1
|
Namoju R, Chilaka KN. Protective effect of alpha‑lipoic acid against in utero cytarabine exposure-induced hepatotoxicity in rat female neonates. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6577-6589. [PMID: 38459988 DOI: 10.1007/s00210-024-03036-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
Cytarabine, an anti-metabolite drug, remains the mainstay of treatment for hematological malignancies. It causes various toxic effects including teratogenicity. Alpha lipoic acid (ALA) is a natural antioxidant reported to offer protection against hepatotoxicity induced by various pathological conditions, drugs, or chemicals. We investigated the protective effect of ALA against prenatal cytarabine exposure-induced hepatotoxicity in rat female neonates. A total of 30 dams were randomly assigned to five groups and received normal saline, ALA 200 mg/kg, cytarabine 12.5 mg/kg, cytarabine 25 mg/kg, and cytarabine 25 mg/kg + ALA 200 mg/kg, respectively, from gestational day (GD)8 to GD21. Cytarabine and ALA were administered via intraperitoneal and oral (gavage) routes, respectively. On postnatal day (PND)1, all the live female neonates (pups) were collected and weighed. The blood and liver from pups were carefully collected and used for histopathological, and biochemical evaluations. A significant and dose-dependent decrease in maternal food intake and weight gain was observed in the pregnant rats (dams) of the cytarabine groups as compared to the dams of the control group. The pups exposed to cytarabine showed a significant and dose-dependent (a) decrease in body weight, liver weight, hepatosomatic index, catalase, superoxide dismutase, glutathione, glutathione peroxidase, serum albumin levels and (b) increase in malondialdehyde, alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase, AST/ALT ratio, and histopathological anomalies. Maternal co-administration of ALA ameliorated these biochemical changes and histopathological abnormalities by combating oxidative stress. Future studies are warranted to explore the molecular mechanisms involved in the ALA's protective effects against prenatal cytarabine-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ramanachary Namoju
- Department of Pharmacology, GITAM School of Pharmacy, GITAM Deemed to be University, Visakhapatnam, Andhra Pradesh, 530045, India.
- Department of Pharmacology, Bhaskar Pharmacy College, Jawaharlal Nehru Technical University, Hyderabad, Telangana, 500075, India.
| | - Kavitha N Chilaka
- Department of Pharmacology, GITAM School of Pharmacy, GITAM Deemed to be University, Visakhapatnam, Andhra Pradesh, 530045, India
| |
Collapse
|
2
|
Madiedo-Podvrsan S, Sebillet L, Martinez T, Bacari S, Zhu F, Cattelin M, Leclerc E, Merlier F, Jellali R, Lacroix G, Vayssade M. Development of a lung-liver in vitro coculture model for inhalation-like toxicity assessment. Toxicol In Vitro 2023; 92:105641. [PMID: 37437822 DOI: 10.1016/j.tiv.2023.105641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/09/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Animal models are considered prime study models for inhalation-like toxicity assessment. However, in light of animal experimentation reduction (3Rs), we developed and investigated an alternative in vitro method to study systemic-like responses to inhalation-like exposures. A coculture platform was established to emulate inter-organ crosstalks between a pulmonary barrier, which constitutes the route of entry of inhaled compounds, and the liver, which plays a major role in xenobiotic metabolism. Both compartments (Calu-3 insert and HepG2/C3A biochip) were jointly cultured in a dynamically-stimulated environment for 72 h. The present model was characterized using acetaminophen (APAP), a well-documented hepatotoxicant, to visibly assess the passage and circulation of a xenobiotic through the device. Based on viability and functionality parameters the coculture model showed that the bronchial barrier and the liver biochip can successfully be maintained viable and function in a dynamic coculture setting for 3 days. In a stress-induced environment, present results reported that the coculture model emulated active and functional in vitro crosstalk that seemingly was responsive to xenobiotic exposure doses. The hepatic and bronchial cellular responses to xenobiotic exposure were modified in the coculture setting as they displayed earlier and stronger detoxification processes, highlighting active and functional organ crosstalk between both compartments.
Collapse
Affiliation(s)
- Sabrina Madiedo-Podvrsan
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Louise Sebillet
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Thomas Martinez
- French National Institute for Industrial Environment and Risks, INERIS, Direction milieux et impacts sur le vivant, Verneuil-en-Halatte, France
| | - Salimata Bacari
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Fengping Zhu
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Marie Cattelin
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Eric Leclerc
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, Japan
| | - Franck Merlier
- Université de technologie de Compiègne, UPJV, CNRS Enzyme and Cell Engineering Laboratory, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Rachid Jellali
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France
| | - Ghislaine Lacroix
- French National Institute for Industrial Environment and Risks, INERIS, Direction milieux et impacts sur le vivant, Verneuil-en-Halatte, France
| | - Muriel Vayssade
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu - CS 60319, 60203 Compiègne Cedex, France.
| |
Collapse
|
3
|
Krausová M, Braun D, Buerki-Thurnherr T, Gundacker C, Schernhammer E, Wisgrill L, Warth B. Understanding the Chemical Exposome During Fetal Development and Early Childhood: A Review. Annu Rev Pharmacol Toxicol 2023; 63:517-540. [PMID: 36202091 DOI: 10.1146/annurev-pharmtox-051922-113350] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Early human life is considered a critical window of susceptibility to external exposures. Infants are exposed to a multitude of environmental factors, collectively referred to as the exposome. The chemical exposome can be summarized as the sum of all xenobiotics that humans are exposed to throughout a lifetime. We review different exposure classes and routes that impact fetal and infant metabolism and the potential toxicological role of mixture effects. We also discuss the progress in human biomonitoring and present possiblemodels for studying maternal-fetal transfer. Data gaps on prenatal and infant exposure to xenobiotic mixtures are identified and include natural biotoxins, in addition to commonly reported synthetic toxicants, to obtain a more holistic assessment of the chemical exposome. We highlight the lack of large-scale studies covering a broad range of xenobiotics. Several recommendations to advance our understanding of the early-life chemical exposome and the subsequent impact on health outcomes are proposed.
Collapse
Affiliation(s)
- Magdaléna Krausová
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Dominik Braun
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Tina Buerki-Thurnherr
- Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Particles Biology Interactions, St. Gallen, Switzerland;
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| | - Eva Schernhammer
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Center for Public Health, Department of Epidemiology, Medical University of Vienna, Vienna, Austria; .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Lukas Wisgrill
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria;
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , , .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| |
Collapse
|
4
|
Li J, Zhou M, Lai X, Wang Y, Zou Y, Li K, Li W, Zheng J. Toxicokinetic and bioavailability studies on retrorsine in mice, and ketoconazole-induced alteration in toxicokinetic properties. Biomed Chromatogr 2021; 36:e5270. [PMID: 34727371 DOI: 10.1002/bmc.5270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 11/11/2022]
Abstract
Retrorsine (RTS) is a toxic retronecine-type pyrrolizidine alkaloid, which is widely distributed. The purpose of this study was to develop a high-performance liquid chromatography-tandem mass spectrometric (LC-MS/MS) method for serum RTS determination in mice. Serum samples were deproteinated by acetonitrile, separated on a C18 -PFP column and delivered at 0.8 ml/min with an eluting system composed of water containing 0.1% (v/v) formic acid and acetonitrile containing 0.1% (v/v) formic acid as mobile phases. RTS and the internal standard S-hexylglutathione (H-GSH) were quantitatively monitored with precursor-to-product transitions of m/z 352.1 → 120.1 and m/z 392.2 → 246.3, respectively. The method showed excellent linearity over the concentration range 0.05-50 μg/ml, with correlation coefficient r2 = 0.9992. The extraction recovery was >86.34%, and the matrix effect was not significant. Inter- and intra-day precisions (RSD) were <4.99%. The validated LC-MS/MS method was successfully applied to study the toxicokinetic profiles of serum RTS in mice after intravenous, oral administration and co-treated with ketoconazole, which showed that RTS displayed a long half-life (~11.05 h) and good bioavailability (81.80%). Co-administration of ketoconazole (KTZ) increased the peak serum concentration and area under the concentration-time curve and decreased the clearance and mean residence time. Summing up, a new standardized method was established for quantitative determination of RTS in sera.
Collapse
Affiliation(s)
- Jing Li
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Mengyue Zhou
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Xiaoqiong Lai
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Yang Wang
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Ying Zou
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Kunna Li
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Weiwei Li
- National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China
| | - Jiang Zheng
- School of Basic Medical Sciences, School of Pharmacy and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guizhou, China.,National Engineering Research Center of Miao's Medicines and Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education and Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, Guizhou, China.,Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
5
|
Dai Y, Luo J, Xiang E, Guo Q, He Z, Gong Z, Sun X, Kou H, Xu K, Fan C, Liu J, Qiu S, Wang Y, Wang H, Guo Y. Prenatal Exposure to Retrorsine Induces Developmental Toxicity and Hepatotoxicity of Fetal Rats in a Sex-Dependent Manner: The Role of Pregnane X Receptor Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3219-3231. [PMID: 33685126 DOI: 10.1021/acs.jafc.0c06748] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Pyrrolizidine alkaloids (PAs) are a type of natural phytotoxin that contaminate food and feed and become an environmental health risk to humans and livestock. PAs exert toxicity that requires metabolic activation by cytochrome P450 (CYP) 3A, and case reports showed that fetuses are quite susceptible to PAs toxicity. The aim of this study was to explore the characteristics of developmental toxicity and fetal hepatotoxicity induced by retrorsine (RTS, a typcial toxic PA) and the underlying mechanism. Pregnant Wistar rats were intragastrically administered with 20 mg/(kg·day) RTS from gestation day (GD) 9 to 20. Results showed that prenatal RTS exposure lowered fetal bodyweights, reduced hepatocyte numbers, and potentiated hepatic apoptosis in fetuses, particularly females. Simutaneously, RTS increased CYP3A expression and pregnane X receptor (PXR) activation in female fetal liver. We further confirmed that RTS was a PXR agonist in LO2 and HepG2 cell lines. Furthermore, agonism or antagonism of androgen receptor (AR) either induced or blocked RTS-mediated PXR activation, respectively. As a PXR agonist, RTS toxicity was exacerbated in female fetus due to the increased CYP3A induction and self-metabolism, while the inhibitory effect of AR on PXR activation reduced the susceptibility of male fetus to RTS. Our findings indicated that PXR may be a potential therapeutic target for PA toxicity.
Collapse
Affiliation(s)
- Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Jinyuan Luo
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province, China
| | - E Xiang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Qi Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Zheng He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zheng Gong
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Xiaoxiang Sun
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Hao Kou
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei Province, China
| | - Kequan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Chengpeng Fan
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
| | - Jie Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Shuaikai Qiu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Yanqing Wang
- Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| |
Collapse
|
6
|
Prenatal developmental toxicity study of an alkaloid-free Ageratum conyzoides extract powder in rats by oral administration. Regul Toxicol Pharmacol 2020; 117:104748. [PMID: 32800848 DOI: 10.1016/j.yrtph.2020.104748] [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: 05/05/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 11/22/2022]
Abstract
A prenatal developmental toxicological study was conducted to evaluate the safety of an alkaloid-free Ageratum conyzoides extract powder administration on pregnant female Wistar rats and on the development of the conceptus in accordance with OECD test guideline (no. 414). Pyrrolizidine alkaloids (PAs) naturally present in A. conyzoides have been shown to induce toxicity in past studies, particularly towards hepatic cells. Therefore our test item preparation of A.conyzoides extract (aerial part of the plant) consisted of the removal of PAs. There were no treatment related adverse effects found during maternal examinations (body weights, food consumption, numbers of pregnant and non-pregnant female rats, endocrine evaluation, gravid uterine weights, and number of corpora lutea), maternal/fetal examinations (numbers of implantation sites, pre-and post-implantation loss (%), dead and live fetuses (%), resorption sites), or fetal examinations (litter size and weights, number of fetuses, sex ratio, or external, visceral, and skeletal variations and malformations) in the Ageratum conyzoides extract powder groups at doses of 500, 1000 and 2000 mg/kw bw/day compared to vehicle control group. The no observed adverse effect level (NOAEL) determined for both maternal and developmental toxicity was 2000 mg/kg bw/day, which was the highest dose tested.
Collapse
|
7
|
Female-specific activation of pregnane X receptor mediates sex difference in fetal hepatotoxicity by prenatal monocrotaline exposure. Toxicol Appl Pharmacol 2020; 406:115137. [PMID: 32682830 DOI: 10.1016/j.taap.2020.115137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/21/2020] [Accepted: 07/10/2020] [Indexed: 01/06/2023]
Abstract
Pyrrolizidine alkaloids (PAs) are a group of hepatic toxicant widely present in plants. Cytochrome P450 (CYP) 3A plays a key role in metabolic activation of PAs to generate electrophilic metabolites, which is the main cause of hepatotoxicity. We have previously demonstrated the sex difference in developmental toxicity and hepatotoxicity in fetal rats exposed to monocrotaline (MCT), a representative toxic PA. The aim of this study was to explore the underlying mechanism. 20 mg·kg-1·d-1 MCT was intragastrically given to pregnant Wistar rats from gestation day 9 to 20. CYP3As expression and pregnane X receptor (PXR) activation were specifically enhanced in female fetal liver. After MCT treatment, we also observed a significant increase of CYP3As expression in LO2 cells (high PXR level) or hPXR-transfected HepG2 cells (low PXR level). Employing hPXR and CYP3A4 dual-luciferase reporter gene assay, we confirmed the agonism effect of MCT on PXR-dependent transcriptional activity of CYP3A4. Agonism and antagonism of the androgen receptor (AR) either induced or blocked MCT-induced PXR activation, respectively. This study was the first report identifying that MCT served as PXR agonist to induce CYP3A expression. CYP3A induction may increase self-metabolic activation of MCT and subsequently lead to more severe hepatotoxicity in female fetus. While in male, during the intrauterine period, activated AR by testosterone secretion from developing testes represses MCT-induced PXR activation and CYP3A induction, which may partially protect male fetus from MCT-induced hepatotoxicity.
Collapse
|
8
|
Guo Y, Guo R, Su Y, Fu J, Wang S, Kong Y, Wu C, Wang J, Tan C, Mo C, Zhao B. The PERK/eIF2α/ATF4/CHOP pathway plays a role in regulating monocrotaline-induced endoplasmic reticulum stress in rat liver. Res Vet Sci 2020; 130:237-239. [DOI: 10.1016/j.rvsc.2020.03.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/02/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023]
|