1
|
Deans JR, Deol P, Titova N, Radi SH, Vuong LM, Evans JR, Pan S, Fahrmann J, Yang J, Hammock BD, Fiehn O, Fekry B, Eckel-Mahan K, Sladek FM. HNF4α isoforms regulate the circadian balance between carbohydrate and lipid metabolism in the liver. Front Endocrinol (Lausanne) 2023; 14:1266527. [PMID: 38111711 PMCID: PMC10726135 DOI: 10.3389/fendo.2023.1266527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/06/2023] [Indexed: 12/20/2023] Open
Abstract
Hepatocyte Nuclear Factor 4α (HNF4α), a master regulator of hepatocyte differentiation, is regulated by two promoters (P1 and P2) which drive the expression of different isoforms. P1-HNF4α is the major isoform in the adult liver while P2-HNF4α is thought to be expressed only in fetal liver and liver cancer. Here, we show that P2-HNF4α is indeed expressed in the normal adult liver at Zeitgeber time (ZT)9 and ZT21. Using exon swap mice that express only P2-HNF4α we show that this isoform orchestrates a distinct transcriptome and metabolome via unique chromatin and protein-protein interactions, including with different clock proteins at different times of the day leading to subtle differences in circadian gene regulation. Furthermore, deletion of the Clock gene alters the circadian oscillation of P2- (but not P1-)HNF4α RNA, revealing a complex feedback loop between the HNF4α isoforms and the hepatic clock. Finally, we demonstrate that while P1-HNF4α drives gluconeogenesis, P2-HNF4α drives ketogenesis and is required for elevated levels of ketone bodies in female mice. Taken together, we propose that the highly conserved two-promoter structure of the Hnf4a gene is an evolutionarily conserved mechanism to maintain the balance between gluconeogenesis and ketogenesis in the liver in a circadian fashion.
Collapse
Affiliation(s)
- Jonathan R. Deans
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
- Genetics, Genomics and Bioinformatics Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Poonamjot Deol
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Nina Titova
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Sarah H. Radi
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
- Biochemistry and Molecular Biology Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Linh M. Vuong
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Jane R. Evans
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Songqin Pan
- Proteomics Core, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, United States
| | - Johannes Fahrmann
- National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis, CA, United States
| | - Jun Yang
- Department of Entomology and Nematology & UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Bruce D. Hammock
- Department of Entomology and Nematology & UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Oliver Fiehn
- National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis, CA, United States
| | - Baharan Fekry
- Department of Biochemistry and Molecular Biology, McGovern Medical School at the University of Texas Health Science Center (UT Health), Houston, TX, United States
| | - Kristin Eckel-Mahan
- Department of Biochemistry and Molecular Biology, McGovern Medical School at the University of Texas Health Science Center (UT Health), Houston, TX, United States
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center (UT Health), Houston, TX, United States
| | - Frances M. Sladek
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
2
|
Pouyabahar D, Chung SW, Pezzutti OI, Perciani CT, Wang X, Ma XZ, Jiang C, Camat D, Chung T, Sekhon M, Manuel J, Chen XC, McGilvray ID, MacParland SA, Bader GD. A rat liver cell atlas reveals intrahepatic myeloid heterogeneity. iScience 2023; 26:108213. [PMID: 38026201 PMCID: PMC10651689 DOI: 10.1016/j.isci.2023.108213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 08/20/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
The large size and vascular accessibility of the laboratory rat (Rattus norvegicus) make it an ideal hepatic animal model for diseases that require surgical manipulation. Often, the disease susceptibility and outcomes of inflammatory pathologies vary significantly between strains. This study uses single-cell transcriptomics to better understand the complex cellular network of the rat liver, as well as to unravel the cellular and molecular sources of inter-strain hepatic variation. We generated single-cell and single-nucleus transcriptomic maps of the livers of healthy Dark Agouti and Lewis rat strains and developed a factor analysis-based bioinformatics analysis pipeline to study data covariates, such as strain and batch. Using this approach, we discovered transcriptomic variation within the hepatocyte and myeloid populations that underlie distinct cell states between rat strains. This finding will help provide a reference for future investigations on strain-dependent outcomes of surgical experiment models.
Collapse
Affiliation(s)
- Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Sai W. Chung
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Olivia I. Pezzutti
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Catia T. Perciani
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xinle Wang
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xue-Zhong Ma
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Chao Jiang
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Damra Camat
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Trevor Chung
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Manmeet Sekhon
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Justin Manuel
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xu-Chun Chen
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Ian D. McGilvray
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Sonya A. MacParland
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D. Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
- Princess Margaret Research Institute, University Health Network, Toronto, ON, Canada
- The Multiscale Human Program, Canadian Institute for Advanced Research, Toronto, ON, Canada
| |
Collapse
|
3
|
Martinez-Calle M, Courbon G, Hunt-Tobey B, Francis C, Spindler J, Wang X, dos Reis LM, Martins CS, Salusky IB, Malluche H, Nickolas TL, Moyses RM, Martin A, David V. Transcription factor HNF4α2 promotes osteogenesis and prevents bone abnormalities in mice with renal osteodystrophy. J Clin Invest 2023; 133:e159928. [PMID: 37079387 PMCID: PMC10231994 DOI: 10.1172/jci159928] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/17/2023] [Indexed: 04/21/2023] Open
Abstract
Renal osteodystrophy (ROD) is a disorder of bone metabolism that affects virtually all patients with chronic kidney disease (CKD) and is associated with adverse clinical outcomes including fractures, cardiovascular events, and death. In this study, we showed that hepatocyte nuclear factor 4α (HNF4α), a transcription factor mostly expressed in the liver, is also expressed in bone, and that osseous HNF4α expression was dramatically reduced in patients and mice with ROD. Osteoblast-specific deletion of Hnf4α resulted in impaired osteogenesis in cells and mice. Using multi-omics analyses of bones and cells lacking or overexpressing Hnf4α1 and Hnf4α2, we showed that HNF4α2 is the main osseous Hnf4α isoform that regulates osteogenesis, cell metabolism, and cell death. As a result, osteoblast-specific overexpression of Hnf4α2 prevented bone loss in mice with CKD. Our results showed that HNF4α2 is a transcriptional regulator of osteogenesis, implicated in the development of ROD.
Collapse
Affiliation(s)
- Marta Martinez-Calle
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Guillaume Courbon
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bridget Hunt-Tobey
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Connor Francis
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jadeah Spindler
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Xueyan Wang
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luciene M. dos Reis
- LIM 16, Nephrology Department, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Carolina S.W. Martins
- LIM 16, Nephrology Department, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Isidro B. Salusky
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Hartmut Malluche
- Division of Nephrology, Bone and Mineral Metabolism, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Thomas L. Nickolas
- Department of Medicine, Columbia Irving University Medical Center, New York, New York, USA
| | - Rosa M.A. Moyses
- LIM 16, Nephrology Department, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Aline Martin
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Valentin David
- Division of Nephrology and Hypertension, Department of Medicine, and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
4
|
Noh SG, Jung HJ, Kim S, Arulkumar R, Kim DH, Park D, Chung HY. Regulation of Circadian Genes Nr1d1 and Nr1d2 in Sex-Different Manners during Liver Aging. Int J Mol Sci 2022; 23:ijms231710032. [PMID: 36077427 PMCID: PMC9456386 DOI: 10.3390/ijms231710032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Circadian rhythm is associated with the aging process and sex differences; however, how age and sex can change circadian regulation systems remains unclear. Thus, we aimed to evaluate age- and sex-related changes in gene expression and identify sex-specific target molecules that can regulate aging. Methods: Rat livers were categorized into four groups, namely, young male, old male, young female, and old female, and the expression of several genes involved in the regulation of the circadian rhythm was confirmed by in silico and in vitro studies. Results: Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses showed that the expression of genes related to circadian rhythms changed more in males than in females during liver aging. In addition, differentially expressed gene analysis and quantitative real-time polymerase chain reaction/western blotting analysis revealed that Nr1d1 and Nr1d2 expression was upregulated in males during liver aging. Furthermore, the expression of other circadian genes, such as Arntl, Clock, Cry1/2, Per1/2, and Rora/c, decreased in males during liver aging; however, these genes showed various gene expression patterns in females during liver aging. Conclusions: Age-related elevation of Nr1d1/2 downregulates the expression of other circadian genes in males, but not females, during liver aging. Consequently, age-related upregulation of Nr1d1/2 may play a more crucial role in the change in circadian rhythms in males than in females during liver aging.
Collapse
Affiliation(s)
- Sang Gyun Noh
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Hee Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Seungwoo Kim
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Radha Arulkumar
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Daeui Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141, Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea
| | - Hae Young Chung
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
- Correspondence: ; Tel.: +82-51-510-2814
| |
Collapse
|
5
|
Kang S, Bo Y, Yang D, Wu G, Yang X, Wei J, Zhao G, An M, Zhao L. Tandem mass tag-based proteomics analysis reveals the effects of Guri Gumu-13 pill on drug-induced liver injury. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1206:123353. [DOI: 10.1016/j.jchromb.2022.123353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/07/2022] [Accepted: 06/24/2022] [Indexed: 10/25/2022]
|
6
|
Marinho AT, Miranda JP, Caixas U, Charneira C, Gonçalves-Dias C, Marques MM, Monteiro EC, Antunes AMM, Pereira SA. Singularities of nevirapine metabolism: from sex-dependent differences to idiosyncratic toxicity. Drug Metab Rev 2019; 51:76-90. [PMID: 30712401 DOI: 10.1080/03602532.2019.1577891] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nevirapine (NVP) is a first-generation non-nucleoside reverse transcriptase inhibitor widely used for the treatment and prophylaxis of human immunodeficiency virus infection. The drug is taken throughout the patient's life and, due to the availability of an extended-release formulation, it is administered once daily. This antiretroviral is one of the scarce examples of drugs with prescription criteria based on sex, in order to prevent adverse reactions. The therapy with NVP has been associated with potentially life-threatening liver and idiosyncratic skin toxicity. Multiple evidence has emerged regarding the formation of electrophilic NVP metabolites as crucial for adverse idiosyncratic reactions. The formation of reactive metabolites that yield covalent adducts with proteins has been demonstrated in patients under NVP-based treatment. Interestingly, several pharmacogenetic- and sex-related factors associated with NVP toxicity can be mechanistically explained by an imbalance toward increased formation of NVP-derived reactive metabolites and/or impaired detoxification capability. Moreover, the haptenation of self-proteins by these reactive species provides a plausible link between NVP bioactivation and immunotoxicity, further supporting the relevance of this toxicokinetics hypothesis. In the current paper, we review the existing knowledge and recent developments on NVP metabolism and their relation to NVP toxicity.
Collapse
Affiliation(s)
- Aline T Marinho
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Joana P Miranda
- b Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy , Universidade de Lisboa , Lisboa , Portugal
| | - Umbelina Caixas
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal.,c Centro Hospitalar de Lisboa Central (CHLC) , Lisboa , Portugal
| | - Catarina Charneira
- d Centro de Química Estrutural (CQE) , Instituto Superior Técnico, ULisboa , Lisboa , Portugal
| | - Clara Gonçalves-Dias
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - M Matilde Marques
- d Centro de Química Estrutural (CQE) , Instituto Superior Técnico, ULisboa , Lisboa , Portugal
| | - Emília C Monteiro
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Alexandra M M Antunes
- d Centro de Química Estrutural (CQE) , Instituto Superior Técnico, ULisboa , Lisboa , Portugal
| | - Sofia A Pereira
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| |
Collapse
|
7
|
Mizukami H, Kim JD, Tabara S, Lu W, Kwon C, Nakashima M, Fukamizu A. KDM5D-mediated H3K4 demethylation is required for sexually dimorphic gene expression in mouse embryonic fibroblasts. J Biochem 2019; 165:335-342. [PMID: 30541083 DOI: 10.1093/jb/mvy106] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/10/2018] [Indexed: 11/14/2022] Open
Abstract
Males and females share the same genetic code, but gene expression profile often displays differences between two sexes. Mouse embryonic fibroblasts (MEFs) have been used to experiment as a useful tool to test gene function. They have also been characterized by gender-based differences in expressed genes such as Y-linked Sry or X-linked Hprt. However, there is no report on sex differences in global gene expression. Here, using the next-generation RNA sequencing, we compared the comprehensive transcriptome of MEFs derived from two sexes. In comparison with the female group, the male group up-regulated 27 differentially expressed genes (DEGs), in which a male-specific histone demethylase KDM5D gene is included, and 7 DEGs were down-regulated. Based on the results by searching the ENCODE analysis, it was shown that the expression of 15 genes identified is potentially regulated by the methylation of H3K4me1 or H3K4me3. Interestingly, we demonstrated that both of H3K4 methylation are induced by knocking down KDM5D, which causes changes in patterns of eight DEGs found in male MEFs. Collectively, these data not only suggest an importance of KDM5D-mediated demethylation of H3K4 involved in the sexually dimorphic gene expression in male MEFs, but also may provide information regarding sex-dependent changes in gene expression when MEFs are used for experiments.
Collapse
Affiliation(s)
- Hayase Mizukami
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA).,Graduate School of Life and Environmental Sciences
| | - Jun-Dal Kim
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA)
| | | | - Weizhe Lu
- Ph.D. Program in Human Biology, School of Integrative Global Majors (SIGMA)
| | - Chulwon Kwon
- Graduate School of Life and Environmental Sciences
| | | | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA).,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| |
Collapse
|
8
|
Capri KM, Maroni MJ, Deane HV, Pierre A, Adams AM, Goncalves FL, Meyer AS, Seggio JA. Effects of time of day and constant light on the behavioral responses and ethanol metabolism to acute alcohol administration in male Black Swiss mice. BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2018.1543640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Kimberly M. Capri
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Marissa J. Maroni
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Hannah V. Deane
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Audeline Pierre
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Abigail M. Adams
- Department of Mathematics, Bridgewater State University, Bridgewater, MA, USA
| | - Fatiana L. Goncalves
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Andrew S. Meyer
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Joseph A. Seggio
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| |
Collapse
|
9
|
Fujita N, Takenaka A. Testosterone represses urinary excretion of the alpha-tocopherol metabolite alpha-carboxymethylhydroxychroman in rats. J Nutr Biochem 2018; 62:59-64. [PMID: 30253278 DOI: 10.1016/j.jnutbio.2018.08.006] [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: 05/22/2018] [Revised: 07/06/2018] [Accepted: 08/16/2018] [Indexed: 12/23/2022]
Abstract
In rats, plasma and tissue concentrations of α-tocopherol, a predominant form of vitamin E in mammals, are known to differ between the sexes. In order to examine sex differences in α-tocopherol metabolism, we investigated urinary excretion of the α-tocopherol metabolite α-carboxymethylhydroxychroman (α-CEHC) using Wistar rats. First, we measured α-CEHC in urine of 9-week-old male and female rats in basal and α-tocopherol-administered conditions. We observed that female rats excrete significantly more α-CEHC than male rats via urine. This sex difference was observed in matured 9-week-old rats but not in premature 3-week-old rats, suggesting that the difference may relate to sex hormones. In order to confirm this, we examined the effect of ovariectomy and orchiectomy on female and male rats, respectively. The results of castration clearly demonstrated that orchiectomy enhanced urinary excretion of α-CEHC, supporting the hypothesis that testosterone repressed α-tocopherol metabolism. We then administered testosterone propionate to orchiectomized rats and observed down-regulation of α-CEHC excretion. Taken together, these results indicate that testosterone represses the metabolism and urinary excretion of α-tocopherol in rats. This is the first report to show a sex-dependent difference in urinary excretion rate of an α-tocopherol metabolite and contributes to the understanding of vitamin E metabolism.
Collapse
Affiliation(s)
- Naoko Fujita
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa 214-8571, Japan
| | - Asako Takenaka
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa 214-8571, Japan.
| |
Collapse
|
10
|
Gα i3 signaling is associated with sexual dimorphic expression of the clock-controlled output gene Dbp in murine liver. Oncotarget 2018; 9:30213-30224. [PMID: 30100984 PMCID: PMC6084400 DOI: 10.18632/oncotarget.25727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/14/2018] [Indexed: 11/25/2022] Open
Abstract
The albumin D-box binding protein (DBP) is a member of the PAR bZip (proline and acidic amino acid-rich basic leucine zipper) transcription factor family and functions as important regulator of circadian core and output gene expression. Gene expression of DBP itself is under the control of E-box-dependent binding by the Bmal1-Clock heterodimer and CRE-dependent binding by the cAMP responsive element binding protein (CREB). However, the signaling mechanism mediating CREB-dependent regulation of DBP expression in the peripheral clock remains elusive. In this study, we examined the role of the GPCR (G-protein-coupled receptor)/Gαi3 (Galphai3) controlled cAMP-CREB signaling pathway in the regulation of hepatic expression of core clock and clock-regulated genes, including Dbp. Analysis of circadian gene expression revealed that rhythmicity of hepatic transcript levels of the majority of core clock (including Per1) and clock-regulated genes were not affected by Gαi3 deficiency. Consistently, the period length of primary Gαi3 deficient tail fibroblasts expressing a Bmal1-Luciferase reporter was not affected. Interestingly, however, Gαi3 deficient female but not male mice showed a tendentiously increased activation of CREB (nuclear pSer133-CREB) accompanied by an advanced peak in Dbp gene expression and elevated mRNA levels of the cytochrome P450 family member Cyp3a11, a target gene of DBP. Accordingly, selective inhibition of CREB led to a strongly decreased expression of DBP and CYP3A4 (human Cyp3a11 homologue) in HepG2 liver cells. In summary, our data suggest that the Gαi3-pCREB signalling pathway functions as a regulator of sexual-dimorphic expression of DBP and its xenobiotic target enzymes Cyp3a11/CYP3A4.
Collapse
|
11
|
Ozturk N, Ozturk D, Pala-Kara Z, Kaptan E, Sancar-Bas S, Ozsoy N, Cinar S, Deniz G, Li XM, Giacchetti S, Lévi F, Okyar A. The immune system as a chronotoxicity target of the anticancer mTOR inhibitor everolimus. Chronobiol Int 2018; 35:705-718. [PMID: 29400578 DOI: 10.1080/07420528.2018.1432632] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The circadian timing system controls many biological functions in mammals including xenobiotic metabolism, detoxification, cell proliferation, apoptosis and immune functions. Everolimus is a mammalian target of rapamycin inhibitor, whose immunosuppressant properties are both desired in transplant patients and unwanted in cancer patients, where it is indicated for its antiproliferative efficacy. Here we sought whether everolimus circadian timing would predictably modify its immunosuppressive effects so as to optimize this drug through timing. C57BL/6J mice were synchronized with light-dark 12h:12h, with L onset at Zeitgeber Time (ZT) 0. Everolimus was administered orally to male (5 mg/kg/day) and female mice (15 mg/kg/day) at ZT1, during early rest span or at ZT13, during early activity span for 4 weeks. Body weight loss, as well as hematological, immunological and biochemical toxicities, were determined. Spleen and thymus were examined histologically. Everolimus toxicity was less severe following dosing at ZT13, as compared to ZT1, as shown with least body weight inhibition in both genders; least reductions in thymus weight both in males (p < 0.01) and females (p < 0.001), least reduction in female spleen weight (p < 0.05), and less severe thymic medullar atrophy both in males (p < 0.001) and females (p < 0.001). The mean circulating counts in total leukocytes, total lymphocytes, T-helper and B lymphocytes displayed minor and non-significant changes following dosing at ZT13, while they were decreased by 56.9% (p < 0.01), 45.5% (p < 0.01), 43.1% (p < 0.05) and 48.7% (p < 0.01) after everolimus at ZT1, respectively, in only male mice. Chronotherapy of everolimus is an effective way to increase the general tolerability and decrease toxicity on the immune system.
Collapse
Affiliation(s)
- Narin Ozturk
- a Department of Pharmacology, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| | - Dilek Ozturk
- b Department of Pharmacology, Faculty of Pharmacy , Bezmialem Vakif University , Fatih-İstanbul , Turkey
| | - Zeliha Pala-Kara
- a Department of Pharmacology, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| | - Engin Kaptan
- c Department of Biology, Faculty of Science , Istanbul University , Vezneciler-Istanbul , Turkey
| | - Serap Sancar-Bas
- c Department of Biology, Faculty of Science , Istanbul University , Vezneciler-Istanbul , Turkey
| | - Nurten Ozsoy
- d Department of Biochemistry, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| | - Suzan Cinar
- e Department of Immunology, Aziz Sancar Institute of Experimental Medicine , Istanbul University , Sehremini- Istanbul , Turkey
| | - Gunnur Deniz
- e Department of Immunology, Aziz Sancar Institute of Experimental Medicine , Istanbul University , Sehremini- Istanbul , Turkey
| | - Xiao-Mei Li
- f INSERM UMRS 935 "Modèles de cellules souches malignes et thérapeutiques" , Campus CNRS , Villejuif-Cedex , France
| | - Sylvie Giacchetti
- g AP-HP, Hôpital Saint-Louis, Breast Disease Unit , University Paris Diderot , Paris , France
| | - Francis Lévi
- f INSERM UMRS 935 "Modèles de cellules souches malignes et thérapeutiques" , Campus CNRS , Villejuif-Cedex , France.,h Warwick Systems Biology Centre, Warwick Medical School , Cancer Chronotherapy Unit , Coventry , UK
| | - Alper Okyar
- a Department of Pharmacology, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| |
Collapse
|
12
|
Sexual Dimorphism in the Response of Mercurialis annua to Stress. Metabolites 2016; 6:metabo6020013. [PMID: 27128954 PMCID: PMC4931544 DOI: 10.3390/metabo6020013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/18/2016] [Accepted: 04/21/2016] [Indexed: 11/17/2022] Open
Abstract
The research presented stemmed from the observations that female plants of the annual dioecious Mercurialis annua outlive male plants. This led to the hypothesis that female plants of M. annua would be more tolerant to stress than male plants. This hypothesis was addressed in a comprehensive way, by comparing morphological, biochemical and metabolomics changes in female and male plants during their development and under salinity. There were practically no differences between the genders in vegetative development and physiological parameters. However, under salinity conditions, female plants produced significantly more new reproductive nodes. Gender-linked differences in peroxidase (POD) and glutathione transferases (GSTs) were involved in anti-oxidation, detoxification and developmental processes in M. annua. 1H NMR metabolite profiling of female and male M. annua plants showed that under salinity the activity of the TCA cycle increased. There was also an increase in betaine in both genders, which may be explainable by its osmo-compatible function under salinity. The concentration of ten metabolites changed in both genders, while ‘Female-only-response’ to salinity was detected for five metabolites. In conclusion, dimorphic responses of M. annua plant genders to stress may be attributed to female plants’ capacity to survive and complete the reproductive life cycle.
Collapse
|
13
|
Zhang F, Yu X, He C, Ouyang X, Wu J, Li J, Zhang J, Duan X, Wan Y, Yue J. Effects of sexually dimorphic growth hormone secretory patterns on arachidonic acid metabolizing enzymes in rodent heart. Toxicol Appl Pharmacol 2015; 289:495-506. [PMID: 26493931 DOI: 10.1016/j.taap.2015.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 11/26/2022]
Abstract
The arachidonic acid (AA) metabolizing enzymes are the potential therapeutic targets of cardiovascular diseases (CVDs). As sex differences have been shown in the risk and outcome of CVDs, we investigated the regulation of heart AA metabolizing enzymes (COXs, LOXs, and CYPs) by sex-dependent growth hormone (GH) secretory patterns. The pulsatile (masculine) GH secretion at a physiological concentration decreased CYP1A1 and CYP2J3 mRNA levels more efficiently in the H9c2 cells compared with the constant (feminine) GH secretion; however, CYP1B1 mRNA levels were higher following the pulsatile GH secretion. Sex differences in CYP1A1, CYP1B1, and CYP2J11 mRNA levels were observed in both the wild-type and GHR deficient mice. No sex differences in the mRNA levels of COXs, LOXs, or CYP2E1 were observed in the wild-type mice. The constant GH infusion induced heart CYP1A1 and CYP2J11, and decreased CYP1B1 in the male C57/B6 mice constantly infused with GH (0.4 μg/h, 7 days). The activity of rat Cyp2j3 promoter was inhibited by the STAT5B protein, but was activated by C/EBPα (CEBPA). Compared with the constant GH administration, the levels of the nuclear phosphorylated STAT5B protein and its binding to the rat Cyp2j3 promoter were higher following the pulsatile GH administration. The constant GH infusion decreased the binding of the nuclear phosphorylated STAT5B protein to the mouse Cyp2j11 promoter. The data suggest the sexually dimorphic transcription of heart AA metabolizing enzymes, which might alter the risk and outcome of CVDs. GHR-STAT5B signal transduction pathway may be involved in the sex difference in heart CYP2J levels.
Collapse
Affiliation(s)
- Furong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xuming Yu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Chunyan He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiufang Ouyang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jinhua Wu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jie Li
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Junjie Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xuejiao Duan
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yu Wan
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jiang Yue
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
14
|
Rose E, Flanagan SP, Jones AG. The Effects of Synthetic Estrogen Exposure on the Sexually Dimorphic Liver Transcriptome of the Sex-Role-Reversed Gulf Pipefish. PLoS One 2015; 10:e0139401. [PMID: 26448558 PMCID: PMC4598134 DOI: 10.1371/journal.pone.0139401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/12/2015] [Indexed: 01/22/2023] Open
Abstract
Species exhibiting sex-role reversal provide an unusual perspective on the evolution of sex roles and sex differences. However, the proximate effects of sex-role reversal are largely unknown. Endocrine disruptors provide an experimental mechanism to address hormonal regulation of sexually dimorphic gene expression in sex-role-reversed taxa. Here, we investigate gene expression patterns in the liver of the sex-role-reversed Gulf pipefish, because the liver is known to be sexually dimorphic and estrogen-regulated in species with conventional sex roles. Using next-generation RNA-sequencing technology (RNA-seq), we detected sexually dimorphic hepatic gene expression patterns, with a total of 482 differentially expressed genes between the sexes in Gulf pipefish. Two-thirds of these genes were over-expressed in females, and the sex-specific transcriptomes of this sex-role-reversed pipefish’s liver were superficially similar to those of fishes with conventional sex-roles. We exposed females, pregnant males, and non-pregnant males to 17α-ethinylestradiol (EE2) at ecologically relevant concentrations of 5ng/L and compared gene expression patterns in the livers of exposed fish to control fish. Several genes that were up-regulated in EE2-exposed males relative to control males were also found to be female-biased in control animals. These genes included several of the classic estrogen biomarkers, such as vitellogenin, choriogenin, and zona pellucida. Thus, estrogen exposure induced feminization of the male liver transcriptome in a sex-role-reversed pipefish. These results suggest that the ancestral state of estrogen-regulated female reproductive physiology has been retained in all sex-role-reversed vertebrates thus far studied, despite substantial evolution of the hormonal regulation of ornamentation and mating behavior in these interesting taxa.
Collapse
Affiliation(s)
- Emily Rose
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, Texas, 77845, United States of America
- * E-mail:
| | - Sarah P. Flanagan
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, Texas, 77845, United States of America
| | - Adam G. Jones
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, Texas, 77845, United States of America
| |
Collapse
|
15
|
Babelova A, Burckhardt BC, Salinas-Riester G, Pommerenke C, Burckhardt G, Henjakovic M. Next generation sequencing of sex-specific genes in the livers of obese ZSF1 rats. Genomics 2015. [PMID: 26200819 DOI: 10.1016/j.ygeno.2015.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Type 2 diabetes induces pathophysiological changes in the liver. The aim of this study was to identify differently expressed genes in the livers of male and female ZSF1 rats (ZDFxSHHF-hybrid, generation F1), a model for type 2 diabetes. Gene expression was investigated using next-generation sequencing (NGS). Selected candidate genes were verified by real-time PCR in the livers of obese and lean rats. 103 sex-different genes, associated to pathways "response to chemical stimulus", "lipid metabolism", and "response to organic substance", were identified. Male-specific genes were involved in hepatic metabolism, detoxification, and secretion, e.g. cytochrome P450 2c11 (Cyp2c11), Cyp4a2, glutathione S-transferases mu 2 (Gstm2), and Slc22a8 (organic anion transporter 3, Oat3). Most female-specific genes were associated to lipid metabolism (e.g. glycerol-3-phosphate acyltransferase 1, Gpam) or glycolysis (e.g. glucokinase, Gck). Our data suggest the necessity to pay attention to sex- and diabetes-dependent changes in pre-clinical testing of hepatic metabolized and secreted drugs.
Collapse
Affiliation(s)
- Andrea Babelova
- Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 83391 Bratislava, Slovak Republic
| | - Birgitta C Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Goettingen, 37073 Goettingen, Germany
| | - Gabriela Salinas-Riester
- Department of Developmental Biochemistry, DNA Microarray and Deep-Sequencing Facility, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Claudia Pommerenke
- Department of Developmental Biochemistry, DNA Microarray and Deep-Sequencing Facility, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Gerhard Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Goettingen, 37073 Goettingen, Germany
| | - Maja Henjakovic
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Goettingen, 37073 Goettingen, Germany.
| |
Collapse
|
16
|
Pinheiro P, Marinho A, Antunes A, Marques M, Pereira S, Miranda J. Sex differences in hepatic and intestinal contributions to nevirapine biotransformation in rats. Chem Biol Interact 2015; 233:115-21. [DOI: 10.1016/j.cbi.2015.03.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/10/2015] [Accepted: 03/22/2015] [Indexed: 01/01/2023]
|
17
|
The incidence of sexually dimorphic gene expression varies greatly between tissues in the rat. PLoS One 2014; 9:e115792. [PMID: 25548914 PMCID: PMC4280129 DOI: 10.1371/journal.pone.0115792] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/01/2014] [Indexed: 11/20/2022] Open
Abstract
The sexually dimorphic expression of genes across 26 somatic rat tissues was using Affymetrix RAE-230 genechips. We considered probesets to be sexually dimorphically expressed (SDE) if they were measurably expressed above background in at least one sex, there was at least a two-fold difference in expression (dimorphism) between the sexes, and the differences were statistically significant after correcting for false discovery. 14.5% of expressed probesets were SDE in at least one tissue, with higher expression nearly twice as prevalent in males compared to females. Most were SDE in a single tissue. Surprisingly, nearly half of the probesets that were (SDE) in multiple tissues were oppositely sex biased in different tissues, and most SDE probesets were also expressed without sex bias in other tissues. Two genes were widely SDE: Xist (female-only) and Eif2s3y (male-only). The frequency of SDE probesets varied widely between tissues, and was highest in the duodenum (6.2%), whilst less than 0.05% in over half of the surveyed tissues. The occurrence of SDE probesets was not strongly correlated between tissues. Within individual tissues, however, relational networks of SDE genes were identified. In the liver, networks relating to differential metabolism between the sexes were seen. The estrogen receptor was implicated in differential gene expression in the duodenum. To conclude, sexually dimorphic gene expression is common, but highly tissue-dependent. Sexually dimorphic gene expression may provide insights into mechanisms underlying phenotypic sex differences. Online data are provided as a resource for further analyses (GEO reference GSE63362).
Collapse
|
18
|
Abstract
Rhythmic events in the female reproductive system depend on the coordinated and synchronized activity of multiple neuroendocrine and endocrine tissues. This coordination is facilitated by the timing of gene expression and cellular physiology at each level of the hypothalamo-pituitary-ovarian (HPO) axis, including the basal hypothalamus and forebrain, the pituitary gland, and the ovary. Central to this pathway is the primary circadian pacemaker in the suprachiasmatic nucleus (SCN) that, through its myriad outputs, provides a temporal framework for gonadotropin release and ovulation. The heart of the timing system, a transcription-based oscillator, imparts SCN pacemaker cells and a company of peripheral tissues with the capacity for daily oscillations of gene expression and cellular physiology. Although the SCN sits comfortably at the helm, peripheral oscillators (such as the ovary) have undefined but potentially critical roles. Each cell type of the ovary, including theca cells, granulosa cells, and oocytes, harbor a molecular clock implicated in the processes of follicular growth, steroid hormone synthesis, and ovulation. The ovarian clock is influenced by the reproductive cycle and diseases that perturb the cycle and/or follicular growth can disrupt the timing of clock gene expression in the ovary. Chronodisruption is known to negatively affect reproductive function and fertility in both rodent models and women exposed to shiftwork schedules. Thus, influencing clock function in the HPO axis with chronobiotics may represent a novel avenue for the treatment of common fertility disorders, particularly those resulting from chronic circadian disruption.
Collapse
Affiliation(s)
- Michael T. Sellix
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
19
|
Murphy ZC, Pezuk P, Menaker M, Sellix MT. Effects of ovarian hormones on internal circadian organization in rats. Biol Reprod 2013; 89:35. [PMID: 23843233 DOI: 10.1095/biolreprod.113.109322] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The circadian clock in the suprachiasmatic nucleus (SCN) of the hypothalamus is the central pacemaker driving rhythms in endocrine physiology. Gonadal steroid hormones affect behavioral rhythms and clock gene expression. However, the impact of fluctuating ovarian steroid levels during the estrous cycle on internal circadian organization remains to be determined. Further, it is not known if steroid hormone depletion, as in menopause, affects the timing system. To determine the influence of estrous cycle stage and steroid depletion on circadian organization, we measured clock gene expression in the SCN and peripheral tissues from cycling and ovariectomized (OVX) period1-luciferase (per1-luc) transgenic rats. The estrous cycle had modest effects on mean phase and phase distribution of per1-luc expression in the SCN. Surprisingly, peak per1-luc expression in the SCN was widely distributed mainly at night, regardless of cycle stage, an effect eliminated by OVX. Treatment of SCN tissue explants with ovarian steroids did not significantly affect per1-luc expression, suggesting that brain regions outside the SCN mediate the phasic effects of steroids. Our data demonstrate that estrous cycle stage has tissue-dependent effects on the phase of per1-luc expression, phase synchrony among oscillators, and the phase relationship between some peripheral clocks and the light-dark cycle. They also reveal that steroid hormone depletion following OVX alters the timing system, suggesting that the decline in hormone levels, common during the transition to menopause, may be associated with irregular internal circadian organization. This effect on the timing system could contribute to the behavioral and physiological changes associated with this transition.
Collapse
Affiliation(s)
- Zachary C Murphy
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | | |
Collapse
|
20
|
Lu YF, Jin T, Xu Y, Zhang D, Wu Q, Zhang YKJ, Liu J. Sex differences in the circadian variation of cytochrome p450 genes and corresponding nuclear receptors in mouse liver. Chronobiol Int 2013; 30:1135-43. [PMID: 23926955 DOI: 10.3109/07420528.2013.805762] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sex differences and circadian variation are two major factors that affect the expression of drug-processing genes. This study aimed to examine sex differences in the circadian variation of hepatic cytochrome P450 (Cyp) genes and corresponding nuclear receptors. Adult mice were acclimated to environmentally controlled facilities for 2 wks, and livers were collected every 4 h during a 24-h period. Total RNA and protein were isolated and subjected to real-time reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis. The mRNA expression of the aryl hydrocarbon receptor (AhR) and AhR-regulated Cyp1a1 and Cyp1a2 were higher in females and higher during the light phase. The mRNA expression of constitutive and rostane receptor (CAR) and CYP2B10 protein was female-predominant and higher in the dark phase. Pregnane X receptor (PXR) peaked around 18:00 h, but PXR-regulated Cyp3a11 and Cyp3a25 were higher at 10:00 h, without apparent sex dimorphism at protein levels. Peroxisome proliferator-activated receptor-α (PPARα), Cyp4a10, and Cyp4a14 were higher in females and peaked between 14:00 and 18:00 h. The mRNA levels of farnesoid X receptor (FXR), Cyp7a1, and Cyp27a1 peaked around 18:00 h and CYP7A1 protein was higher during the dark phase and higher in females. Cyp7b1(male-predominant) and Cyp2a4 (female-predominant) both showed circadian variation. Circadian variation of hepatic clock genes such as nuclear receptor Rev-erbα, cryptochrome 1 (Cry1), and brain muscle ARNT-like protein 1 (Bmal1) showed distinct patterns. Sex differences and circadian rhythmicity of Cyp genes and corresponding nuclear receptors exist in mouse liver that could impact xenobiotic metabolism and toxicity at different times of the day.
Collapse
Affiliation(s)
- Yuan-Fu Lu
- Key Laboratory of Basic Pharmacology of Guizhou, Zunyi Medical College , Zunyi , China
| | | | | | | | | | | | | |
Collapse
|
21
|
Walker DM, Goetz BM, Gore AC. Dynamic postnatal developmental and sex-specific neuroendocrine effects of prenatal polychlorinated biphenyls in rats. Mol Endocrinol 2013; 28:99-115. [PMID: 24284824 DOI: 10.1210/me.2013-1270] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gestational exposures to estrogenic compounds, both endogenous hormones and exogenous endocrine-disrupting chemicals (EDCs), have long-term effects on reproductive physiology and behavior. We tested the hypothesis that prenatal treatment of rats with low doses of Aroclor 1221 (A1221), a weakly estrogenic polychlorinated biphenyl mix previously used in industry, or estradiol benzoate (EB), alters development of the hypothalamus in a sexually dimorphic manner and subsequently perturbs reproductive function. Pregnant Sprague-Dawley rats were injected on embryonic days 16 and 18 with vehicle (dimethylsulfoxide), A1221 (1 mg/kg), or EB (50 μg/kg). Developmental milestones were monitored, and on postnatal days 15, 30, 45, and 90, 1 male and 1 female per litter were euthanized. Because of their key roles in the mediation of steroid actions on reproductive function, the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC) were punched for a low-density quantitative PCR array of 48 neuroendocrine genes and analysis of DNA methylation of a subset of genes. Gestational exposure to A1221 or EB delayed the timing of puberty in males and disrupted estrous cyclicity in females. In the AVPV, 28 genes were affected by treatment in a developmental stage-specific manner, mostly in females, which exhibited a masculinized expression profile. This included 2 clock genes, Per2 and Arntl, implicating circadian circuits as being vulnerable to endocrine disruption. DNA methylation analysis of 2 genes, Per2 and Ar, showed no effect of EDCs and suggested alternative mechanisms for the altered mRNA levels. In the ARC, 12 genes were affected by treatment, mostly in males, again with dynamic developmental changes. Bionetwork analysis of relationships among genes, hormones, and physiological markers showed sexually dimorphic effects of estrogenic EDC exposures, with the female AVPV and the male ARC being most vulnerable, and provided novel relationships among hypothalamic genes and postnatal reproductive maturation.
Collapse
Affiliation(s)
- Deena M Walker
- The Institute for Neuroscience (D.M.W., A.C.G.), Center for Computational Biology and Bioinformatics (B.M.G.), Division of Pharmacology and Toxicology (A.C.G.), College of Pharmacy, and Institute for Cell and Molecular Biology (A.C.G.), The University of Texas at Austin, Austin, Texas 78712
| | | | | |
Collapse
|
22
|
Yamatoya H, Kawaguchi H, Yajima K, Kadokura H, Yoshikawa T, Yamashita R, Shiraishi M, Miyamoto A, Miyoshi N. Data on Wistar Hannover rats from a general toxicity study. Exp Anim 2012; 61:467-76. [PMID: 22850647 DOI: 10.1538/expanim.61.467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The aim of this study was to collect data on chronological changes in clinical laboratory tests, pathological examinations, and hepatic drug-metabolizing enzymes from Wistar Hannover rats at 8, 10, 19, and 32 weeks of age. The serum triglyceride concentration and the serum LDL cholesterol level were higher in males than in females at all ages. In contrast, serum total protein and creatinine concentrations and cholinesterase activity were lower in males than in females. In addition, sex differences were confirmed in pituitary weight and hepatic CYP3A2 and CYP2C11 activities. In conclusion, the general toxicological data noted in clinical laboratory tests, pathological examinations, and hepatic drug-metabolizing enzymes relating to chronological changes and sex differences may be useful in assessing drug-related toxicity in this strain.
Collapse
Affiliation(s)
- Hideyuki Yamatoya
- The United Graduate School of Veterinary Science, Yamaguchi University, 1677–1 Yoshida, Yamaguchi 753-8515, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wang SH, Yeh SH, Lin WH, Yeh KH, Yuan Q, Xia NS, Chen DS, Chen PJ. Estrogen receptor α represses transcription of HBV genes via interaction with hepatocyte nuclear factor 4α. Gastroenterology 2012; 142:989-998.e4. [PMID: 22240483 DOI: 10.1053/j.gastro.2011.12.045] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 12/08/2011] [Accepted: 12/26/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Women with hepatitis B virus (HBV) infection usually have lower viral loads than men, reducing their risk of liver cancer. There are 2 androgen-responsive elements in the HBV enhancer I that contribute to higher viral titers in men. We investigated whether and how estrogen signaling affects progression of HBV infection. METHODS Ovariectomy and estrogen supplementation were used to evaluate the effect of estrogen on HBV titers in transgenic mice with replicating HBV in hepatocytes. The effect of estrogen signaling on transcription of HBV genes, and the mechanisms of regulation, were studied in HepG2 cells. RESULTS HBV titers increased in female mice after ovariectomy and decreased in male mice supplemented with estrogen. Hepatic expression of estrogen receptor (ER)-α was increased by estrogen exposure. In HepG2 cells, up-regulation of ER-α reduced HBV transcription, which required a specific region within enhancer I. Direct DNA binding of ER-α and histone deacetylase activity were not required for ER-α-mediated repression of HBV genes. Overexpression of hepatocyte nuclear factor (HNF)-4α, which binds to this region, overcame the repressive effect of ER-α. ER-α did not repress transcription of an HBV replicon with a mutant HNF-4α binding site within enhancer I. Coimmunoprecipitation assays showed an interaction between ER-α and HNF-4α; this interaction prevented HNF-4α binding to enhancer I and activation of HBV transcription. CONCLUSIONS Estrogen can repress transcription of HBV genes by up-regulating ER-α, which interacts with and alters binding of HNF-4α to the HBV enhancer I. These findings might account for the lower viral load and reduced incidence of liver cancer in HBV-infected women than men.
Collapse
Affiliation(s)
- Sheng-Han Wang
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Nakamura K, Kato N, Aizawa K, Mizutani R, Yamauchi J, Tanoue A. Expression of albumin and cytochrome P450 enzymes in HepG2 cells cultured with a nanotechnology-based culture plate with microfabricated scaffold. J Toxicol Sci 2012; 36:625-33. [PMID: 22008537 DOI: 10.2131/jts.36.625] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The Nanoculture plate (NCP) is a recently developed plate which essentially consists of a textured surface with specific characteristics that induce spheroid formation: microfabrications with a micro-square pattern on the culture surface. The NCP can be used to generate uniform adhesive spheroids of cancer cell lines using conventional techniques without the need of any animal compounds. In this study, we assessed the performance of human hepatoma cell line HepG2 cells cultured with an NCP to evaluate the effects of the NCP on their hepatocyte-specific functions. The NCP facilitated the formation of three-dimensional (3D) HepG2 cell architecture. HepG2 cells cultured with an NCP exhibited enhanced mRNA expression levels of albumin and cytochrome P450 (CYP) enzymes compared to those cultured with a two-dimensional (2D) conventional plate. The expression levels of two specific liver-enriched transcription factors, hepatocyte nuclear factor 4α (HNF4α) and CCAAT/enhancer binding protein α (C/EBPα), were higher in HepG2 cells grown with the NCP than those in HepG2 cells grown with conventional plates before albumin and CYP enzymes expression levels were increased. The inducibility of CYP1A2 and CYP3A4 mRNA following exposure to inducers in HepG2 cells cultured with an NCP was comparable to that in HepG2 cells cultured with conventional plates, while the expression levels of CYP1A2 and CYP3A4 mRNA following exposure to inducers were higher when using an NCP than when using conventional plates. These results suggest that the use of an NCP enhances the hepatocyte-specific functions of HepG2 cells, such as drug-metabolizing enzyme expression, making the NCP/HepG2 system a useful tool for evaluating drug metabolism in vitro.
Collapse
Affiliation(s)
- Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|