1
|
Yu J, Gao X, Shi H, Zhang L, Nie W, Zhang R, Fang M, Liu Y, Yan Y, Fan B, Wu C, Huang C, Fan S. Activation of Nuclear Receptor CAR: A Pathway to Delay Aging through Enhanced Capacity for Xenobiotic Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416823. [PMID: 39887667 PMCID: PMC11948022 DOI: 10.1002/advs.202416823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/13/2025] [Indexed: 02/01/2025]
Abstract
Environmental factors are linked to aging and age-related diseases. Emerging evidence suggests that enhancing body's resistance to xenobiotics might be an anti-aging strategy. The constitutive androstane receptor (CAR) regulates drug-metabolizing enzymes and transporters, coordinating metabolism and immune responses to adapt to stress triggered by exogenous exposure. However, the impact of activating CAR on aging remains unknown. In this study, Caenorhabditis elegans (C. elegans), drug-induced premature aging mice, and senescence accelerated P8 (SAMP8) mice are used as models to explore the effects of CAR activation on lifespan and healthspan, along with the underlying mechanisms. The results showed that hCAR agonist CITCO and mCAR agonist TCPOBOP prolonged the lifespan and healthspan in model organism. The longevity effects of CITCO and TCPOBOP were attenuated in CAR homozygous nhr-8/daf-12 mutant C. elegans as well as CAR-/- mice. In C. elegans, CITCO activated both anti-stress and detoxification genes, and increased the resistance to environmental adversities. Additionally, the lifespan-extending and xenobiotic resistant effects of CITCO might be related to the regulation of age-related pathways. Furthermore, CITCO improved age-related neurodegeneration in C. elegans models. Taken together, the results suggest that the longevity effects of CAR agonists may be related to the enhancement of xenobiotic resistance of animals.
Collapse
Affiliation(s)
- Jing Yu
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Xiaoyan Gao
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hang Shi
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Lijun Zhang
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Wenlong Nie
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Ruochen Zhang
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Minglv Fang
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Ying Liu
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Yingxuan Yan
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Bingbing Fan
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Chengyuan Wu
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Cheng Huang
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Shengjie Fan
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
2
|
Fashe MM, Miner TA, Collazo VL, Grieco JT, Fallon JK, Jackson KD, Lee CR. Impact of sex and pregnancy on hepatic CYP3A4 expression and activity in a humanized mouse model. Drug Metab Dispos 2025; 53:100025. [PMID: 40023573 DOI: 10.1016/j.dmd.2024.100025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 03/04/2025] Open
Abstract
Cytochrome P450 (CYP) 3A4 is an essential drug-metabolizing enzyme in humans, which shows substantial interindividual variation in response to various intrinsic and extrinsic factors such as sex and pregnancy. In humans, higher CYP3A4 metabolism has been observed in females compared with that in males and in pregnant compared with that in nonpregnant individuals, which has been linked to increased CYP3A4 expression in liver. However, sex differences and pregnancy-mediated changes in hepatic CYP3A4 expression and activity in vivo are not fully understood. In this study, we investigated the utility of a genetically engineered humanized mouse model that carries human CYP3A4/7, pregnane X receptor (PXR) and constitutive androstane receptor (CAR) (huPXR/CAR/CYP3A4/7) to recapitulate sex-associated and pregnancy-associated differences in the hepatic CYP3A4 expression and metabolism observed in humans. We found that female huPXR/CAR/CYP3A4/7 mice exhibited higher basal CYP3A4 mRNA levels and CYP3A4 absolute protein concentrations in liver, and higher 1-hydroxymidazolam formation in liver microsomes, compared with male humanized mice. In contrast, pregnant huPXR/CAR/CYP3A4/7 mice exhibited lower CYP3A4 mRNA levels, CYP3A4 absolute protein concentrations, and 1-hydroxymidazolam formation compared with nonpregnant and postpartum humanized mice. Expression of CAR and Cyp2b10 (a CAR responsive gene) were also higher in females and decreased during pregnancy and were positively correlated with hepatic CYP3A4 mRNA levels. Overall, the huPXR/CAR/CYP3A4/7 mouse model demonstrated utility to study higher basal hepatic CYP3A4 metabolism in females compared with that in males in vivo; however, this humanized mouse model did not demonstrate utility to study pregnancy-mediated increases in CYP3A4 drug substrate metabolism and clearance observed in humans. SIGNIFICANCE STATEMENT: This study assessed the impact of sex and pregnancy on hepatic CYP3A4 protein concentrations and metabolism in humanized PXR/CAR/CYP3A4 mice. Consistent with humans, female mice demonstrated higher hepatic CYP3A4 expression and activity than male mice. In contrast, pregnant mice showed decreased CYP3A4 expression and metabolism compared with nonpregnant mice. The humanized mouse model appeared useful to evaluate sex differences in basal hepatic CYP3A4 metabolism in vivo, but not to study the pregnancy-mediated increase in CYP3A4 metabolism observed during human pregnancy.
Collapse
Affiliation(s)
- Muluneh M Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Taryn A Miner
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Valeria Laboy Collazo
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph T Grieco
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John K Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
3
|
Morris-Schaffer K, Higgins L, Kocabas NA, Faulhammer F, Cordova A, Freeman E, Kamp H, Nahar M, Richmond E, Rooseboom M. A weight of evidence review on the mode of action, adversity, and the human relevance of xylene's observed thyroid effects in rats. Crit Rev Toxicol 2025; 55:1-26. [PMID: 39785829 DOI: 10.1080/10408444.2024.2422890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 01/12/2025]
Abstract
Xylene substances have wide industrial and consumer uses and are currently undergoing dossier and substance evaluation under Registration, Evaluation, Authorization and Restriction of Chemicals (REACH) for further toxicological testing including consideration of an additional neurotoxicological testing cohort to an extended one-generation reproduction toxicity (EOGRT) study. New repeated dose study data on xylenes identify the thyroid as a potential target tissue, and therefore a weight of evidence review is provided to investigate whether or not xylene-mediated changes on the hypothalamus-pituitary-thyroid (HPT) axis are secondary to liver enzymatic induction and are of a magnitude that is relevant for neurological human health concerns. Multiple published studies confirm xylene-mediated increases in liver weight, hepatocellular hypertrophy, and liver enzymatic induction via the oral or inhalation routes, including an increase in uridine 5'-diphospho-glucuronosyltransferase (UDP-GT) activity, the key step in thyroid hormone metabolism in rodents. Only minimal to slight increases in thyroid follicular cell hypertrophy have been observed in some xylene repeated dose studies, with no associated robust or consistent perturbance of thyroid hormone changes across the studies or carried through to offspring indicating adaptive homeostatic maintenance of the HPT axis. Also importantly, in vitro human cell line data from the United States Environmental Protection Agency (US EPA) Toxicity Forecasting (ToxCast) provides supporting evidence of xylene's inability to directly perturb thyroidal functionality. A further supplemental in-depth metabolomics analysis (MetaMap®Tox) of xylene showed a tentative match to compounds that also demonstrate extra-thyroidal effects on the HPT axis as a consequence of liver enzyme induction. Lastly, the slight HPT axis changes mediated by xylene were well-below the published literature thresholds for developmental neurotoxicological outcomes established for thyroidal changes in animals and humans. In summary, the data and various lines of scientific evidence presented herein individually and collectively demonstrate that xylene's mediated changes in the HPT axis, via a secondary extra-thyroidal MOA (i.e. liver enzyme induction), do not raise a human health concern with regards to developmental neurotoxicity. As such, the available toxicological data do not support the classification of xylene as a known or suspected endocrine disruptor, specifically through the thyroid modality, per Regulations Commission Delegated Regulation (EU) 2023/707 of 19 December 2022 amending Regulation (EC) No 1272/2008 and do not support the need for a neurotoxicological cohort evaluation in any subsequent EOGRTS.
Collapse
Affiliation(s)
| | - Larry Higgins
- Scientific Services, Penman Consulting bvba, Brussels, Belgium
| | | | - Frank Faulhammer
- Global Toxicology & Ecotoxicology, BASF SE, Ludwigshafen, Germany
| | - Alexandra Cordova
- Environmental & Earth Sciences, Exponent Incorporated, Austin, TX, USA
| | - Elaine Freeman
- Exponent Incorporated, Chemical Regulation and Food Safety, Washington, D.C., USA
| | | | - Muna Nahar
- Exponent Incorporated, Chemical Regulation and Food Safety, Washington, D.C., USA
| | - Emily Richmond
- Chemical Regulation and Food Safety, Exponent International, UK
| | - Martijn Rooseboom
- Product Stewardship, Science & Regulatory, Shell Global Solutions International B.V. The Hague, the Netherlands
| |
Collapse
|
4
|
Monrose M, Holota H, Martinez G, Damon-Soubeyrand C, Thirouard L, Martinot E, Battistelli E, de Haze A, Bravard S, Tamisier C, Caira F, Coutton C, Barbotin AL, Boursier A, Lakhal L, Beaudoin C, Volle DH. Constitutive Androstane Receptor Regulates Germ Cell Homeostasis, Sperm Quality, and Male Fertility via Akt-Foxo1 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402082. [PMID: 39318179 DOI: 10.1002/advs.202402082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/02/2024] [Indexed: 09/26/2024]
Abstract
Male sexual function can be disrupted by exposure to exogenous compounds that cause testicular physiological alterations. The constitutive androstane receptor (Car) is a receptor for both endobiotics and xenobiotics involved in detoxification. However, its role in male fertility, particularly in regard to the reprotoxic effects of environmental pollutants, remains unclear. This study aims to investigate the role of the Car signaling pathway in male fertility. In vivo, in vitro, and pharmacological approaches are utilized in wild-type and Car-deficient mouse models. The results indicate that Car inhibition impaired male fertility due to altered sperm quality, specifically histone retention, which is correlated with an increased percentage of dying offspring in utero. The data highlighted interactions among Car, Akt, Foxo1, and histone acetylation. This study demonstrates that Car is crucial in germ cell homeostasis and male fertility. Further research on the Car signaling pathway is necessary to reveal unidentified causes of altered fertility and understand the harmful impact of environmental molecules on male fertility and offspring health.
Collapse
Affiliation(s)
- Mélusine Monrose
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Hélène Holota
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Guillaume Martinez
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, F-38000, France
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Grenoble, F-38000, France
| | - Christelle Damon-Soubeyrand
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Plateform Anipath, Clermont-Ferrand, F-63001, France
| | - Laura Thirouard
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Emmanuelle Martinot
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Edwige Battistelli
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Angélique de Haze
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Stéphanie Bravard
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Plateform Anipath, Clermont-Ferrand, F-63001, France
| | - Christelle Tamisier
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Françoise Caira
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - Charles Coutton
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, F-38000, France
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Grenoble, F-38000, France
| | - Anne-Laure Barbotin
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, F-59000, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, F-59000, France
| | - Angèle Boursier
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, F-59000, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, F-59000, France
| | - Laila Lakhal
- INRAe UMR1331, ToxAlim, University of Toulouse, Toulouse, F-31027, France
| | - Claude Beaudoin
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| | - David H Volle
- INSERM U1103, Université Clermont Auvergne, CNRS UMR-6293, GReD Institute, Team-Volle, Clermont-Ferrand, F-63001, France
| |
Collapse
|
5
|
Zheng M, Zhai Y, Yu Y, Shen J, Chu S, Focaccia E, Tian W, Wang S, Liu X, Yuan X, Wang Y, Li L, Feng B, Li Z, Guo X, Qiu J, Zhang C, Hou J, Sun Y, Yang X, Zuo X, Heikenwalder M, Li Y, Yuan D, Li S. TNF compromises intestinal bile-acid tolerance dictating colitis progression and limited infliximab response. Cell Metab 2024; 36:2086-2103.e9. [PMID: 38971153 DOI: 10.1016/j.cmet.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Accepted: 06/07/2024] [Indexed: 07/08/2024]
Abstract
The intestine constantly encounters and adapts to the external environment shaped by diverse dietary nutrients. However, whether and how gut adaptability to dietary challenges is compromised in ulcerative colitis is incompletely understood. Here, we show that a transient high-fat diet exacerbates colitis owing to inflammation-compromised bile acid tolerance. Mechanistically, excessive tumor necrosis factor (TNF) produced at the onset of colitis interferes with bile-acid detoxification through the receptor-interacting serine/threonine-protein kinase 1/extracellular signal-regulated kinase pathway in intestinal epithelial cells, leading to bile acid overload in the endoplasmic reticulum and consequent apoptosis. In line with the synergy of bile acids and TNF in promoting gut epithelial damage, high intestinal bile acids correlate with poor infliximab response, and bile acid clearance improves infliximab efficacy in experimental colitis. This study identifies bile acids as an "opportunistic pathogenic factor" in the gut that would represent a promising target and stratification criterion for ulcerative colitis prevention/therapy.
Collapse
Affiliation(s)
- Mengqi Zheng
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China
| | - Yunjiao Zhai
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Yanbo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jing Shen
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Shuzheng Chu
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Enrico Focaccia
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wenyu Tian
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Sui Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xuesong Liu
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Xi Yuan
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Yue Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Bingcheng Feng
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiaohuan Guo
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing 100084, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cuijuan Zhang
- Institute of Pathology and Pathophysiology, Shandong University School of Medicine, Jinan 250012, China; Department of Pathology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jiajie Hou
- Cancer Centre, Faculty of Health Sciences University of Macau, Macau SAR, China; MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Yiyuan Sun
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiaoyun Yang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiuli Zuo
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; The M3 Research Center, Medical faculty, University Tübingen, Ottfried-Müller Strasse 37, Tübingen, Germany.
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan 250012, China.
| | - Detian Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China.
| | - Shiyang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Provincial Clinical Research Center for Digestive Diseases, Jinan, China; Advanced Medical Research Institute, Shandong University, Jinan 250012, China; Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, China.
| |
Collapse
|
6
|
Heintz MM, Klaren WD, East AW, Haws LC, McGreal SR, Campbell RR, Thompson CM. Comparison of transcriptomic profiles between HFPO-DA and prototypical PPARα, PPARγ, and cytotoxic agents in wild-type and PPARα knockout mouse hepatocytes. Toxicol Sci 2024; 200:183-198. [PMID: 38574385 PMCID: PMC11199908 DOI: 10.1093/toxsci/kfae045] [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: 04/06/2024] Open
Abstract
Recent in vitro transcriptomic analyses for the short-chain polyfluoroalkyl substance, HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate), support conclusions from in vivo data that HFPO-DA-mediated liver effects in mice are part of the early key events of the peroxisome proliferator-activated receptor alpha (PPARα) activator-induced rodent hepatocarcinogenesis mode of action (MOA). Transcriptomic responses in HFPO-DA-treated rodent hepatocytes have high concordance with those treated with a PPARα agonist and lack concordance with those treated with PPARγ agonists or cytotoxic agents. To elucidate whether HFPO-DA-mediated transcriptomic responses in mouse liver are PPARα-dependent, additional transcriptomic analyses were conducted on samples from primary PPARα knockout (KO) and wild-type (WT) mouse hepatocytes exposed for 12, 24, or 72 h with various concentrations of HFPO-DA, or well-established agonists of PPARα (GW7647) and PPARγ (rosiglitazone), or cytotoxic agents (acetaminophen or d-galactosamine). Pathway and predicted upstream regulator-level responses were highly concordant between HFPO-DA and GW7647 in WT hepatocytes. A similar pattern was observed in PPARα KO hepatocytes, albeit with a distinct temporal and concentration-dependent delay potentially mediated by compensatory responses. This delay was not observed in PPARα KO hepatocytes exposed to rosiglitazone, acetaminophen, d-galactosamine. The similarity in transcriptomic signaling between HFPO-DA and GW7647 in both the presence and absence of PPARα in vitro indicates these compounds share a common MOA.
Collapse
|
7
|
Jayanti S, Vitek L, Verde CD, Llido JP, Sukowati C, Tiribelli C, Gazzin S. Role of Natural Compounds Modulating Heme Catabolic Pathway in Gut, Liver, Cardiovascular, and Brain Diseases. Biomolecules 2024; 14:63. [PMID: 38254662 PMCID: PMC10813662 DOI: 10.3390/biom14010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
The crucial physiological process of heme breakdown yields biliverdin (BV) and bilirubin (BR) as byproducts. BV, BR, and the enzymes involved in their production (the "yellow players-YP") are increasingly documented as endogenous modulators of human health. Mildly elevated serum bilirubin concentration has been correlated with a reduced risk of multiple chronic pro-oxidant and pro-inflammatory diseases, especially in the elderly. BR and BV per se have been demonstrated to protect against neurodegenerative diseases, in which heme oxygenase (HMOX), the main enzyme in the production of pigments, is almost always altered. HMOX upregulation has been interpreted as a tentative defense against the ongoing pathologic mechanisms. With the demonstration that multiple cells possess YP, their propensity to be modulated, and their broad spectrum of activity on multiple signaling pathways, the YP have assumed the role of an adjustable system that can promote health in adults. Based on that, there is an ongoing effort to induce their activity as a therapeutic option, and natural compounds are an attractive alternative to the goal, possibly requiring only minimal changes in the life style. We review the most recent evidence of the potential of natural compounds in targeting the YP in the context of the most common pathologic condition of adult and elderly life.
Collapse
Affiliation(s)
- Sri Jayanti
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, General University Hospital and 1st Faculty of Medicine, Charles University, 12000 Prague, Czech Republic;
| | - Camilla Dalla Verde
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Department of Life Sciences, University of Trieste, 34139 Trieste, Italy
| | - John Paul Llido
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Department of Life Sciences, University of Trieste, 34139 Trieste, Italy
- Department of Science and Technology, Philippine Council for Health Research and Development, Bicutan, Taguig City 1631, Philippines
| | - Caecilia Sukowati
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Claudio Tiribelli
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
| | - Silvia Gazzin
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
| |
Collapse
|
8
|
Huillet M, Lasserre F, Gratacap MP, Engelmann B, Bruse J, Polizzi A, Fougeray T, Martin CMP, Rives C, Fougerat A, Naylies C, Lippi Y, Garcia G, Rousseau-Bacquie E, Canlet C, Debrauwer L, Rolle-Kampczyk U, von Bergen M, Payrastre B, Boutet-Robinet E, Gamet-Payrastre L, Guillou H, Loiseau N, Ellero-Simatos S. Pharmacological activation of constitutive androstane receptor induces female-specific modulation of hepatic metabolism. JHEP Rep 2024; 6:100930. [PMID: 38149074 PMCID: PMC10749885 DOI: 10.1016/j.jhepr.2023.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 12/28/2023] Open
Abstract
Background & Aims The constitutive androstane receptor (CAR) is a nuclear receptor that binds diverse xenobiotics and whose activation leads to the modulation of the expression of target genes involved in xenobiotic detoxification and energy metabolism. Although CAR hepatic activity is considered to be higher in women than in men, its sex-dependent response to an acute pharmacological activation has seldom been investigated. Methods The hepatic transcriptome, plasma markers, and hepatic metabolome, were analysed in Car+/+ and Car-/- male and female mice treated either with the CAR-specific agonist 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) or with vehicle. Results Although 90% of TCPOBOP-sensitive genes were modulated in a sex-independent manner, the remaining 10% showed almost exclusive female liver specificity. These female-specific CAR-sensitive genes were mainly involved in xenobiotic metabolism, inflammation, and extracellular matrix organisation. CAR activation also induced higher hepatic oxidative stress and hepatocyte cytolysis in females than in males. Hepatic expression of flavin monooxygenase 3 (Fmo3) was almost abolished and was associated with a decrease in hepatic trimethylamine-N-oxide (TMAO) concentration in TCPOBOP-treated females. In line with a potential role in the control of TMAO homeostasis, CAR activation decreased platelet hyper-responsiveness in female mice supplemented with dietary choline. Conclusions More than 10% of CAR-sensitive genes are sex-specific and influence hepatic and systemic responses such as platelet aggregation. CAR activation may be an important mechanism of sexually-dimorphic drug-induced liver injury. Impact and implications CAR is activated by many drugs and pollutants. Its pharmacological activation had a stronger impact on hepatic gene expression and metabolism in females than in males, and had a specific impact on liver toxicity and trimethylamine metabolism. Sexual dimorphism should be considered when testing and/or prescribing xenobiotics known to activate CAR.
Collapse
Affiliation(s)
- Marine Huillet
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Frédéric Lasserre
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Marie-Pierre Gratacap
- INSERM, UMR-1297 and Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires (I2MC), CHU-Rangueil, Toulouse, France
| | - Beatrice Engelmann
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Justine Bruse
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Arnaud Polizzi
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Tiffany Fougeray
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Céline Marie Pauline Martin
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Clémence Rives
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Anne Fougerat
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Claire Naylies
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Yannick Lippi
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Géraldine Garcia
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Elodie Rousseau-Bacquie
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Cécile Canlet
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Laurent Debrauwer
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Bernard Payrastre
- INSERM, UMR-1297 and Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires (I2MC), CHU-Rangueil, Toulouse, France
- Laboratoire d’Hématologie, CHU de Toulouse, Toulouse, France
| | - Elisa Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Nicolas Loiseau
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| |
Collapse
|
9
|
Liu S, Zhang J, Li R, Zhang C, Wang L, Liang H, Feng G, Xiong D. Triazophos exposure on maternal Daphnia magna at environmental-related concentrations revealed toxic effects to its offspring. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 196:105607. [PMID: 37945248 DOI: 10.1016/j.pestbp.2023.105607] [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: 04/23/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 11/12/2023]
Abstract
Due to chemical and photochemical stability, triazophos has been frequently detected in rivers and oceans over the years with extensive use for pest control in agriculture, and it has become a worldwide ecological concern to the aquatic environment. Until now, fewer data are available regarding the potential long-term adverse effects of triazophos on aquatic invertebrates, which plays an essential role in aquatic food webs, as a key group for water ecosystems. In this experiment, the F1- and F2 progenies of Daphnia magna were recovered when daphnias (F0) exposure to triazophos at environmental-related concentrations (0.1 and 1.0 μg/L) for 21 d; and the indexes related to phenotypic traits, reproduction and gene expression were measured in tested animals. The results showed that heart rate and total number of neonates in exposed F0-daphnias were significantly lower than those of control group, and the detoxification genes (HR96 and P-gp) were up-regulated while genes related reproduction (Vtg) and molting (Nvd and Shd) were significantly down-regulated. The heart rate and individual size of F1-daphnias (<24 h) were significantly reduced in the treatment group. After 21-d recovery, the heart rate and expression of HR96, P-gp, Vtg, Nvd and Shd were declined in F1-daphnias. There was no obvious difference of morphological traits and heart rate between treatment and control in F2-daphnias (<24 h). In summary, daphnias (F0) exposure to triazophos with environmental dose could raise toxic effects on its offspring (F1), which is mainly manifested by reduced heart rate, the accumulated number and individual size of offspring and decreased expression of genes related to molting and reproduction.
Collapse
Affiliation(s)
- Shaoquan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jianlu Zhang
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Zoology, Xi'an 710032, China
| | - Ruijiao Li
- Fisheries Research & Technology Extension Center of Shaanxi, Yellow River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Xi'an 710086, China
| | - Chunyun Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lixin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hao Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Guangpeng Feng
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China.
| | - Dongmei Xiong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
10
|
Palaniyandi J, Bruin JE, Kumarathasan P, MacPherson S, Borghese MM, Ashley-Martin J. Prenatal exposure to perfluoroalkyl substances and inflammatory biomarker concentrations. Environ Epidemiol 2023; 7:e262. [PMID: 37545803 PMCID: PMC10403040 DOI: 10.1097/ee9.0000000000000262] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) are persistent environmental contaminants that induce immunotoxicity in experimental studies; however, epidemiological evidence-particularly during pregnancy-is scarce. We quantified associations between first trimester plasma perfluorooctanoic acid (PFOA), perfluorooctane sulfonate (PFOS), and perfluorohexane sulfonate (PFHxS) concentrations and third trimester concentrations of inflammatory biomarkers and determined if these associations were modified by fetal sex. Methods We analyzed data from 1411 participants, recruited between 2008 and 2011, in the Maternal-Infant Research on Environmental Chemicals study. Our primary outcome was a composite inflammatory index derived by summing the z-scores of eight proinflammatory biomarkers. Using multivariable linear regression models, we quantified associations between each PFAS and the inflammatory index and individual biomarkers. We quantified the effects of the PFAS mixture using weighted quantile sum regression, and evaluated effect modification using product terms and sex-stratified models. Results Each doubling of PFOA and PFHxS was associated with a 0.38 (95% CI, 0.09, 0.67) and 0.21 (95% CI, 0.01, 0.41) SD increase in the proinflammatory index, respectively. A one-quartile increase in the PFAS mixture was associated with a 0.40 (95% CI, 0.09, 0.71) SD increase in the proinflammatory index. In individual models, we observed positive associations between PFAS and concentrations of monocyte chemoattractant protein-1, macrophage inflammatory protein-1β, and matrix metalloproteinases-9; however, the magnitude and precision varied according to the specific PFAS. Sex-specific findings were identified in few PFAS-biomarker associations. Conclusions PFOA, PFOS, and PFHxS, individually and as a mixture, were positively associated with proinflammatory biomarkers during pregnancy.
Collapse
Affiliation(s)
- Jana Palaniyandi
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario
| | - Jennifer E. Bruin
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario
| | | | - Susan MacPherson
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario
| | - Michael M. Borghese
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario
| | | |
Collapse
|
11
|
Husain I, Dale OR, Martin K, Gurley BJ, Adams SJ, Avula B, Chittiboyina AG, Khan IA, Khan SI. Screening of medicinal plants for possible herb-drug interactions through modulating nuclear receptors, drug-metabolizing enzymes and transporters. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115822. [PMID: 36223846 DOI: 10.1016/j.jep.2022.115822] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The last three decades have witnessed a surge in popularity and consumption of herbal products. An unintended consequence of such popularity is that chronic consumption of these products can often modulate the functions of various proteins involved in drug disposition and may, in turn, impose risks for herb-drug interactions (HDIs), leading to serious adverse health outcomes. Identifying plants that may give rise to clinically relevant HDIs is essential, and proactive dissemination of such research outcomes is necessary for researchers, clinicians, and average consumers. AIM OF THE STUDY The main objective of this study was to evaluate the HDI potential of plants commonly used as ingredients in many herbal products, including BDS. MATERIALS AND METHODS The dried material of 123 plants selected from the NCNPR repository was extracted with 95% ethanol. The extracts were screened for agonistic effects on nuclear receptors (PXR and AhR) by reporter gene assays in PXR-transfected HepG2 and AhR-reporter cells. For cytochrome P450 enzyme (CYP) inhibition studies, CYP450 baculosomes were incubated with enzyme-specific probe substrates by varying concentrations of extracts. The inhibitory effect on the efflux transporter P-glycoprotein (P-gp) was investigated via rhodamine (Rh-123) uptake assay in P-gp overexpressing MDR1-MDCK cells. RESULTS Out of 123 plants, 16 increased transcriptional activity of human PXR up to 4 to 7-fold at 60 μg/mL, while 18 plants were able to increase AhR activity up to 10 to 40-fold at 30 μg/mL. Thirteen plants inhibited the activity of CYP3A4, while 10 plants inhibited CYP1A2 activity with IC50 values in the range of 1.3-10 μg/mL. Eighteen plants (at 50 μg/mL) increased intracellular accumulation of Rh-123 (>150%) in MDR1-MDCK cells. Additionally, other plants tested in this study were able to activate PXR, AhR, or both to lesser extents, and several inhibited the catalytic activity of CYPs at higher concentrations (IC50 >10 μg/mL). CONCLUSIONS The results indicate that prolonged or excessive consumption of herbal preparations rich in such plants (presented in Figs. 1a, 2a, 3a, 4a, and 5a) may pose a risk for CYP- and P-gp-mediated HDIs, leading to unwanted side effects due to the altered pharmacokinetics of concomitantly ingested medications.
Collapse
Affiliation(s)
- Islam Husain
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Olivia R Dale
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Katherine Martin
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Bill J Gurley
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Sebastian J Adams
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Bharathi Avula
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Amar G Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Ikhlas A Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States; Department of Bio-Molecular Sciences, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States
| | - Shabana I Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States; Department of Bio-Molecular Sciences, School of Pharmacy, The University of Mississippi, Mississippi, 38677, United States.
| |
Collapse
|
12
|
Heintz MM, Eccles JA, Olack EM, Maner-Smith KM, Ortlund EA, Baldwin WS. Human CYP2B6 produces oxylipins from polyunsaturated fatty acids and reduces diet-induced obesity. PLoS One 2022; 17:e0277053. [PMID: 36520866 PMCID: PMC9754190 DOI: 10.1371/journal.pone.0277053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple factors in addition to over consumption lead to obesity and non-alcoholic fatty liver disease (NAFLD) in the United States and worldwide. CYP2B6 is the only human detoxification CYP whose loss is associated with obesity, and Cyp2b-null mice show greater diet-induced obesity with increased steatosis than wildtype mice. However, a putative mechanism has not been determined. LC-MS/MS revealed that CYP2B6 metabolizes PUFAs, with a preference for metabolism of ALA to 9-HOTrE and to a lesser extent 13-HOTrE with a preference for metabolism of PUFAs at the 9- and 13-positions. To further study the role of CYP2B6 in vivo, humanized-CYP2B6-transgenic (hCYP2B6-Tg) and Cyp2b-null mice were fed a 60% high-fat diet for 16 weeks. Compared to Cyp2b-null mice, hCYP2B6-Tg mice showed reduced weight gain and metabolic disease as measured by glucose tolerance tests, however hCYP2B6-Tg male mice showed increased liver triglycerides. Serum and liver oxylipin metabolite concentrations increased in male hCYP2B6-Tg mice, while only serum oxylipins increased in female hCYP2B6-Tg mice with the greatest increases in LA oxylipins metabolized at the 9 and 13-positions. Several of these oxylipins, specifically 9-HODE, 9-HOTrE, and 13-oxoODE, are PPAR agonists. RNA-seq data also demonstrated sexually dimorphic changes in gene expression related to nuclear receptor signaling, especially CAR > PPAR with qPCR suggesting PPARγ signaling is more likely than PPARα signaling in male mice. Overall, our data indicates that CYP2B6 is an anti-obesity enzyme, but probably to a lesser extent than murine Cyp2b's. Therefore, the inhibition of CYP2B6 by xenobiotics or dietary fats can exacerbate obesity and metabolic disease potentially through disrupted PUFA metabolism and the production of key lipid metabolites.
Collapse
Affiliation(s)
- Melissa M. Heintz
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Jazmine A. Eccles
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Emily M. Olack
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Kristal M. Maner-Smith
- Emory Integrated Metabolomics and Lipodomics Core, Emory University, Atlanta, Georgia, United States of America
| | - Eric A. Ortlund
- Department of Biochemistry, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
13
|
Short-Term High-Fat Diet Alters Acetaminophen Metabolism in Healthy Individuals. Ther Drug Monit 2022; 44:797-804. [PMID: 35500453 DOI: 10.1097/ftd.0000000000000993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/28/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Acetaminophen is metabolized through a nontoxic sulfation and glucuronidation pathway and toxic oxidation pathway (via CYP2E1 and CYP1A2). A short-term high-fat diet induces alterations in the steatotic liver and may alter hepatic drug enzyme activity. In the case of acetaminophen, these alterations may result in an increased risk of hepatotoxicity. Therefore, this study was conducted to assess the effect of a 3-day hypercaloric high-fat diet on the plasma levels of acetaminophen metabolites. METHODS Nine healthy subjects participated in this randomized, crossover intervention study. The subjects consumed a regular diet or a regular diet supplemented with 500 mL of cream (1700 kcal) for 3 days and then fasted overnight. After ingesting 1000-mg acetaminophen, the plasma concentration of acetaminophen (APAP) and its metabolites [acetaminophen glucuronide, acetaminophen sulfate, 3-cysteinyl-acetaminophen, and 3-(N-acetyl-L-cystein-S-yl)-acetaminophen, and 3-methoxy-acetaminophen] were measured. RESULTS The 3-day high-fat diet increased the extrapolated area under the concentration-time curve from 0 to infinity (area under the curve 0-inf ) of APAP-Cys by approximately 20% ( P = 0.02) and that from 0 to 8 hours (area under the curve 0-8 ) of APAP-Cys-NAC by approximately 39% ( P = 0.01). The 3-day high-fat diet did not alter the pharmacokinetic parameters of the parent compound acetaminophen and other metabolites. CONCLUSIONS A short-term, hypercaloric, high-fat diet increases the plasma levels of the APAP metabolites formed by the oxidation pathway, which may increase the risk of hepatotoxicity.
Collapse
|
14
|
Içoglu Aksakal F, Yilmaz A, Koc K, Özdemir S. A comparative study on aquatic toxicity of chemically-synthesized and green synthesis silver nanoparticles on daphnia magna. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:2149-2159. [PMID: 34191661 DOI: 10.1080/09603123.2021.1947991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/22/2021] [Indexed: 06/13/2023]
Abstract
The steady increase in the employment of silver nanoparticles (AgNPs) in consumer products entails the determination of the aquatic toxicity of AgNPs. Various AgNP characteristics including particle size, and shape, surface charge, and material have prominent effects on ecotoxicity. In the present study, we investigated the aquatic toxicity of chemically-synthesized AgNPs (Che-AgNPs) and green synthesis AgNPs (Gr-AgNPs) to Daphnia magna as a model organism. In each case, Che-AgNPs and Gr-AgNPs showed dose-dependent toxicity in the range of 5-50 ppb. It was also detected that the size and surface coverage material of AgNPs has a significant impact on the survival rate of D. magna. We also analyzed the expression of some genes related to detoxification and the reproductive system. These observations presented that in both NP types the significant alterations were detected in genes of the model organism in a dose-dependent manner.
Collapse
Affiliation(s)
- Feyza Içoglu Aksakal
- Department of Agricultural Biotechnology, Faculty of Agriculture, Atatürk University, Erzurum, Turkey
| | - Asli Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Kubra Koc
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, TURKEY
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, Erzurum, Turkey
| |
Collapse
|
15
|
Baumert BO, Goodrich JA, Hu X, Walker DI, Alderete TL, Chen Z, Valvi D, Rock S, Berhane K, Gilliland FD, Goran MI, Jones DP, Conti DV, Chatzi L. Plasma concentrations of lipophilic persistent organic pollutants and glucose homeostasis in youth populations. ENVIRONMENTAL RESEARCH 2022; 212:113296. [PMID: 35447156 PMCID: PMC9831292 DOI: 10.1016/j.envres.2022.113296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/15/2022] [Accepted: 04/09/2022] [Indexed: 05/16/2023]
Abstract
BACKGROUND Exposure to lipophilic persistent organic pollutants (POPs) is ubiquitous. POPs are metabolic disrupting chemicals and are potentially diabetogenic. METHODS Using a multi-cohort study including overweight adolescents from the Study of Latino Adolescents at Risk (SOLAR, N = 301, 2001-2012) and young adults from the Southern California Children's Health Study (CHS, N = 135, 2014-2018), we examined associations of POPs and risk factors for type 2 diabetes. SOLAR participants underwent annual visits for a median of 2.2 years and CHS participants performed a single visit, during which a 2-h oral glucose tolerance test was performed. Linear mixed models were used to examine associations between plasma concentrations of POPs [4,4'-dichlorodiphenyldichloroethylene (4,4'-DDE), hexachlorobenzene (HCB), PCBs-153, 138, 118, 180 and PBDEs-154, 153, 100, 85, 47] and changes in glucose homeostasis across age and pubertal stage. RESULTS In SOLAR, exposure to HCB, PCB-118, and PBDE-153 was associated with dysregulated glucose metabolism. For example, each two-fold increase in HCB was associated with approximately 2 mg/dL higher glucose concentrations at 30 min (p = 0.001), 45 min (p = 0.0006), and 60 min (p = 0.03) post glucose challenge. Compared to individuals with low levels of PCB-118, individuals with high levels exhibited a 4.7 mg/dL (p = 0.02) higher glucose concentration at 15 min and a 3.6 mg/dL (p = 0.01) higher glucose concentration at 30 min. The effects observed with exposure to organochlorine compounds were independent of pubertal stages. PBDE-153 was associated with the development of dysregulated glucose metabolism beginning in late puberty. At Tanner stage 4, exposure to PBDE-153 was associated with a 12.7 mg/dL higher 60-min glucose concentration (p = 0.009) and a 16.1 mg*dl-1*hr-1 higher glucose AUC (p = 0.01). These associations persisted at Tanner 5. In CHS, PBDE-153 and total PBDE were associated with similar increases in glucose concentrations. CONCLUSION Our results suggest that childhood exposure to lipophilic POPs is associated with dysregulated glucose metabolism.
Collapse
Affiliation(s)
- Brittney O Baumert
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States.
| | - Jesse A Goodrich
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xin Hu
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Zhanghua Chen
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sarah Rock
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kiros Berhane
- Department of Biostatistics, Columbia University, New York, NY, United States
| | - Frank D Gilliland
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michael I Goran
- Department of Pediatrics, Children's Hospital of Los Angeles, The Saban Research Institute, United States
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States
| | - David V Conti
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Leda Chatzi
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
16
|
Bone C, Squires EJ. Nuclear Receptor Pathways Mediating the Development of Boar Taint. Metabolites 2022; 12:metabo12090785. [PMID: 36144190 PMCID: PMC9503508 DOI: 10.3390/metabo12090785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022] Open
Abstract
The nuclear receptors PXR, CAR, and FXR are activated by various ligands and function as transcription factors to control the expression of genes that regulate the synthesis and metabolism of androstenone and skatole. These compounds are produced in entire male pigs and accumulate in the fat to cause the development of a meat quality issue known as boar taint. The extent of this accumulation is influenced by the synthesis and hepatic clearance of androstenone and skatole. For this reason, PXR, CAR, and FXR-mediated signaling pathways have garnered interest as potential targets for specialized treatments designed to reduce the development of boar taint. Recent research has also identified several metabolites produced by gut microbes that act as ligands for these nuclear receptors (e.g., tryptophan metabolites, short-chain fatty acids, bile acids); however, the connection between the gut microbiome and boar taint development is not clear. In this review, we describe the nuclear receptor signaling pathways that regulate the synthesis and metabolism of boar taint compounds and outline the genes involved. We also discuss several microbial-derived metabolites and dietary additives that are known or suspected nuclear receptor ligands and suggest how these compounds could be used to develop novel treatments for boar taint.
Collapse
|
17
|
Guo H, Chen J, Zhang H, Yao J, Sheng N, Li Q, Guo Y, Wu C, Xie W, Dai J. Exposure to GenX and Its Novel Analogs Disrupts Hepatic Bile Acid Metabolism in Male Mice. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:6133-6143. [PMID: 34427428 DOI: 10.1021/acs.est.1c02471] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Due to its wide usage and recent detection in environmental matrices, hexafluoropropylene oxide dimer acid (HFPO-DA, commercial name GenX) has attracted considerable attention. Here, we explored and compared the toxicity of GenX and its novel analogs with that of perfluorooctanoic acid (PFOA) to provide guidance on the structural design and optimization of novel alternatives to poly- and perfluoroalkyl substances (PFASs). Adult male BALB/c mice were continuously exposed to PFOA, GenX, perfluoro-2-methyl-3,6-dioxo-heptanoic acid (PFMO2HpA), and perfluoro-2-methyl-3,6,8-trioxo-nonanoic acid (PFMO3NA; 0, 0.4, 2, or 10 mg/kg/d) via oral gavage for 28 days. The PFOA, GenX, and PFMO3NA treatment groups showed an increase in relative liver weight, and bile acid metabolism was the most significantly affected pathway in all treatment groups, as shown via weighted gene coexpression network analysis. The highest total bile acid levels were observed in the 2 and 10 mg/kg/d PFMO3NA groups. The ratios of primary bile acids to all bile acids increased in the high-dose groups, while the ratios of secondary bile acids showed a downward trend. Thus, bile acid metabolism disorder may be a prominent adverse effect induced by exposure to GenX, its analogs, and PFOA. Results also showed that the hepatotoxicity of PFMO2HpA was lower than that of GenX, whereas the hepatotoxicity of PFMO3NA was stronger, suggesting that PFMO2HpA may be a potential alternative to GenX.
Collapse
Affiliation(s)
- Hua Guo
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiamiao Chen
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Hongxia Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingzhi Yao
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Nan Sheng
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Qi Li
- Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Chengying Wu
- Sanming Hexafluo Chemicals Co., Ltd., Fluorinated New Material Industry Park, Mingxi, Fujian 365200, China
| | - Weidong Xie
- Sanming Hexafluo Chemicals Co., Ltd., Fluorinated New Material Industry Park, Mingxi, Fujian 365200, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
18
|
Wang X, Yu Y, Wang P, Yang K, Wang Y, Yan L, Zhong XB, Zhang L. Long Noncoding RNAs Hepatocyte Nuclear Factor 4A Antisense RNA 1 and Hepatocyte Nuclear Factor 1A Antisense RNA 1 Are Involved in Ritonavir-Induced Cytotoxicity in Hepatoma Cells. Drug Metab Dispos 2022; 50:704-715. [PMID: 34949673 PMCID: PMC9132102 DOI: 10.1124/dmd.121.000693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022] Open
Abstract
Ritonavir (RTV), a pharmacoenhancer used in anti-HIV regimens, can induce liver damage. RTV is primarily metabolized by cytochrome P450 3A4 (CYP3A4) in the liver. HNF4A antisense RNA 1 (HNF4A-AS1) and HNF1A antisense RNA 1 (HNF1A-AS1) are long noncoding RNAs that regulate the expression of pregnane X receptor (PXR) and CYP3A4. This study investigated the role and underlying mechanisms of HNF4A-AS1 and HNF1A-AS1 in RTV-induced hepatotoxicity. HNF4A-AS1 and HNF1A-AS1 were knocked down by small hairpin RNAs in Huh7 and HepG2 cells. Lactate dehydrogenase and reactive oxygen species assays were performed to assess RTV-induced hepatotoxicity. Chromatin immunoprecipitation quantitative real-time polymerase chain reaction was used to detect PXR enrichment and histone modifications in the CYP3A4 promoter. HNF4A-AS1 knockdown increased PXR and CYP3A4 expression and exacerbated RTV-induced cytotoxicity, whereas HNF1A-AS1 knockdown generated the opposite phenotype. Mechanistically, enrichment of PXR and trimethylation of histone 3 lysine 4 (H3K4me3) in the CYP3A4 promoter was increased, and trimethylation of histone 3 lysine 27 (H3K27me3) was decreased after HNF4A-AS1 knockdown. However, PXR and H3K4me3 enrichment decreased after HNF1A-AS1 knockdown. Alterations in RTV-induced hepatotoxicity caused by decreasing HNF4A-AS1 or HNF1A-AS1 were reversed by knockdown or overexpression of PXR. Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. SIGNIFICANCE STATEMENT: HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as long noncoding RNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. This discovery provides directions for further research on the mechanisms of RTV-induced liver injury.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Yihang Yu
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Kun Yang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Yiting Wang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Liang Yan
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Xiao-Bo Zhong
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences (X.W., Y.Y., P.W., K.Y., Y.W., L.Z.) and State Key Laboratory for Esophageal Cancer Prevention and Treatment, (L.Z.) Zhengzhou University, Zhengzhou, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-B.Z.)
| |
Collapse
|
19
|
Hoyeck MP, Matteo G, MacFarlane EM, Perera I, Bruin JE. Persistent organic pollutants and β-cell toxicity: a comprehensive review. Am J Physiol Endocrinol Metab 2022; 322:E383-E413. [PMID: 35156417 PMCID: PMC9394781 DOI: 10.1152/ajpendo.00358.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 01/09/2023]
Abstract
Persistent organic pollutants (POPs) are a diverse family of contaminants that show widespread global dispersion and bioaccumulation. Humans are continuously exposed to POPs through diet, air particles, and household and commercial products; POPs are consistently detected in human tissues, including the pancreas. Epidemiological studies show a modest but consistent correlation between exposure to POPs and increased diabetes risk. The goal of this review is to provide an overview of epidemiological evidence and an in-depth evaluation of the in vivo and in vitro evidence that POPs cause β-cell toxicity. We review evidence for six classes of POPs: dioxins, polychlorinated biphenyls (PCBs), organochlorine pesticides (OCPs), organophosphate pesticides (OPPs), flame retardants, and per- and polyfluoroalkyl substances (PFAS). The available data provide convincing evidence implicating POPs as a contributing factor driving impaired glucose homeostasis, β-cell dysfunction, and altered metabolic and oxidative stress pathways in islets. These findings support epidemiological data showing that POPs increase diabetes risk and emphasize the need to consider the endocrine pancreas in toxicity assessments. Our review also highlights significant gaps in the literature assessing islet-specific endpoints after both in vivo and in vitro POP exposure. In addition, most rodent studies do not consider the impact of biological sex or secondary metabolic stressors in mediating the effects of POPs on glucose homeostasis and β-cell function. We discuss key gaps and limitations that should be assessed in future studies.
Collapse
Affiliation(s)
- Myriam P Hoyeck
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Geronimo Matteo
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Erin M MacFarlane
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Ineli Perera
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Jennifer E Bruin
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Dutta M, Lim JJ, Cui JY. Pregnane X Receptor and the Gut-Liver Axis: A Recent Update. Drug Metab Dispos 2022; 50:478-491. [PMID: 34862253 PMCID: PMC11022899 DOI: 10.1124/dmd.121.000415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023] Open
Abstract
It is well-known that the pregnane X receptor (PXR)/Nr1i2 is a critical xenobiotic-sensing nuclear receptor enriched in liver and intestine and is responsible for drug-drug interactions, due to its versatile ligand binding domain (LBD) and target genes involved in xenobiotic biotransformation. PXR can be modulated by various xenobiotics including pharmaceuticals, nutraceuticals, dietary factors, and environmental chemicals. Microbial metabolites such as certain secondary bile acids (BAs) and the tryptophan metabolite indole-3-propionic acid (IPA) are endogenous PXR activators. Gut microbiome is increasingly recognized as an important regulator for host xenobiotic biotransformation and intermediary metabolism. PXR regulates and is regulated by the gut-liver axis. This review summarizes recent research advancements leveraging pharmaco- and toxico-metagenomic approaches that have redefined the previous understanding of PXR. Key topics covered in this review include: (1) genome-wide investigations on novel PXR-target genes, novel PXR-DNA interaction patterns, and novel PXR-targeted intestinal bacteria; (2) key PXR-modulating activators and suppressors of exogenous and endogenous sources; (3) novel bidirectional interactions between PXR and gut microbiome under physiologic, pathophysiological, pharmacological, and toxicological conditions; and (4) modifying factors of PXR-signaling including species and sex differences and time (age, critical windows of exposure, and circadian rhythm). The review also discusses critical knowledge gaps and important future research topics centering around PXR. SIGNIFICANCE STATEMENT: This review summarizes recent research advancements leveraging O'mics approaches that have redefined the previous understanding of the xenobiotic-sensing nuclear receptor pregnane X receptor (PXR). Key topics include: (1) genome-wide investigations on novel PXR-targeted host genes and intestinal bacteria as well as novel PXR-DNA interaction patterns; (2) key PXR modulators including microbial metabolites under physiological, pathophysiological, pharmacological, and toxicological conditions; and (3) modifying factors including species, sex, and time.
Collapse
Affiliation(s)
- Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Age- and Diet-Dependent Changes in Hepatic Lipidomic Profiles of Phospholipids in Male Mice: Age Acceleration in Cyp2b-Null Mice. J Lipids 2022; 2022:7122738. [PMID: 35391786 PMCID: PMC8983274 DOI: 10.1155/2022/7122738] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/03/2022] [Indexed: 11/17/2022] Open
Abstract
Increases in traditional serum lipid profiles are associated with obesity, cancer, and cardiovascular disease. Recent lipidomic analysis has indicated changes in serum lipidome profiles, especially in regard to specific phosphatidylcholines, associated with obesity. However, little work has evaluated murine hepatic liver lipidomic profiles nor compared these profiles across age, high-fat diet, or specific genotypes, in this case the lack of hepatic Cyp2b enzymes. In this study, the effects of age (9 months old), high-fat diet (4.5 months old), and the loss of three primarily hepatic xeno- and endobiotic metabolizing cytochrome P450 (Cyp) enzymes, Cyp2b9, Cyp2b10, and Cyp2b13 (Cyp2b-null mice), on the male murine hepatic lipidome were compared. Hierarchical clustering and principal component analysis show that age perturbs hepatic phospholipid profiles and serum lipid markers the most compared to young mice, followed by a high-fat diet and then loss of Cyp2b. Several lipid biomarkers such as PC/PE ratios, PE 38 : 6, and LPC concentrations indicate greater potential for NAFLD and hypertension with mixed effects in Cyp2b-null mice(less NAFLD and greater hypertension-associated markers). Lipid profiles from older mice contain greater total and n-6 fatty acids than normal diet (ND)-fed young mice; however, surprisingly, young Cyp2b-null mice contain high n-6 : n-3 ratios. Overall, the lack of Cyp2b typically enhanced adverse physiological parameters observed in the older (9 mo) mice with increased weight gain combined with a deteriorating cholesterol profile, but not necessarily all phospholipid profiles were adversely perturbed.
Collapse
|
22
|
Rahunen R, Kummu O, Koivukangas V, Hautajärvi H, Hakkola J, Rysä J, Hukkanen J. Pregnane X Receptor‒4β-Hydroxycholesterol Axis in the Regulation of Overweight- and Obesity-Induced Hypertension. J Am Heart Assoc 2022; 11:e023492. [PMID: 35229613 PMCID: PMC9075316 DOI: 10.1161/jaha.121.023492] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Mechanisms mediating hypertensive effects of overweight and obesity have not been fully elucidated. We showed previously that activation of pregnane X receptor (PXR) by rifampicin elevates 24-hour blood pressure (BP) and plasma 4β-hydroxycholesterol (4βHC), agonist for liver X receptor (LXR). Methods and Results In combined "PXR activation data set" (n=62) of 4 clinical trials, 1 week rifampicin dosing increased office systolic BP (SBP) by 3.1 mm Hg, DBP 1.8 mm Hg, and mean arterial pressure 2.2 mm Hg in comparison with placebo (P<0.01). Plasma 4βHC had negative correlation with SBP both in rifampicin (r=-0.46, P=0.0002) and placebo (r=-0.45, P=0.0003) arms, although 4βHC was elevated >3-fold by rifampicin. In "non-intervention data set" (n=102) of patients with obesity and healthy volunteers (body mass index, 19.2-55.2 kg/m2), 4βHC had negative correlations (P<0.00001) with office SBP (r=-0.51), diastolic BP (r=-0.50), and mean arterial pressure (r=-0.54). Lean women had higher 4βHC than men, with increasing weight repressing 4βHC (r=-0.62, P<0.00001) in both sexes. In multiple linear regression analysis, the only statistically significant predictor for SBP was 4βHC. Six-day PXR agonist dosing elevated SBP in rats (n=7-8/group). PXR activation elevated 4βHC and after PXR agonist was withdrawn and elevated 4βHC was left to act alone, SBP was reduced on days 7 to 14 in comparison with control rats. Conclusions PXR activation elevates SBP. Elevated circulating 4βHC lowers SBP in rats, and higher 4βHC is an independent predictor of lower SBP in humans. PXR-4βHC-LXR is novel BP-regulating pathway deregulated in overweight and obesity by repressed 4βHC, with implications for sex-specific BP regulation. Registration URL: https://www.clinicaltrials.gov; Unique identifiers: NCT00985270, NCT01293422, NCT01690104, NCT02329405, and NCT01330251.
Collapse
Affiliation(s)
- Roosa Rahunen
- Research Unit of Internal Medicine University of Oulu Finland.,Biocenter Oulu University of Oulu Finland.,Medical Research Center Oulu Oulu University Hospital and University of Oulu Finland
| | - Outi Kummu
- Biocenter Oulu University of Oulu Finland.,Medical Research Center Oulu Oulu University Hospital and University of Oulu Finland.,Research Unit of Biomedicine Pharmacology and Toxicology University of Oulu Finland
| | - Vesa Koivukangas
- Medical Research Center Oulu Oulu University Hospital and University of Oulu Finland.,Department of Surgery Oulu University Hospital and University of Oulu Finland
| | - Heidi Hautajärvi
- Admescope Ltd. Oulu Finland.,Now with Finnish Customs Laboratory Finland
| | - Jukka Hakkola
- Biocenter Oulu University of Oulu Finland.,Medical Research Center Oulu Oulu University Hospital and University of Oulu Finland.,Research Unit of Biomedicine Pharmacology and Toxicology University of Oulu Finland
| | - Jaana Rysä
- School of Pharmacy Faculty of Health Sciences University of Eastern Finland Kuopio Finland
| | - Janne Hukkanen
- Research Unit of Internal Medicine University of Oulu Finland.,Biocenter Oulu University of Oulu Finland.,Medical Research Center Oulu Oulu University Hospital and University of Oulu Finland
| |
Collapse
|
23
|
Shin A, Waxman DJ. Impact of neonatal activation of nuclear receptor CAR (Nr1i3) on Cyp2 gene expression in adult mouse liver. Toxicol Sci 2022; 187:298-310. [PMID: 35285501 DOI: 10.1093/toxsci/kfac032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Perinatal exposure to environmental chemicals is proposed to reprogram development and alter disease susceptibility later in life. Supporting this, neonatal activation of the nuclear receptor CAR (Nr1i3) by TCPOBOP induces persistent expression of mouse hepatic Cyp2 genes into adulthood, attributed to long-term epigenetic memory of the early life exposure [Hepatology (2012) 56:1499-1509]. Here, we confirm that the same high-dose (15x ED50) neonatal TCPOBOP exposure used in that work induces prolonged (12 weeks) increases in hepatic Cyp2 expression; however, we show that the persistence of expression can be fully explained by the persistence of residual TCPOBOP in liver tissue. When the long-term presence of TCPOBOP in tissue was eliminated by decreasing the neonatal TCPOBOP dose 22-fold (0.67x ED50), strong neonatal increases in hepatic Cyp2 expression were still obtained but did not persist into adulthood. Furthermore, the neonatal ED50-range TCPOBOP exposure did not sensitize mice to a subsequent, low-dose TCPOBOP treatment. In contrast, neonatal treatment with phenobarbital, a short half-life (t1/2=8 h) agonist of CAR and of PXR (Nr1i2), induced high-level neonatal activation of Cyp2 genes and also altered their responsiveness to low-dose phenobarbital exposure at adulthood by either increasing (Cyp2b10) or decreasing (Cyp2c55) expression. Thus, neonatal xenobiotic exposure can reprogram hepatic Cyp2 genes and alter their responsiveness to exposures later in life. These findings highlight the need to carefully consider xenobiotic dose, half-life and persistence in tissue when evaluating the long-term effects of early life environmental chemical exposures.
Collapse
Affiliation(s)
- Aram Shin
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| |
Collapse
|
24
|
A biologically based model to quantitatively assess the role of the nuclear receptors liver X (LXR), and pregnane X (PXR) on chemically induced hepatic steatosis. Toxicol Lett 2022; 359:46-54. [PMID: 35143881 PMCID: PMC9644840 DOI: 10.1016/j.toxlet.2022.02.002] [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: 10/05/2021] [Revised: 01/24/2022] [Accepted: 02/03/2022] [Indexed: 11/21/2022]
Abstract
Hepatic steatosis is characterized by the intracellular increase of free fatty acids (FFAs) in the form of triglycerides in hepatocytes. This hepatic adverse outcome can be caused by many factors, including exposure to drugs or environmental toxicants. Mechanistically, accumulation of lipids in the liver can take place via several mechanisms such as de novo synthesis and/or uptake of FFAs from serum via high fat content diets. De novo synthesis of FFAs within the liver is mediated by the liver X receptor (LXR), and their uptake into the liver is mediated through the pregnane X receptor (PXR). We investigated the impact of chemical exposure on FFAs hepatic content via activation of LXR and PXR by integrating chemical-specific physiologically based pharmacokinetic (PBPK) models with a quantitative toxicology systems (QTS) model of hepatic lipid homeostasis. Three known agonists of LXR and/or PXR were modeled: T0901317 (antagonist for both receptors), GW3965 (LXR only), and Rifampicin (PXR only). Model predictions showed that T0901317 caused the most FFAs build-up in the liver, followed by Rifampicin and then GW3965. These modeling results highlight the importance of PXR activation for serum FFAs uptake into the liver while suggesting that increased hepatic FAAs de novo synthesis alone may not be enough to cause appreciable accumulation of lipids in the liver under normal environmental exposure levels. Moreover, the overall PBPK-hepatic lipids quantitative model can be used to screen chemicals for their potential to cause in vivo hepatic lipid content buildup in view of their in vitro potential to activate the nuclear receptors and their exposure levels.
Collapse
|
25
|
Khalil Y, Carrino S, Lin F, Ferlin A, Lad HV, Mazzacuva F, Falcone S, Rivers N, Banks G, Concas D, Aguilar C, Haynes AR, Blease A, Nicol T, Al-Shawi R, Heywood W, Potter P, Mills K, Gale DP, Clayton PT. Tissue Proteome of 2-Hydroxyacyl-CoA Lyase Deficient Mice Reveals Peroxisome Proliferation and Activation of ω-Oxidation. Int J Mol Sci 2022; 23:ijms23020987. [PMID: 35055171 PMCID: PMC8781152 DOI: 10.3390/ijms23020987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Peroxisomal fatty acid α-oxidation is an essential pathway for the degradation of β-carbon methylated fatty acids such as phytanic acid. One enzyme in this pathway is 2-hydroxyacyl CoA lyase (HACL1), which is responsible for the cleavage of 2-hydroxyphytanoyl-CoA into pristanal and formyl-CoA. Hacl1 deficient mice do not present with a severe phenotype, unlike mice deficient in other α-oxidation enzymes such as phytanoyl-CoA hydroxylase deficiency (Refsum disease) in which neuropathy and ataxia are present. Tissues from wild-type and Hacl1−/− mice fed a high phytol diet were obtained for proteomic and lipidomic analysis. There was no phenotype observed in these mice. Liver, brain, and kidney tissues underwent trypsin digestion for untargeted proteomic liquid chromatography-mass spectrometry analysis, while liver tissues also underwent fatty acid hydrolysis, extraction, and derivatisation for fatty acid gas chromatography-mass spectrometry analysis. The liver fatty acid profile demonstrated an accumulation of phytanic and 2-hydroxyphytanic acid in the Hacl1−/− liver and significant decrease in heptadecanoic acid. The liver proteome showed a significant decrease in the abundance of Hacl1 and a significant increase in the abundance of proteins involved in PPAR signalling, peroxisome proliferation, and omega oxidation, particularly Cyp4a10 and Cyp4a14. In addition, the pathway associated with arachidonic acid metabolism was affected; Cyp2c55 was upregulated and Cyp4f14 and Cyp2b9 were downregulated. The kidney proteome revealed fewer significantly upregulated peroxisomal proteins and the brain proteome was not significantly different in Hacl1−/− mice. This study demonstrates the powerful insight brought by proteomic and metabolomic profiling of Hacl1−/− mice in better understanding disease mechanism in fatty acid α-oxidation disorders.
Collapse
Affiliation(s)
- Youssef Khalil
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
| | - Sara Carrino
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy
| | - Fujun Lin
- Department of Renal Medicine, University College London, London NW3 2PF, UK; (F.L.); (A.F.); (D.P.G.)
- Department of Nephrology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200082, China
| | - Anna Ferlin
- Department of Renal Medicine, University College London, London NW3 2PF, UK; (F.L.); (A.F.); (D.P.G.)
- Clinical Genetics and Genomics Laboratory, Royal Brompton Hospital, London SW3 6NP, UK
| | - Heena V. Lad
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Francesca Mazzacuva
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
- Department of Bioscience, University of East London, London E15 4LZ, UK
| | - Sara Falcone
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Natalie Rivers
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Gareth Banks
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Danilo Concas
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Carlos Aguilar
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Andrew R. Haynes
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Andy Blease
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Thomas Nicol
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Raya Al-Shawi
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, University College London, London NW3 2PF, UK;
| | - Wendy Heywood
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
| | - Paul Potter
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Kevin Mills
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
| | - Daniel P. Gale
- Department of Renal Medicine, University College London, London NW3 2PF, UK; (F.L.); (A.F.); (D.P.G.)
| | - Peter T. Clayton
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
- Correspondence:
| |
Collapse
|
26
|
Koga T, Peters JM. Targeting Peroxisome Proliferator-Activated Receptor-β/δ (PPARβ/δ) for the Treatment or Prevention of Alcoholic Liver Disease. Biol Pharm Bull 2021; 44:1598-1606. [PMID: 34719638 DOI: 10.1248/bpb.b21-00486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Excessive, chronic alcohol consumption can lead to alcoholic liver disease. The etiology of alcoholic liver disease is multifactorial and is influenced by alterations in gene expression and changes in fatty acid metabolism, oxidative stress, and insulin resistance. These events can lead to steatosis, fibrosis, and eventually to cirrhosis and liver cancer. Many of these functions are regulated by peroxisome proliferator-activated receptors (PPARs). Thus, it is not surprising that PPARs can modulate the mechanisms that cause alcoholic liver disease. While the roles of PPARα and PPARγ are clearer, the role of PPARβ/δ in alcoholic liver disease requires further clarification. This review summarizes the current understanding based on recent studies that indicate that PPARβ/δ can likely be targeted for the treatment and/or the prevention of alcoholic liver disease.
Collapse
Affiliation(s)
- Takayuki Koga
- Laboratory of Hygienic Chemistry, Department of Health Science and Hygiene, Daiichi University of Pharmacy
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University
| |
Collapse
|
27
|
Yuan KK, Duan GF, Liu QY, Li HY, Yang WD. Inhibition of Diarrheal Shellfish Toxins Accumulation in the Mussel Perna viridis by Curcumin and Underlying Mechanisms. Toxins (Basel) 2021; 13:toxins13080578. [PMID: 34437449 PMCID: PMC8402306 DOI: 10.3390/toxins13080578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 01/04/2023] Open
Abstract
Diarrheal shellfish toxins (DSTs) are among the most widely distributed phytotoxins, and are associated with diarrheal shellfish poisoning (DSP) events in human beings all over the world. Therefore, it is urgent and necessary to identify an effective method for toxin removal in bivalves. In this paper, we found that curcumin (CUR), a phytopolylphenol pigment, can inhibit the accumulation of DSTs (okadaic acid-eq) in the digestive gland of Perna viridis after Prorocentrum lima exposure. qPCR results demonstrated that CUR inhibited the induction of DSTs on the aryl hydrocarbon receptor (AhR), hormone receptor 96 (HR96) and CYP3A4 mRNA, indicating that the CUR-induced reduction in DSTs may be correlated with the inhibition of transcriptional induction of AhR, HR96 and CYP3A4. The histological examination showed that P. lima cells caused severe damage to the digestive gland of P. viridis, and the addition of curcumin effectively alleviated the damage induced by P. lima. In conclusion, our findings provide a potential method for the effective removal of toxins from DST-contaminated shellfish.
Collapse
|
28
|
Yang H, Lu G, Yan Z, Liu J. Influence of suspended sediment on the bioavailability of benzophenone-3: Focus on accumulation and multi-biological effects in Daphnia magna. CHEMOSPHERE 2021; 275:129974. [PMID: 33639549 DOI: 10.1016/j.chemosphere.2021.129974] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 06/12/2023]
Abstract
The UV-filter benzophenone-3 (BP3) tends to associate with suspended sediment (SPS) due to hydrophobicity, which could alter its toxicological effects on non-target aquatic organisms. In this study, the freshwater cladoceran Daphnia magna (D. magna) was selected as a model organism to investigate the impacts of the source and composition of SPS on the accumulation and multiple toxicological effects (from the molecular level to individual level) of BP3. Among the three components of SPS, amorphous organic carbon (AOC) and minerals promoted the body burden of BP3, while black carbon (BC) inhibited the bioaccumulation. The inhibition effects of BP3 on swimming and feeding behaviors of D. magna were also enhanced due to the presence of AOC and BC. Compared with BP3 exposure alone, higher oxidative stress and neurotoxicity were observed in the presence of SPS containing AOC, BC and minerals, corresponding to that superoxide dismutase, catalase and glutathione-S-transferase activities were further induced, and acetylcholinesterase activity was inhibited. Furthermore, BP3 induced mRNA expression levels of the endocrine system (ecdysone receptor, cytochrome P450 CYP314) and metabolic system (toxicant nuclear receptor HR96, P-glycoprotein), and the presence of SPS containing AOC, BC and minerals exhibited an enhanced effect. Combined with all endpoints, evident relationship was observed between the bioaccumulation level and the response of individual behavior and molecular biomarkers. The results demonstrated that the effects of SPS compositions on bioaccumulation and toxicological effects of organic UV-filters should be considered in aquatic environments.
Collapse
Affiliation(s)
- Haohan Yang
- Key Laboratory for Integrated Regulation and Resources Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, China
| | - Guanghua Lu
- Key Laboratory for Integrated Regulation and Resources Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, China; Water Conservancy Project & Civil Engineering College, Tibet Agriculture & Animal Husbandry University, Linzhi, 860000, China.
| | - Zhenhua Yan
- Key Laboratory for Integrated Regulation and Resources Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, China
| | - Jianchao Liu
- Key Laboratory for Integrated Regulation and Resources Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing, 210098, China
| |
Collapse
|
29
|
Wang P, Chen S, Wang Y, Wang X, Yan L, Yang K, Zhong XB, Han S, Zhang L. The Long Noncoding RNA Hepatocyte Nuclear Factor 4 α Antisense RNA 1 Negatively Regulates Cytochrome P450 Enzymes in Huh7 Cells via Histone Modifications. Drug Metab Dispos 2021; 49:361-368. [PMID: 33674270 DOI: 10.1124/dmd.120.000316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/02/2021] [Indexed: 01/22/2023] Open
Abstract
The maintenance of homeostasis of cytochromes P450 enzymes (P450s) under both physiologic and xenobiotic exposure conditions is ensured by the action of positive and negative regulators. In the current study, the hepatocyte nuclear factor 4α (HNF4A) antisense RNA 1 (HNF4A-AS1), an antisense long noncoding RNA of HNF4A, was found to be a negative regulator of the basal and rifampicin (RIF)-induced expression of nuclear receptors and downstream P450s. In Huh7 cells, knockdown of HNF4A-AS1 resulted in elevated expression of HNF4A, pregnane X receptor (PXR), and P450s (including CYP3A4) under both basal and RIF-induced conditions. Conversely, overexpression of HNF4A-AS1 led to decreased basal expression of constitutive androstane receptor, aryl hydrocarbon receptor, PXR, and all studied P450s. Of note, significantly diminished induction levels of PXR and CYP1A2, 2C8, 2C19, and 3A4 by RIF were also observed in HNF4A-AS1 plasmid-transfected Huh7 cells. Moreover, the negative feedback of HNF4A on HNF4A-AS1-mediated gene expression was validated using a loss-of-function experiment in this study. Strikingly, our data showed that increased enrichment levels of histone 3 lysine 4 trimethylation and HNF4A in the CYP3A4 promoter contribute to the elevated CYP3A4 expression after HNF4A-AS1 knockdown. Overall, the current study reveals that histone modifications contribute to the negative regulation of nuclear receptors and P450s by HNF4A-AS1 in basal and drug-induced levels. SIGNIFICANCE STATEMENT: Utilizing loss-of-function and gain-of-function experiments, the current study systematically investigated the negative regulation of HNF4A-AS1 on the expression of nuclear receptors (including HNF4A, constitutive androstane receptor, aryl hydrocarbon receptor, and pregnane X receptor) and P450s (including CYP1A2, 2E1, 2B6, 2D6, 2C8, 2C9, 2C19, and 3A4) in both basal and rifampicin-induced levels in Huh7 cells. Notably, this study is the first to reveal the contribution of histone modification to the HNF4A-AS1-mediated expression of CYP3A4 in Huh7 cells.
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Shitong Chen
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Yiting Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Xiaofei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Liang Yan
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Kun Yang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Xiao-Bo Zhong
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Shengna Han
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China (P.W., S.C., Y.W., X.W., K.Y., S.H., L.Z.); Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.-b.Z.)
| |
Collapse
|
30
|
Heusinkveld H, Braakhuis H, Gommans R, Botham P, Corvaro M, van der Laan JW, Lewis D, Madia F, Manou I, Schorsch F, Wolterink G, Woutersen R, Corvi R, Mehta J, Luijten M. Towards a mechanism-based approach for the prediction of nongenotoxic carcinogenic potential of agrochemicals. Crit Rev Toxicol 2020; 50:725-739. [DOI: 10.1080/10408444.2020.1841732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Harm Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Hedwig Braakhuis
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Robin Gommans
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | | | | | | | | | - Federica Madia
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Irene Manou
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Brussels, Belgium
| | | | - Gerrit Wolterink
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Ruud Woutersen
- TNO Quality of Life, Zeist, and Wageningen University & Research, Wageningen, the Netherlands
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
31
|
Oliviero F, Lukowicz C, Boussadia B, Forner-Piquer I, Pascussi JM, Marchi N, Mselli-Lakhal L. Constitutive Androstane Receptor: A Peripheral and a Neurovascular Stress or Environmental Sensor. Cells 2020; 9:E2426. [PMID: 33171992 PMCID: PMC7694609 DOI: 10.3390/cells9112426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
Xenobiotic nuclear receptors (NR) are intracellular players involved in an increasing number of physiological processes. Examined and characterized in peripheral organs where they govern metabolic, transport and detoxification mechanisms, accumulating data suggest a functional expression of specific NR at the neurovascular unit (NVU). Here, we focus on the Constitutive Androstane Receptor (CAR), expressed in detoxifying organs such as the liver, intestines and kidneys. By direct and indirect activation, CAR is implicated in hepatic detoxification of xenobiotics, environmental contaminants, and endogenous molecules (bilirubin, bile acids). Importantly, CAR participates in physiological stress adaptation responses, hormonal and energy homeostasis due to glucose and lipid sensing. We next analyze the emerging evidence supporting a role of CAR in NVU cells including the blood-brain barrier (BBB), a key vascular interface regulating communications between the brain and the periphery. We address the emerging concept of how CAR may regulate specific P450 cytochromes at the NVU and the associated relevance to brain diseases. A clear understanding of how CAR engages during pathological conditions could enable new mechanistic, and perhaps pharmacological, entry-points within a peripheral-brain axis.
Collapse
Affiliation(s)
- Fabiana Oliviero
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| | - Céline Lukowicz
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| | - Badreddine Boussadia
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Isabel Forner-Piquer
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Jean-Marc Pascussi
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Institute of Functional Genomics (UMR 5203 CNRS–U 1191 INSERM, University of Montpellier), 34094 Montpellier, France; (B.B.); (I.F.-P.); (J.-M.P.)
| | - Laila Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31027 Toulouse, France; (F.O.); (C.L.)
| |
Collapse
|
32
|
Strupp C, Quesnot N, Weber-Parmentier C, Richert L, Bomann WH, Singh P. Weight of Evidence and Human Relevance Evaluation of the Benfluralin Mode of Action in Rats (Part II): Thyroid carcinogenesis. Regul Toxicol Pharmacol 2020; 117:104736. [PMID: 32798613 DOI: 10.1016/j.yrtph.2020.104736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 11/08/2022]
Abstract
Benfluralin is an herbicide of the dinitroaniline class used to control grasses and weeds. In a 2 year dietary study in rats, benfluralin increased incidences of thyroid follicular adenoma and carcinoma at high dietary concentrations (≥2500 ppm). The benfluralin toxicology database suggests the mode of action (MOA) is initiated by induction of liver metabolizing enzymes, particularly thyroid hormone specific UGTs, a major pathway for T4 clearance in rats. As reported with phenobarbital, this effect triggers negative feedback regulation, increasing thyroid stimulating hormone (TSH) release into circulating blood. When sustained over time, this leads to thyroid changes such as follicular hypertrophy, hyperplasia and thyroid follicular tumors with chronic exposures. The described MOA was previously established in rat studies with various chemical activators of xenobiotic receptors in the liver. It is generally considered as non-relevant in humans, due to differences between humans and rats in T4 turnover and susceptibility to this carcinogenic MOA. A structured methodology based on the IPCS/MOA/Human Relevance framework was used in the evaluation of available benfluralin data, and the conclusion was determined that the carcinogenic potential of benfluralin in the thyroid is not relevant in humans.
Collapse
Affiliation(s)
- Christian Strupp
- Gowan, Highlands House, Basingstoke Road, Spencers Wood Reading, Berkshire, RG7 1NT, United Kingdom.
| | - Nicolas Quesnot
- Charles River Laboratories Evreux, 27005, Evreux Cedex, France.
| | | | | | - Werner H Bomann
- ToxConsult®, 9393 W 110th Street, 51 Corporate Woods, Suite 500, Overland Park, KS, 66210, USA.
| | - Pramila Singh
- Charles River Laboratories Evreux, 27005, Evreux Cedex, France.
| |
Collapse
|
33
|
Petrakis D, Margină D, Tsarouhas K, Tekos F, Stan M, Nikitovic D, Kouretas D, Spandidos DA, Tsatsakis A. Obesity ‑ a risk factor for increased COVID‑19 prevalence, severity and lethality (Review). Mol Med Rep 2020; 22:9-19. [PMID: 32377709 PMCID: PMC7248467 DOI: 10.3892/mmr.2020.11127] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Coronaviruses (CoVs), enveloped positive-sense RNA viruses, are a group of viruses that cause infections in the human respiratory tract, which can be characterized clinically from mild to fatal. The severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) is the virus responsible. The global spread of COVID‑19 can be described as the worst pandemic in humanity in the last century. To date, COVID‑19 has infected more than 3,000,000 people worldwide and killed more than 200,000 people. All age groups can be infected from the virus, but more serious symptoms that can possibly result in death are observed in older people and those with underlying medical conditions such as cardiovascular and pulmonary disease. Novel data report more severe symptoms and even a negative prognosis for the obese patients. A growing body of evidence connects obesity with COVID‑19 and a number of mechanisms from immune system activity attenuation to chronic inflammation are implicated. Lipid peroxidation creates reactive lipid aldehydes which in a patient with metabolic disorder and COVID‑19 will affect its prognosis. Finally, pregnancy‑associated obesity needs to be studied further in connection to COVID‑19 as this infection could pose high risk both to pregnant women and the fetus.
Collapse
Affiliation(s)
- Demetrios Petrakis
- Laboratory of Toxicology, Medical School, University of Crete, 71409 Heraklion, Greece
| | - Denisa Margină
- ‘Carol Davila’ University of Medicine and Pharmacy, Faculty of Pharmacy, Department of Biochemistry, 020956 Bucharest, Romania
| | | | - Fotios Tekos
- Department of Biochemistry-Biotechnology, University of Thessaly, 41500 Larissa, Greece
| | - Miriana Stan
- ‘Carol Davila’ University of Medicine and Pharmacy, Faculty of Pharmacy, Department of Toxicology, 020956 Bucharest, Romania
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Demetrios Kouretas
- Department of Biochemistry-Biotechnology, University of Thessaly, 41500 Larissa, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71409 Heraklion, Greece
| |
Collapse
|
34
|
He C, Liu S, Liang J, Zeng Y, Wang S, Wu Q, Xie W, Zhang Y. Genome-wide identification and analysis of nuclear receptors genes for lethal screening against Bemisia tabaci Q. PEST MANAGEMENT SCIENCE 2020; 76:2040-2048. [PMID: 31943718 DOI: 10.1002/ps.5738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/14/2019] [Accepted: 01/13/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Nuclear receptors (NRs) play an essential role in diverse biological processes, such as insect metamorphosis. Here, transcriptome analysis and functional studies were used to determine whether NRs are involved in metamorphosis of whitefly Bemisia tabaci Q, a serious pest to crops, and to find some potential insecticide targets. RESULTS Twenty NRs were identified in the Bemisia tabaci Q genome and categorized into the NR0-NR6 subfamilies. The phylogenetic tree of NRs from Bemisia tabaci Q and other representative species was constructed, which provided evolutionary insight into their genetic distances. The results of spatiotemporal gene expression indicated that the majority of NR gene expression was higher in the head than the abdomen and higher in eggs than adults. Further functional analysis using RNA interference (RNAi) showed that NR genes play an important role in Bemisia tabaci Q pupation and eclosion. With respect to high mortality and effects on growth, this was reflected in the unable to become pupa when the third-stage nymph treated with double-stranded RNA (dsRNA) and the developmental time delay (4-7 days) when pupae were treated with dsRNA for the 12 NR genes during molting compared with the development time in the control. CONCLUSION This study provides insight into NR functions during the metamorphosis stages of Bemisia tabaci Q. Several candidate genes could be potential insecticide targets for whitefly pest control due to their important roles in insect development. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chao He
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Shaonan Liu
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
- Institute of Insect Sciences, College of Agriculture, Yangtze University, Jingzhou, P. R. China
| | - Jinjin Liang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Yang Zeng
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Shaoli Wang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Qingjun Wu
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Wen Xie
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Youjun Zhang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| |
Collapse
|
35
|
Ding R, Liu S, He C, Nie X. Paracetamol affects the expression of detoxification- and reproduction-related genes and alters the life traits of Daphnia magna. ECOTOXICOLOGY (LONDON, ENGLAND) 2020; 29:398-406. [PMID: 32300985 DOI: 10.1007/s10646-020-02199-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/15/2020] [Indexed: 06/11/2023]
Abstract
Paracetamol (APAP) is a widely used non-steroidal anti-inflammatory drug and has been frequently detected in aquatic environment. However, limited information is provided about the toxic effects and detoxification mechanism of APAP in aquatic invertebrates. In the present study, the change of life traits of Daphnia magna (e.g., body length, growth rate and reproduction) was investigated under the chronic APAP exposure (0-5000 μg/L) for 21 day, and the effects of APAP on the expression of the detoxification- and reproduction-related genes including HR96, CYP360A8, CYP314, MRP4, P-gp, EcR and Vtg in the acute exposure (up to 96 h) were also determined. Results showed that the molting frequency, days to the first brood and days to the first egg production of D. magna were affected under the 50 μg/L concentration of APAP in the chronic exposure test. In the acute test, the transcriptional expression of HR96 was up-regulated under APAP exposure for 24 and 48 h. Similar performances were also observed in the expression of CYP360A8, CYP314, MRP4 and P-gp. However, with exposure time extended to 96 h, the induction of HR96 decreased or even reversed in some cases. It may indicate that the defense system in Daphnia is activated for a short time of exposure or becomes adaptive after longer term of exposure. APAP exposure also affected reproduction-related genes expression, which was related to the exposure time and concentration of APAP. In summary, APAP significantly affected the expression of genes associated with detoxification metabolism and altered some physiological parameters in D. magna.
Collapse
Affiliation(s)
- Rui Ding
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Sijia Liu
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Cuiping He
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
36
|
Heintz MM, McRee R, Kumar R, Baldwin WS. Gender differences in diet-induced steatotic disease in Cyp2b-null mice. PLoS One 2020; 15:e0229896. [PMID: 32155178 PMCID: PMC7064244 DOI: 10.1371/journal.pone.0229896] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/16/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease; however, progression to nonalcoholic steatohepatitis (NASH) is associated with most adverse outcomes. CYP2B metabolizes multiple xeno- and endobiotics, and male Cyp2b-null mice are diet-induced obese (DIO) with increased NAFLD. However, the DIO study was not performed long enough to assess progression to NASH. Therefore, to assess the role of Cyp2b in fatty liver disease progression from NAFLD to NASH, we treated wildtype (WT) and Cyp2b-null mice with a normal diet (ND) or choline-deficient, L-amino acid-defined high fat diet (CDAHFD) for 8 weeks and determined metabolic and molecular changes. CDAHFD-fed WT female mice gained more weight and had greater liver and white adipose tissue mass than their Cyp2b-null counterparts; males experienced diet-induced weight loss regardless of genotype. Serum biomarkers of liver injury increased in both CDAHFD-fed female and male mice; however CDAHFD-fed Cyp2b-null females exhibited significantly lower serum ALT, AST, and ASP concentrations compared to WT mice, indicating Cyp2b-null females were protected from liver injury. In both genders, hierarchical clustering of RNA-seq data demonstrates several gene ontologies responded differently in CDAHFD-fed Cyp2b-null mice compared to WT mice (lipid metabolism > fibrosis > inflammation). Oil Red O staining and direct triglycerides measurements confirmed that CDAHFD-fed Cyp2b-null females were protected from NAFLD. CDAHFD-fed Cyp2b-null mice showed equivocal changes in fibrosis with transcriptomic and serum markers suggesting less inflammation due to glucocorticoid-mediated repression of immune responses. In contrast to females, CDAHFD-fed Cyp2b-null males had higher triglyceride levels. Results indicate that female Cyp2b-null mice are protected from NAFLD while male Cyp2b-null mice are more susceptible to NAFLD, with few significant changes in NASH development. This study confirms that increased NAFLD development does not necessarily lead to progressive NASH. Furthermore, it indicates a role for Cyp2b in fatty liver disease that differs based on gender.
Collapse
Affiliation(s)
- Melissa M. Heintz
- Environmental Toxicology Program, Clemson University, Clemson, SC, United States of America
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - Rebecca McRee
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - Ramiya Kumar
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - William S. Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC, United States of America
- Biological Sciences, Clemson University, Clemson, SC, United States of America
- * E-mail:
| |
Collapse
|
37
|
Lammers LA, Achterbergh R, Mathôt RAA, Romijn JA. The effects of fasting on drug metabolism. Expert Opin Drug Metab Toxicol 2019; 16:79-85. [PMID: 31851534 DOI: 10.1080/17425255.2020.1706728] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: There is considerable variability in the rates and extent of drug metabolism between patients due to physiological, genetic, pharmacologic, environmental and nutritional factors such as fasting. This variability in drug metabolism may result in treatment failure or, conversely, in increased side effects or toxicity. Preclinical studies have shown that fasting alters drug metabolism by modulating the activity of drug metabolizing enzymes involved. However, until recently little was known about the effects of fasting on drug metabolism in humans.Areas covered: This review describes the effects of fasting on drug metabolism based on both preclinical studies and studies performed in humans.Expert opinion: A better understanding of the effects of fasting may improve the efficacy and safety of pharmacotherapy for individual patients. Fasting contributes to variability in human drug metabolism by differentially affecting drug metabolizing enzymes. Although the effects of fasting on drug metabolism appear to be small (between 10-20%), fasting may be relevant for drugs with a small therapeutic range and/or in combination with other factors that contribute to variability in drug metabolism such as physiological, genetic or pharmacological factors. Therefore, additional research on this topic is warranted.
Collapse
Affiliation(s)
- Laureen A Lammers
- Department of Hospital Pharmacy, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roos Achterbergh
- Department of Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ron A A Mathôt
- Department of Hospital Pharmacy, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Johannes A Romijn
- Department of Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
38
|
Rooney JP, Oshida K, Kumar R, Baldwin WS, Corton JC. Chemical Activation of the Constitutive Androstane Receptor Leads to Activation of Oxidant-Induced Nrf2. Toxicol Sci 2019; 167:172-189. [PMID: 30203046 DOI: 10.1093/toxsci/kfy231] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Exposure to environmentally relevant chemicals that activate the xenobiotic receptors aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), and peroxisome proliferator-activated receptor alpha (PPARα) in rodent test systems often leads to increases in oxidative stress (OS) that contributes to liver cancer induction. We hypothesized that activation of the oxidant-induced transcription factor Nrf2 could be used as a surrogate endpoint for increases in OS. We examined the relationships between activation of xenobiotic receptors and Nrf2 using previously characterized gene expression biomarkers that accurately predict modulation. Using a correlation approach (Running Fisher Test), the biomarkers were compared with microarray profiles in a mouse liver gene expression compendium. Out of the 163 chemicals examined, 47% from 53 studies activated Nrf2. We found consistent coupling between CAR and Nrf2 activation. Out of the 41 chemicals from 32 studies that activated CAR, 90% also activated Nrf2. CAR was activated earlier and at lower doses than Nrf2, indicating CAR activation preceded Nrf2 activation. Nrf2 activation by 2 CAR activators was abolished in CAR-null mice. We hypothesized that Nrf2 is activated by reactive oxygen species from the increased activity of enzymes encoded by Cyp2b family members. However, Nrf2 was similarly activated in the livers of both TCPOBOP-treated wild-type and Cyp2b9/10/13-null mice. This study provides evidence that Nrf2 activation (1) often occurs after exposure to xenobiotic chemicals, (2) is tightly linked to activation of CAR, and (3) does not require induction of 3 Cyp2b genes secondary to CAR activation.
Collapse
Affiliation(s)
- John P Rooney
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711.,Oak Ridge Institute for Science and Education (ORISE) participant at the National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Keiyu Oshida
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711.,Toray Industries, Inc, Kanagawa, Japan
| | - Ramiya Kumar
- Environmental Toxicology Program and Biological Sciences Department, Clemson University, Clemson, South Carolina 29634
| | - William S Baldwin
- Environmental Toxicology Program and Biological Sciences Department, Clemson University, Clemson, South Carolina 29634
| | - J Christopher Corton
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
39
|
Abe T, Shizu R, Sasaki T, Shimizu Y, Hosaka T, Kodama S, Matsuzawa A, Yoshinari K. Functional Interaction between Pregnane X Receptor and Yes-Associated Protein in Xenobiotic-Dependent Liver Hypertrophy and Drug Metabolism. J Pharmacol Exp Ther 2019; 371:590-601. [PMID: 31533970 DOI: 10.1124/jpet.119.258632] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/10/2019] [Indexed: 03/08/2025] Open
Abstract
Pregnane X receptor (PXR), a xenobiotic-responsive nuclear receptor, plays key roles in drug disposition. PXR activation induces liver hypertrophy in rodents, but the molecular mechanism of this effect remains unclear, although the PXR-mediated induction of cytochrome P450s (P450s) is proposed to be involved. Since yes-associated protein (YAP), an effector protein of the Hippo pathway, functions as a transcriptional cofactor that controls organ size via TEA domain family members (TEADs) or other transcription factors, we investigated the functional interaction of PXR with YAP in liver hypertrophy and drug metabolism in this study. The treatment of mice with a PXR activator induced liver hypertrophy, promoted nuclear YAP accumulation, and increased the expression of YAP/TEAD target genes in the liver, suggesting the coactivation of PXR and YAP. Through chronological analyses of this in vivo model, no clear association between PXR-dependent liver hypertrophy and P450 induction was observed. In reporter assays, ligand-activated PXR enhanced YAP-mediated gene transcription, whereas YAP overexpression inhibited PXR-dependent gene transcription. No clear species differences in these transcriptional interactions between humans and mice were observed. Furthermore, in human hepatocarcinoma and primary hepatocyte-like cells, YAP suppressed the expression of liver-enriched transcription factors, including hepatocyte nuclear factor 4α, PXR, the constitutive androstane receptor, and their target genes. These results suggest that YAP is involved in PXR-induced liver hypertrophy and that YAP activation interferes with gene expression associated with various liver functions. SIGNIFICANCE STATEMENT: We have investigated the functional interaction between PXR and YAP, an effector protein of the Hippo pathway. PXR plays central roles in various liver functions including drug metabolism, and the Hippo pathway and YAP regulate organ size through interacting with several transcription factors, including TEADs. Our results suggest that YAP is involved in PXR-mediated liver hypertrophy and that YAP activation interferes with the expression of liver-enriched transcription factors and thus drug-metabolizing enzymes.
Collapse
Affiliation(s)
- Taiki Abe
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Ryota Shizu
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Takamitsu Sasaki
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Yuki Shimizu
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Takuomi Hosaka
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Susumu Kodama
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Atsushi Matsuzawa
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| | - Kouichi Yoshinari
- Division of Drug Metabolism and Molecular Toxicology (T.A., S.K., K.Y.) and Laboratory of Health Chemistry (T.A., A.M., K.Y.), Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan; and Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Japan (T.A., R.S., T.S., Y.S., T.H., K.Y.)
| |
Collapse
|
40
|
Baldwin WS. Phase 0 of the Xenobiotic Response: Nuclear Receptors and Other Transcription Factors as a First Step in Protection from Xenobiotics. NUCLEAR RECEPTOR RESEARCH 2019; 6:101447. [PMID: 31815118 PMCID: PMC6897393 DOI: 10.32527/2019/101447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This mini-review examines the crucial importance of transcription factors as a first line of defense in the detoxication of xenobiotics. Key transcription factors that recognize xenobiotics or xenobiotic-induced stress such as reactive oxygen species (ROS), include AhR, PXR, CAR, MTF, Nrf2, NF-κB, and AP-1. These transcription factors constitute a significant portion of the pathways induced by toxicants as they regulate phase I-III detoxication enzymes and transporters as well as other protective proteins such as heat shock proteins, chaperones, and anti-oxidants. Because they are often the first line of defense and induce phase I-III metabolism, could these transcription factors be considered the phase 0 of xenobiotic response?
Collapse
Affiliation(s)
- William S Baldwin
- Clemson University, Biological Sciences/Environmental Toxicology, 132 Long Hall, Clemson, SC 29634
| |
Collapse
|
41
|
Abe T, Amaike Y, Shizu R, Takahashi M, Kano M, Hosaka T, Sasaki T, Kodama S, Matsuzawa A, Yoshinari K. Role of YAP Activation in Nuclear Receptor CAR-Mediated Proliferation of Mouse Hepatocytes. Toxicol Sci 2019; 165:408-419. [PMID: 29893953 DOI: 10.1093/toxsci/kfy149] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Constitutive androstane receptor (CAR) is a xenobiotic-responsive nuclear receptor that is highly expressed in the liver. CAR activation induces hepatocyte proliferation and hepatocarcinogenesis in rodents, but the mechanisms remain unclear. In this study, we investigated the association of CAR-dependent cell proliferation with Yes-associated protein (YAP), which is a transcriptional cofactor controlling organ size and cell growth through the interaction with various transcriptional factors including TEA domain family member (TEAD). In mouse livers, 1,4-bis-(2-[3,5-dichloropyridyloxy])benzene (TCPOBOP) (a mouse CAR [mCAR] activator) treatment increased the nuclear YAP accumulation and mRNA levels of YAP target genes as well as cell-cycle related genes along with liver hypertrophy and verteporfin (an inhibitor of YAP/TEAD interaction) cotreatment tended to attenuate them. Furthermore, in cell-based reporter gene assays, CAR activation enhanced the YAP/TEAD-dependent transcription. To investigate the role of YAP/TEAD activation in the CAR-dependent hepatocyte proliferation, we sought to establish an in vitro system completely reproducing CAR-dependent cell proliferation. Since CAR was only slightly expressed in cultured mouse primary hepatocytes compared with mouse livers and no proliferation was observed after treatment with TCPOBOP, we overexpressed CAR using mCAR expressing adenovirus (Ad-mCAR-V5) in mouse primary hepatocytes. Ad-mCAR-V5 infection and TCPOBOP treatment induced hepatocyte proliferation. Similar results were obtained with immortalized normal mouse hepatocytes as well. In the established in vitro system, CAR-dependent proliferation was strongly inhibited by Yap knockdown and completely abolished by verteporfin treatment. Our present results obtained in in vivo and in vitro experiments suggest that YAP/TEAD activation plays key roles in CAR-dependent proliferation of murine hepatocytes.
Collapse
Affiliation(s)
- Taiki Abe
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan.,Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Yuto Amaike
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ryota Shizu
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Miki Takahashi
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan.,Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Makoto Kano
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takamitsu Sasaki
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Susumu Kodama
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Kouichi Yoshinari
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan.,Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
42
|
Shmarakov IO, Lee YJ, Jiang H, Blaner WS. Constitutive androstane receptor mediates PCB-induced disruption of retinoid homeostasis. Toxicol Appl Pharmacol 2019; 381:114731. [PMID: 31449830 DOI: 10.1016/j.taap.2019.114731] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 11/18/2022]
Abstract
Environmental exposure to polychlorinated biphenyls (PCBs) is associated with an increased risk of incidence of metabolic disease, however the molecular mechanisms underlying this phenomenon are not fully understood. Our study provides new insights into molecular interactions between PCBs and retinoids (vitamin A and its metabolites) by defining a role for constitutive androstane receptor (CAR) in the disruption of retinoid homeostasis by non-coplanar 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153). Administration of four weekly 50 mg/kg doses of PCB153 to C57BL/6 male mice resulted in a significant decline in the tissue concentrations of retinyl esters, retinol and all-trans-retinoic acid (atRA), while no decline in hepatic and adipose tissue retinoid levels were detected in Car-null littermates. Our data imply that disrupted retinoid homeostasis occurs as a consequence of PCB153-induced activation of CAR, and raise the possibility that CAR signaling can affect atRA homeostasis in vivo. A strong correlation between the changes in retinoid metabolism and extensive upregulation of hepatic CAR-driven Cyp2b10 expression implicates this CYP isoform as contributing to retinoid homeostasis disruption via atRA oxidation during PCB153 exposure. In response to PCB153-induced CAR activation and disruption of retinoid homeostasis, expression of hepatic Pepck, Cd36 and adipose tissue Pparγ, Cd36, Adipoq, and Rbp4 were altered; however, this was reversed by administration of exogenous dietary retinoids (300 IU daily for 4 weeks). Our study establishes that PCB153 exposure enables a significant disruption of retinoid homeostasis in a CAR-dependent manner. We propose that this contributes to the obesogenic properties of PCB153 and may contribute to the predisposition to the metabolic disease.
Collapse
Affiliation(s)
- Igor O Shmarakov
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | - Yun Jee Lee
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Hongfeng Jiang
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - William S Blaner
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| |
Collapse
|
43
|
Barretto SA, Lasserre F, Fougerat A, Smith L, Fougeray T, Lukowicz C, Polizzi A, Smati S, Régnier M, Naylies C, Bétoulières C, Lippi Y, Guillou H, Loiseau N, Gamet-Payrastre L, Mselli-Lakhal L, Ellero-Simatos S. Gene Expression Profiling Reveals that PXR Activation Inhibits Hepatic PPARα Activity and Decreases FGF21 Secretion in Male C57Bl6/J Mice. Int J Mol Sci 2019; 20:ijms20153767. [PMID: 31374856 PMCID: PMC6696478 DOI: 10.3390/ijms20153767] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 01/25/2023] Open
Abstract
The pregnane X receptor (PXR) is the main nuclear receptor regulating the expression of xenobiotic-metabolizing enzymes and is highly expressed in the liver and intestine. Recent studies have highlighted its additional role in lipid homeostasis, however, the mechanisms of these regulations are not fully elucidated. We investigated the transcriptomic signature of PXR activation in the liver of adult wild-type vs. Pxr-/- C57Bl6/J male mice treated with the rodent specific ligand pregnenolone 16α-carbonitrile (PCN). PXR activation increased liver triglyceride accumulation and significantly regulated the expression of 1215 genes, mostly xenobiotic-metabolizing enzymes. Among the down-regulated genes, we identified a strong peroxisome proliferator-activated receptor α (PPARα) signature. Comparison of this signature with a list of fasting-induced PPARα target genes confirmed that PXR activation decreased the expression of more than 25 PPARα target genes, among which was the hepatokine fibroblast growth factor 21 (Fgf21). PXR activation abolished plasmatic levels of FGF21. We provide a comprehensive signature of PXR activation in the liver and identify new PXR target genes that might be involved in the steatogenic effect of PXR. Moreover, we show that PXR activation down-regulates hepatic PPARα activity and FGF21 circulation, which could participate in the pleiotropic role of PXR in energy homeostasis.
Collapse
Affiliation(s)
- Sharon Ann Barretto
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Frédéric Lasserre
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Anne Fougerat
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Lorraine Smith
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Tiffany Fougeray
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Céline Lukowicz
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Arnaud Polizzi
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Sarra Smati
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Marion Régnier
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Claire Naylies
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Colette Bétoulières
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Yannick Lippi
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Hervé Guillou
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Nicolas Loiseau
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Laurence Gamet-Payrastre
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Laila Mselli-Lakhal
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Sandrine Ellero-Simatos
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France.
| |
Collapse
|
44
|
Heintz MM, Kumar R, Rutledge MM, Baldwin WS. Cyp2b-null male mice are susceptible to diet-induced obesity and perturbations in lipid homeostasis. J Nutr Biochem 2019; 70:125-137. [PMID: 31202118 DOI: 10.1016/j.jnutbio.2019.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/26/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
Abstract
Obesity is an endemic problem in the United States and elsewhere, and data indicate that in addition to overconsumption, exposure to specific chemicals enhances obesity. CYP2B metabolizes multiple endo- and xenobiotics, and recent data suggests that repression of Cyp2b activity increases dyslipidemia and age-onset obesity, especially in males. To investigate the role played by Cyp2b in lipid homeostasis and obesity, we treated wildtype and Cyp2b-null mice with a normal (ND) or 60% high-fat diet (HFD) for 10 weeks and determined metabolic and molecular changes. Male HFD-fed Cyp2b-null mice weigh 15% more than HFD-fed wildtype mice, primarily due to an increase in white adipose tissue (WAT); however, Cyp2b-null female mice did not demonstrate greater body mass or WAT. Serum parameters indicate increased ketosis, leptin and cholesterol in HFD-fed Cyp2b-null male mice compared to HFD-fed wildtype mice. Liver triglycerides and liver:serum triglyceride ratios were higher than their similarly treated wildtype counterparts in Cyp2b-null male mice, indicating a role for Cyp2b in fatty acid metabolism regardless of diet. Furthermore, RNAseq demonstrates that hepatic gene expression in ND-fed Cyp2b-null male mice is similar to HFD-fed WT male mice, suggestive of fatty liver disease progression and a role for Cyp2b in lipid homeostasis. Females did not show as demonstrative changes in liver health, and significantly fewer changes in gene expression, as well as gene expression associated with liver disease. Overall our data indicates that the repression or inhibition of CYP2B may exacerbate metabolic disorders and cause obesity by perturbing fatty acid metabolism, especially in males.
Collapse
Affiliation(s)
- Melissa M Heintz
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634
| | - Ramiya Kumar
- Biological Sciences, Clemson University, Clemson, SC 29634
| | | | - William S Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634; Biological Sciences, Clemson University, Clemson, SC 29634.
| |
Collapse
|
45
|
Dempsey JL, Wang D, Siginir G, Fei Q, Raftery D, Gu H, Yue Cui J. Pharmacological Activation of PXR and CAR Downregulates Distinct Bile Acid-Metabolizing Intestinal Bacteria and Alters Bile Acid Homeostasis. Toxicol Sci 2019; 168:40-60. [PMID: 30407581 PMCID: PMC6821357 DOI: 10.1093/toxsci/kfy271] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome regulates important host metabolic pathways including xenobiotic metabolism and intermediary metabolism, such as the conversion of primary bile acids (BAs) into secondary BAs. The nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are well-known regulators for xenobiotic biotransformation in liver. However, little is known regarding the potential effects of PXR and CAR on the composition and function of the gut microbiome. To test our hypothesis that activation of PXR and CAR regulates gut microbiota and secondary BA synthesis, 9-week-old male conventional and germ-free mice were orally gavaged with corn oil, PXR agonist PCN (75 mg/kg), or CAR agonist TCPOBOP (3 mg/kg) once daily for 4 days. PCN and TCPOBOP decreased two taxa in the Bifidobacterium genus, which corresponded with decreased gene abundance of the BA-deconjugating enzyme bile salt hydrolase. In liver and small intestinal content of germ-free mice, there was a TCPOBOP-mediated increase in total, primary, and conjugated BAs corresponding with increased Cyp7a1 mRNA. Bifidobacterium, Dorea, Peptociccaceae, Anaeroplasma, and Ruminococcus positively correlated with T-UDCA in LIC, but negatively correlated with T-CDCA in serum. In conclusion, PXR and CAR activation downregulates BA-metabolizing bacteria in the intestine and modulates BA homeostasis in a gut microbiota-dependent manner.
Collapse
Affiliation(s)
- Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| | - Dongfang Wang
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
- Chongqing Blood Center, Chongqing 400015, P.R. China
| | - Gunseli Siginir
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| | - Qiang Fei
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
- Department of Chemistry, Jilin University, Changchun, Jilin Province 130061, P.R. China
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, Center for Metabolic and Vascular Biology, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Phoenix, Arizona 85004
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105
| |
Collapse
|
46
|
Liu C, Wang B, Zhou B, Jian X, Zhang X, Wang Y. The responses of Oncorhynchus mykiss coping with BDE-47 stress via PXR-mediated detoxification and Nrf2-mediated antioxidation system. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 207:63-71. [PMID: 30530205 DOI: 10.1016/j.aquatox.2018.11.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 06/09/2023]
Abstract
The low brominated polybrominated diphenyl ether (PBDE) 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) is ubiquitous in the marine environment. To elucidate the stress response and possible mechanisms underlying BDE-47, the rainbow trout fish Oncorhynchus mykiss were selected and orally fed bait with BDE-47 concentrations of 50 ng/g and 500 ng/g. BDE-47 was found to be mainly accumulated in head kidney and caused lipid peroxidation after prolonged exposure. We studied the detoxification system genes pregnane X receptor (PXR) and downstream genes (cytochrome 3 A, CYP3 A; glutathione S-transferase, GST) and their corresponding enzyme activity and found that the above indicators in the treatment groups increased first and then decreased with time, while the 500 ng/g group showed more significant changes. Further, the antioxidant system gene expression levels of the NF-E2-related factor 2 (Nrf2) and downstream genes (superoxide dismutase, SOD; catalase, CAT) were found significantly up-regulated with concentration and time. The change in the enzyme activity of SOD and CAT showed the same tendency as that of indicators of detoxifying system. The results showed that BDE-47 can accumulated in head kidney and caused activate and fast increase of genes and enzymes of detoxification and antioxidant system in the short-term and then damage the response systems in longer times. After Pearson correlation analysis, the Integrated Biomarker Response (IBR) Index was established with malondialdehyde (MDA) content; PXR, Nrf2, SOD, and CAT gene expression; and CYP3 A, GST, and CAT enzymatic activity, which were significantly related to BDE-47 bioaccumulation (P < 0.5). The IBR value can indicate the ecotoxicological responses of the head kidney to different BDE-47 concentrations exposure, but the high activity of the antioxidant system might obscure the damage of the detoxification system.
Collapse
Affiliation(s)
- Chunchen Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Boyuan Wang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Bin Zhou
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
| | - Xiaoyang Jian
- North China Sea Environmental Monitoring Center, State Oceanic Administration, Fushun Road 22, Qingdao, Shandong Province, 266033, China.
| | - Xinxin Zhang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - You Wang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
| |
Collapse
|
47
|
Tian J, Hu J, Liu G, Yin H, Chen M, Miao P, Bai P, Yin J. Altered Gene expression of ABC transporters, nuclear receptors and oxidative stress signaling in zebrafish embryos exposed to CdTe quantum dots. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 244:588-599. [PMID: 30384064 DOI: 10.1016/j.envpol.2018.10.092] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/11/2018] [Accepted: 10/18/2018] [Indexed: 06/08/2023]
Abstract
Adenosine triphosphate-binding cassette (ABC) transporters, including P-glycoprotein (Pgp) and multi-resistance associated proteins (Mrps), have been considered important participants in the self-protection of zebrafish embryos against environmental pollutants, but their possible involvement in the efflux and detoxification of quantum dots (QDs), as well as their regulation mechanism are currently unclear. In this work, gene expression alterations of ABC transporters, nuclear receptors, and oxidative stress signaling in zebrafish embryos after the treatment of mercaptopropionic acid (MPA)CdTe QDs and MPA-CdSCdTe QDs were investigated. It was observed that both QDs caused concentration-dependent delayed hatching effects and the subsequent induction of transporters like mrp1&2 in zebrafish embryos, indicating the protective role of corresponding proteins against CdTe QDs. Accompanying these alterations, expressions of nuclear receptors including the pregnane X receptor (pxr), aryl hydrocarbon receptor (ahr) 1b, and peroxisome proliferator-activated receptor (ppar)-β were induced by QDs in a concentration- and time-dependent manner. Moreover, elevated oxidative stress, reflected by the reduction of glutathione (GSH) level and superoxide dismutase (SOD) activities, as well as the dramatic induction of nuclear factor E2 related factor (nrf) 2, was also found. More importantly, alterations of pxr and nrf2 were more pronounced than that of mrps, and these receptors exhibited an excellent correlation with delayed hatching rate in the same embryos (R2 > 0.8). Results from this analysis demonstrated that the induction of mrp1 and mrp2 could be important components for the detoxification of QDs in zebrafish embryos. These transporters could be modulated by nuclear receptors and oxidative stress signaling. In addition, up-regulation of pxr and nrf2 could be developed as toxic biomarkers of CdTe QDs.
Collapse
Affiliation(s)
- Jingjing Tian
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Academy for Engineering & Technology, Fudan University, Shanghai 200433, PR China
| | - Jia Hu
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Guangxing Liu
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China
| | - Huancai Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China
| | - Mingli Chen
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China
| | - Peng Miao
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China
| | - Pengli Bai
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China
| | - Jian Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China.
| |
Collapse
|
48
|
Farmahin R, Gannon AM, Gagné R, Rowan-Carroll A, Kuo B, Williams A, Curran I, Yauk CL. Hepatic transcriptional dose-response analysis of male and female Fischer rats exposed to hexabromocyclododecane. Food Chem Toxicol 2018; 133:110262. [PMID: 30594549 DOI: 10.1016/j.fct.2018.12.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
Hexabromocyclododecane (HBCD) is a brominated flame retardant found in the environment and human tissues. The toxicological effects of HBCD exposure are not clearly understood. We employed whole-genome RNA-sequencing on liver samples from male and female Fischer rats exposed to 0, 250, 1250, and 5000 mg technical mixture of HBCD/kg diet for 28 days to gain further insight into HBCD toxicity. HBCD altered 428 and 250 gene transcripts in males and females, respectively, which were involved in metabolism of xenobiotics, oxidative stress, immune response, metabolism of glucose and lipids, circadian regulation, cell cycle, fibrotic activity, and hormonal balance. Signature analysis supported that HBCD operates through the constitutive androstane and pregnane X receptors. The median transcriptomic benchmark dose (BMD) for the lowest statistically significant pathway was within 1.5-fold of the BMD for increased liver weight, while the BMD for the lowest pathway with at least three modeled genes (minimum 5% of pathway) was similar to the lowest apical endpoint BMD. The results show how transcriptional analyses can inform mechanisms underlying chemical toxicity and the doses at which potentially adverse effects occur. This experiment is part of a larger study exploring the use of toxicogenomics and high-throughput screening for human health risk assessment.
Collapse
Affiliation(s)
- Reza Farmahin
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Anne Marie Gannon
- Regulatory Toxicology Research Division, Health Products and Food Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Rémi Gagné
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Andrea Rowan-Carroll
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Byron Kuo
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Ivan Curran
- Regulatory Toxicology Research Division, Health Products and Food Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| |
Collapse
|
49
|
Damiri B, Baldwin WS. Cyp2b-Knockdown Mice Poorly Metabolize Corn Oil and Are Age-Dependent Obese. Lipids 2018; 53:871-884. [PMID: 30421529 DOI: 10.1002/lipd.12095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023]
Abstract
We previously made a RNAi-based cytochrome P450 2b (Cyp2b)-knockdown (Cyp2b-KD) mouse to determine the in vivo role of the Cyp2b subfamily in xenobiotic detoxification. Further studies reported here indicate a role for Cyp2b in unsaturated fatty-acid (UFA) metabolism and in turn obesity. Mice were treated intraperitoneally (i.p.) with 100 μL corn oil as a carrier or the potent Cyp2b-inducer 3,3',5,5'-Tetrachloro-1,4-bis(pyridyloxy)benzene (TCPOBOP (TC)) dissolved in corn oil. Surprisingly, female Cyp2b-KD mice but not male mice showed increased liver lipid accumulation. Male Cyp2b-KD mice had higher serum triacylglycerols, cholesterol, very low-density lipoprotein (VLDL), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) than wildtype (WT) mice; females had higher cholesterol, LDL, and HDL. Thus, Cyp2b-KD mice are unable to clear a high bolus dose of corn oil, potentially because the Cyp2b-KD mice were unable to metabolize the UFA in the corn oil. Therefore, WT and Cyp2b-KD mice were housed for 35 weeks and necropsies performed to test whether Cyp2b-KD mice develop age onset obesity. Cyp2b-KD mice exhibited a significant increase in body weight caused by an increase in white adipose tissue deposition relative to WT mice. Serum cholesterol, triacylglycerol, LDL, and VLDL were significantly greater in 35-week-old Cyp2b-KD males compared to WT males; only serum triacylglycerol and LDL were higher in females. In conclusion, changes in Cyp2b expression led to perturbation in lipid metabolism and depuration in Cyp2b-KD mice. This suggests that Cyp2b is more than a detoxification enzyme, but also involved in the metabolism of UFA, as Cyp2b-KD mice have increased the body weight, fat deposition, and serum lipids.
Collapse
Affiliation(s)
- Basma Damiri
- Medicine and Health Sciences Faculty, Drugs and Toxicology Division, An-Najah National University, Omar Ibn Al-Khattab St., PO Box 7, Nablus, West Bank, Palestinian Territories
| | - William S Baldwin
- Biological Sciences, Clemson University, 132 Long Hall St., Clemson, SC 29634, USA.,Environmental Toxicology Program, 132 Long Hall St., Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
50
|
Braeuning A, Kollotzek F, Zeller E, Knorpp T, Templin MF, Schwarz M. Mouse Hepatomas with Ha-ras and B-raf Mutations Differ in Mitogen-Activated Protein Kinase Signaling and Response to Constitutive Androstane Receptor Activation. Drug Metab Dispos 2018; 46:1462-1465. [PMID: 30115646 DOI: 10.1124/dmd.118.083014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/14/2018] [Indexed: 12/31/2022] Open
Abstract
Nuclear receptors mediate the hepatic induction of drug-metabolizing enzymes by xenobiotics. Not much is known about enzyme induction in liver tumors. Here, we treated tumor-bearing mice with phenobarbital, an activator of the constitutive androstane receptor (CAR), to analyze the response of chemically induced Ha-ras- and B-raf-mutated mouse liver adenoma to CAR activation in vivo. Both tumor subpopulations possess almost identical gene expression profiles. CAR target gene induction in the tumors was studied at the mRNA and protein levels, and a reverse-phase protein microarray approach was chosen to characterize important signaling cascades. CAR target gene induction was pronounced in B-raf-mutated but not in Ha-ras-mutated tumors. Phosphoproteomic profiling revealed that phosphorylation-activated extracellular signal-regulated kinase (ERK) 1/2 was more abundant in Ha-ras-mutated than in B-raf-mutated tumors. ERK activation in tumor tissue was negatively correlated with CAR target induction. ERK activation is known to inhibit CAR-dependent transcription. In summary, profound differences exist between the two closely related tumor subpopulations with respect to the activation of mitogenic signaling cascades, and these dissimilarities might explain the differences in xenobiotic induction of CAR target genes.
Collapse
Affiliation(s)
- Albert Braeuning
- Department of Toxicology, University of Tübingen, Tübingen, Germany (A.B., F.K., E.Z., M.S.); Natural and Medical Sciences Institute, Reutlingen, Germany (T.K., M.F.T.); and Department Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany (A.B.)
| | - Ferdinand Kollotzek
- Department of Toxicology, University of Tübingen, Tübingen, Germany (A.B., F.K., E.Z., M.S.); Natural and Medical Sciences Institute, Reutlingen, Germany (T.K., M.F.T.); and Department Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany (A.B.)
| | - Eva Zeller
- Department of Toxicology, University of Tübingen, Tübingen, Germany (A.B., F.K., E.Z., M.S.); Natural and Medical Sciences Institute, Reutlingen, Germany (T.K., M.F.T.); and Department Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany (A.B.)
| | - Thomas Knorpp
- Department of Toxicology, University of Tübingen, Tübingen, Germany (A.B., F.K., E.Z., M.S.); Natural and Medical Sciences Institute, Reutlingen, Germany (T.K., M.F.T.); and Department Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany (A.B.)
| | - Markus F Templin
- Department of Toxicology, University of Tübingen, Tübingen, Germany (A.B., F.K., E.Z., M.S.); Natural and Medical Sciences Institute, Reutlingen, Germany (T.K., M.F.T.); and Department Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany (A.B.)
| | - Michael Schwarz
- Department of Toxicology, University of Tübingen, Tübingen, Germany (A.B., F.K., E.Z., M.S.); Natural and Medical Sciences Institute, Reutlingen, Germany (T.K., M.F.T.); and Department Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany (A.B.)
| |
Collapse
|