcutaneous carbon tetrachloride (CCl4) administration, but not after oral (p.o.) administration. In the present study, we focused on the up-regulation of stress-inducible proteins induced by IL-6 after i.p. CCl4 administration. The expression of heme oxygenase-1 (HO-1) (EC 1.14.99.3) mRNA and protein were induced more in rats administered CCl4 via the i.p. route, compared with the p.o. route; however, expression of heat shock protein (HSP) 72 and HSP90 mRNA were increased to similar extents in both experimental groups. The induction of HO-1 mRNA and protein after i.p. CCl4 administration were significantly reduced after pretreatment with anti-rat IL-6 antibody. Activation of the signal transducer and activator of transcription factor 3 (STAT3), which promotes HO-1 expression, peaked together with plasma levels of IL-6 after i.p. CCl4 administration, suggesting that hepatic HO-1 expression was increased by IL-6 via the Janus kinase/STAT3 pathway. The present data indicate that hepatic HO-1 is up-regulated by endogenously produced IL-6, in addition to its up-regulation by heme derived from cytochrome P450 which has already been reported in rats administered i.p. CCl4. The up-regulation of hepatic HO-1 expression may reduce the tissue injury to livers caused by CCl4.
Collapse
Affiliation(s)
- Kenzaburo Yamaji
- Department of Hygienic Chemistry, Showa Pharmaceutical University, 3-3165 Higashitamagawagakuen, Machida, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|