1
|
Zhu H, Xiao H, Li L, Yang M, Lin Y, Zhou J, Zhang X, Zhou Y, Lan X, Liu J, Zeng J, Wang L, Zhong Y, Qian X, Cao Z, Liu P, Mei H, Cai M, Cai X, Tang Z, Hu L, Zhou R, Xu X, Yang H, Wang J, Jin X, Zhou A. Novel insights into the genetic architecture of pregnancy glycemic traits from 14,744 Chinese maternities. CELL GENOMICS 2024; 4:100631. [PMID: 39389014 PMCID: PMC11602577 DOI: 10.1016/j.xgen.2024.100631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 12/14/2023] [Accepted: 07/17/2024] [Indexed: 10/12/2024]
Abstract
Glycemic traits are critical indicators of maternal and fetal health during pregnancy. We performed genetic analysis for five glycemic traits in 14,744 Chinese pregnant women. Our genome-wide association study identified 25 locus-trait associations, including established links between gestational diabetes mellitus (GDM) and the genes CDKAL1 and MTNR1B. Notably, we discovered a novel association between fasting glucose during pregnancy and the ESR1 gene (estrogen receptor), which was validated by an independent study in pregnant women. The ESR1-GDM link was recently reported by the FinnGen project. Our work enhances the findings in East Asian populations and highlights the need for independent studies. Further analyses, including genetic correlation, Mendelian randomization, and transcriptome-wide association studies, provided genetic insights into the relationship between pregnancy glycemic traits and hypertension. Overall, our findings advance the understanding of genetic architecture of pregnancy glycemic traits, especially in East Asian populations.
Collapse
Affiliation(s)
- Huanhuan Zhu
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China
| | - Han Xiao
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Ying Lin
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jieqiong Zhou
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xinyi Zhang
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Zhou
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xianmei Lan
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiuying Liu
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Jingyu Zeng
- BGI Research, Shenzhen 518083, China; College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lin Wang
- BGI Research, Shenzhen 518083, China
| | - Yuanyuan Zhong
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xiaobo Qian
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongqiang Cao
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Hong Mei
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Xiaonan Cai
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Liqin Hu
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518120, China
| | - Huanming Yang
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, BGI, Shenzhen 518120, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | | | - Xin Jin
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China.
| | - Aifen Zhou
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China; Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China.
| |
Collapse
|
2
|
Barton BE, Erickson JA, Allred SI, Jeffries JM, Stephens KK, Hunter MI, Woodall KA, Winuthayanon W. Reversible female contraceptives: historical, current, and future perspectives†. Biol Reprod 2024; 110:14-32. [PMID: 37941453 PMCID: PMC10790348 DOI: 10.1093/biolre/ioad154] [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: 06/06/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023] Open
Abstract
Contraception is a practice with extensive and complicated social and scientific histories. From cycle tracking, to the very first prescription contraceptive pill, to now having over-the-counter contraceptives on demand, family planning is an aspect of healthcare that has undergone and will continue to undergo several transformations through time. This review provides a comprehensive overview of current reversible hormonal and non-hormonal birth control methods as well as their mechanism of action, safety, and effectiveness specifically for individuals who can become pregnant. Additionally, we discuss the latest Food and Drug Administration (FDA)-approved hormonal method containing estetrol and drospirenone that has not yet been used worldwide as well as the first FDA-approved hormonal over-the-counter progestin-only pills. We also review available data on novel hormonal delivery through microchip, microneedle, and the latest FDA-approved non-hormonal methods such as vaginal pH regulators. Finally, this review will assist in advancing female contraceptive method development by underlining constructive directions for future pursuits. Information was gathered from the NCBI and Google Scholars databases using English and included publications from 1900 to present. Search terms included contraceptive names as well as efficacy, safety, and mechanism of action. In summary, we suggest that investigators consider the side effects and acceptability together with the efficacy of contraceptive candidate towards their development.
Collapse
Affiliation(s)
- Brooke E Barton
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Jeffery A Erickson
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
- Translational Bioscience Program, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Stephanie I Allred
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jenna M Jeffries
- College of Art & Science, Washington State University, Pullman, WA, USA
| | - Kalli K Stephens
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
- Translational Bioscience Program, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mark I Hunter
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Kirby A Woodall
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Wipawee Winuthayanon
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
3
|
Birgersson M, Indukuri R, Lindquist L, Stepanauskaite L, Luo Q, Deng Q, Archer A, Williams C. Ovarian ERβ cistrome and transcriptome reveal chromatin interaction with LRH-1. BMC Biol 2023; 21:277. [PMID: 38031019 PMCID: PMC10688478 DOI: 10.1186/s12915-023-01773-1] [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: 03/03/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Estrogen receptor beta (ERβ, Esr2) plays a pivotal role in folliculogenesis and ovulation, yet its exact mechanism of action is mainly uncharacterized. RESULTS We here performed ERβ ChIP-sequencing of mouse ovaries followed by complementary RNA-sequencing of wild-type and ERβ knockout ovaries. By integrating the ERβ cistrome and transcriptome, we identified its direct target genes and enriched biological functions in the ovary. This demonstrated its strong impact on genes regulating organism development, cell migration, lipid metabolism, response to hypoxia, and response to estrogen. Cell-type deconvolution analysis of the bulk RNA-seq data revealed a decrease in luteal cells and an increased proportion of theca cells and a specific type of cumulus cells upon ERβ loss. Moreover, we identified a significant overlap with the gene regulatory network of liver receptor homolog 1 (LRH-1, Nr5a2) and showed that ERβ and LRH-1 extensively bound to the same chromatin locations in granulosa cells. Using ChIP-reChIP, we corroborated simultaneous ERβ and LRH-1 co-binding at the ERβ-repressed gene Greb1 but not at the ERβ-upregulated genes Cyp11a1 and Fkbp5. Transactivation assay experimentation further showed that ERβ and LRH-1 can inhibit their respective transcriptional activity at classical response elements. CONCLUSIONS By characterizing the genome-wide endogenous ERβ chromatin binding, gene regulations, and extensive crosstalk between ERβ and LRH-1, along with experimental corroborations, our data offer genome-wide mechanistic underpinnings of ovarian physiology and fertility.
Collapse
Affiliation(s)
- Madeleine Birgersson
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Rajitha Indukuri
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
| | - Linnéa Lindquist
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Lina Stepanauskaite
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Qing Luo
- Department of Physiology and Pharmacology, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Amena Archer
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Cecilia Williams
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden.
| |
Collapse
|
4
|
Le Ciclé C, Pacini V, Rama N, Tauszig-Delamasure S, Airaud E, Petit F, de Beco S, Cohen-Tannoudji J, L'hôte D. The Neurod1/4-Ntrk3-Src pathway regulates gonadotrope cell adhesion and motility. Cell Death Discov 2023; 9:327. [PMID: 37658038 PMCID: PMC10474047 DOI: 10.1038/s41420-023-01615-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 09/03/2023] Open
Abstract
Pituitary gonadotrope cells are essential for the endocrine regulation of reproduction in vertebrates. These cells emerge early during embryogenesis, colonize the pituitary glands and organize in tridimensional networks, which are believed to be crucial to ensure proper regulation of fertility. However, the molecular mechanisms regulating the organization of gonadotrope cell population during embryogenesis remain poorly understood. In this work, we characterized the target genes of NEUROD1 and NEUROD4 transcription factors in the immature gonadotrope αT3-1 cell model by in silico functional genomic analyses. We demonstrated that NEUROD1/4 regulate genes belonging to the focal adhesion pathway. Using CRISPR/Cas9 knock-out approaches, we established a double NEUROD1/4 knock-out αT3-1 cell model and demonstrated that NEUROD1/4 regulate cell adhesion and cell motility. We then characterized, by immuno-fluorescence, focal adhesion number and signaling in the context of NEUROD1/4 insufficiency. We demonstrated that NEUROD1/4 knock-out leads to an increase in the number of focal adhesions associated with signaling abnormalities implicating the c-Src kinase. We further showed that the neurotrophin tyrosine kinase receptor 3 NTRK3, a target of NEUROD1/4, interacts physically with c-Src. Furthermore, using motility rescue experiments and time-lapse video microscopy, we demonstrated that NTRK3 is a major regulator of gonadotrope cell motility. Finally, using a Ntrk3 knock-out mouse model, we showed that NTRK3 regulates gonadotrope cells positioning in the developing pituitary, in vivo. Altogether our study demonstrates that the Neurod1/4-Ntrk3-cSrc pathway is a major actor of gonadotrope cell mobility, and thus provides new insights in the regulation of gonadotrope cell organization within the pituitary gland.
Collapse
Affiliation(s)
- Charles Le Ciclé
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Vincent Pacini
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Nicolas Rama
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université Lyon1, 69008, Lyon, France
| | - Servane Tauszig-Delamasure
- Institut NeuroMyoGène - CNRS UMR 5310 - Inserm U1217 de Lyon - UCBL Lyon 1, Faculté de Médecine et de Pharmacie, Lyon, France
| | - Eloïse Airaud
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Florence Petit
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Simon de Beco
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Joëlle Cohen-Tannoudji
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - David L'hôte
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France.
| |
Collapse
|
5
|
Moreira ARS, Lim J, Urbaniak A, Banik J, Bronson K, Lagasse A, Hardy L, Haney A, Allensworth M, Miles TK, Gies A, Byrum SD, Wilczynska A, Boehm U, Kharas M, Lengner C, MacNicol MC, Childs GV, MacNicol AM, Odle AK. Musashi Exerts Control of Gonadotrope Target mRNA Translation During the Mouse Estrous Cycle. Endocrinology 2023; 164:bqad113. [PMID: 37477898 PMCID: PMC10402870 DOI: 10.1210/endocr/bqad113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023]
Abstract
The anterior pituitary controls key biological processes, including growth, metabolism, reproduction, and stress responses through distinct cell types that each secrete specific hormones. The anterior pituitary cells show a remarkable level of cell type plasticity that mediates the shifts in hormone-producing cell populations that are required to meet organismal needs. The molecular mechanisms underlying pituitary cell plasticity are not well understood. Recent work has implicated the pituitary stem cell populations and specifically, the mRNA binding proteins of the Musashi family in control of pituitary cell type identity. In this study we have identified the target mRNAs that mediate Musashi function in the adult mouse pituitary and demonstrate the requirement for Musashi function in vivo. Using Musashi RNA immunoprecipitation, we identify a cohort of 1184 mRNAs that show specific Musashi binding. Identified Musashi targets include the Gnrhr mRNA, which encodes the gonadotropin-releasing hormone receptor (GnRHR), and the Fshb mRNA, encoding follicle-stimulating hormone (FSH). Reporter assays reveal that Musashi functions to exert repression of translation of the Fshb mRNA, in addition to the previously observed repression of the Gnrhr mRNA. Importantly, mice engineered to lack Musashi in gonadotropes demonstrate a failure to repress translation of the endogenous Gnrhr and Fshb mRNAs during the estrous cycle and display a significant heterogeneity in litter sizes. The range of identified target mRNAs suggests that, in addition to these key gonadotrope proteins, Musashi may exert broad regulatory control over the pituitary proteome in a cell type-specific manner.
Collapse
Affiliation(s)
- Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Katherine Bronson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Linda Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melody Allensworth
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tiffany K Miles
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Allen Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Arkansas Children's Research Institute, Arkansas Children's Hospital, Little Rock, AR 72202, USA
| | - Ania Wilczynska
- Bit.bio, The Dorothy Hodgkin Building, Babraham Research Campus, Cambridge CB22 3FH, UK
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg 66421, Germany
| | - Michael Kharas
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christopher Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
6
|
Ouyang Q, Xie H, Ran M, Zhang X, He Z, Lin Y, Hu S, Hu J, He H, Li L, Liu H, Wang J. Estrogen Receptor Gene 1 ( ESR1) Mediates Lipid Metabolism in Goose Hierarchical Granulosa Cells Rather than in Pre-Hierarchical Granulosa Cells. BIOLOGY 2023; 12:962. [PMID: 37508392 PMCID: PMC10376489 DOI: 10.3390/biology12070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/18/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
(1) Background: The role of estrogen receptor gene 1 (ESR1) in female reproduction and lipid metabolism has been extensively investigated. However, its contribution to lipid metabolism during the development of poultry follicles remains unclear. (2) Methods: This study aimed to explore the function of ESR1 via overexpressing (ESR1ov) and interfering (ESR1si) with its expression in pre-hierarchical granulosa cells (phGCs) and hierarchical granulosa cells (poGCs). (3) Results: We successfully cloned and obtained an 1866 bp segment of the full-length CDS region of the Sichuan white goose ESR1 gene. In phGCs of the ESR1ov and ESR1si groups, there were no significant changes compared to the control group. However, in poGCs, the ESR1ov group exhibited decreased lipid deposition, triglycerides, and cholesterol compared to the control group, while the ESR1si group showed increased lipid deposition, triglycerides, and cholesterol. The expression of APOB and PPARα was significantly reduced in the ESR1ov group compared to the ESR1ov-NC group. Moreover, significant changes in the expression of ACCα, DGAT1, SCD, CPT1, and ATGL were observed between the ESR1si and ESR1si-NC group. (4) Conclusions: These findings shed light on the function and molecular mechanism of ESR1 in lipid metabolism in goose poGCs, providing a better understanding of the physiological process of goose follicular development.
Collapse
Affiliation(s)
- Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hengli Xie
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingxia Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xi Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiyu He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yueyue Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
7
|
Isola JVV, Ko S, Ocañas SR, Stout MB. Role of Estrogen Receptor α in Aging and Chronic Disease. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230005. [PMID: 37425648 PMCID: PMC10327608 DOI: 10.20900/agmr20230005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Estrogen receptor alpha (ERα) plays a crucial role in reproductive function in both sexes. It also mediates cellular responses to estrogens in multiple nonreproductive organ systems, many of which regulate systemic metabolic homeostasis and inflammatory processes in mammals. The loss of estrogens and/or ERα agonism during aging is associated with the emergence of several comorbid conditions, particularly in females undergoing the menopausal transition. Emerging data also suggests that male mammals likely benefit from ERα agonism if done in a way that circumvents feminizing characteristics. This has led us, and others, to speculate that tissue-specific ERα agonism may hold therapeutic potential for curtailing aging and chronic disease burden in males and females that are at high-risk of cancer and/or cardiovascular events with traditional estrogen replacement therapies. In this mini-review, we emphasize the role of ERα in the brain and liver, summarizing recent evidence that indicates these two organs systems mediate the beneficial effects of estrogens on metabolism and inflammation during aging. We also discuss how 17α-estradiol administration elicits health benefits in an ERα-dependent manner, which provides proof-of-concept that ERα may be a druggable target for attenuating aging and age-related disease burden.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Chen KH, Doliba N, May CL, Roman J, Ustione A, Tembo T, Negron A, Radovick S, Piston DW, Glaser B, Kaestner KH, Matschinsky FM. Genetic activation of glucokinase in a minority of pancreatic beta cells causes hypoglycemia in mice. Life Sci 2022; 309:120952. [PMID: 36100080 PMCID: PMC10312065 DOI: 10.1016/j.lfs.2022.120952] [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: 01/17/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 01/05/2023]
Abstract
AIMS Glucokinase (GK) is expressed in the glucose-sensing cells of the islets of Langerhans and plays a critical role in glucose homeostasis. Here, we tested the hypothesis that genetic activation of GK in a small subset of β-cells is sufficient to change the glucose set-point of the whole islet. MATERIAL AND METHODS Mouse models of cell-type specific GK deficiency (GKKO) and genetic enzyme activation (GKKI) in a subset of β-cells were obtained by crossing the αGSU (gonadotropin alpha subunit)-Cre transgene with the appropriate GK mutant alleles. Metabolic analyses consisted of glucose tolerance tests, perifusion of isolated islets and intracellular calcium measurements. KEY FINDINGS The αGSU-Cre transgene produced genetically mosaic islets, as Cre was active in 15 ± 1.2 % of β-cells. While mice deficient for GK in a subset of islet cells were normal, unexpectedly, GKKI mice were chronically hypoglycemic, glucose intolerant, and had a lower threshold for glucose stimulated insulin secretion. GKKI mice exhibited an average fasting blood glucose level of 3.5 mM. GKKI islets responded with intracellular calcium signals that spread through the whole islets at 1 mM and secreted insulin at 3 mM glucose. SIGNIFICANCE Genetic activation of GK in a minority of β-cells is sufficient to change the glucose threshold for insulin secretion in the entire islet and thereby glucose homeostasis in the whole animal. These data support the model in which β-cells with higher GK activity function as 'hub' or 'trigger' cells and thus control insulin secretion by the β-cell collective within the islet.
Collapse
Affiliation(s)
- Kevin H Chen
- Department of Biochemistry and Biophysics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Nicolai Doliba
- Department of Biochemistry and Biophysics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Catherine L May
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Jeffrey Roman
- Department of Biochemistry and Biophysics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Alessandro Ustione
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Teguru Tembo
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Ariel Negron
- Department of Medicine and Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Sally Radovick
- Department of Medicine and Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Benjamin Glaser
- Endocrinology and Metabolism Department, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA.
| | - Franz M Matschinsky
- Department of Biochemistry and Biophysics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19014, USA.
| |
Collapse
|
9
|
Saito K, Dickey JE, Rodeghiero SR, Toth BA, Kelly MJ, Deng Y, Singh U, Deng G, Jiang J, Cui H. Hypomorphism of a Novel Long ERα Isoform Causes Severe Reproductive Dysfunctions in Female Mice. Endocrinology 2022; 163:6742225. [PMID: 36181426 DOI: 10.1210/endocr/bqac160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 11/19/2022]
Abstract
Estrogen receptor alpha (ERα)-mediated estrogen signaling plays a pivotal role in both reproductive and nonreproductive functions. Transcriptional regulation of the ERα gene is highly complex, with multiple transcript variants being differentially produced across the tissues. However, tissue-specific variation and physiological specificity of the ERα variants are not yet fully understood. In an attempt to generate a Cre-dependently restorable ERα-null mouse for functional genetic studies, we unexpectedly produced ERα hypomorphic mice with biased downregulation of a previously unappreciated long ERα isoform that is enriched in the female reproductive organs (uterus and ovaries) and the pituitary but minimally expressed in the brain. Female homozygous mutant mice were capable of pregnancy but displayed irregular estrus cycle and rarely kept newborn pups alive. No significant morphological and pathological changes in reproductive system or disruption of body weight homeostasis were seen in female homozygous mutant mice. Collectively, our results define a tissue-specific enriched long ERα isoform and its preferential role in female reproductive function rather than body weight homeostasis.
Collapse
Affiliation(s)
- Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Jacob E Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Samuel R Rodeghiero
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Matthew J Kelly
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
- F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52241, USA
| |
Collapse
|
10
|
Henriques PC, Aquino NSS, Campideli-Santana AC, Silva JF, Araujo-Lopes R, Franci CR, Coimbra CC, Szawka RE. Hypothalamic Expression of Estrogen Receptor Isoforms Underlies Estradiol Control of Luteinizing Hormone in Female Rats. Endocrinology 2022; 163:6631316. [PMID: 35789268 DOI: 10.1210/endocr/bqac101] [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: 03/05/2022] [Indexed: 11/19/2022]
Abstract
Luteinizing hormone (LH) secretion during the ovarian cycle is governed by fluctuations in circulating estradiol (E2) that oppositely regulate kisspeptin neurons in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) of the hypothalamus. However, how these effects are orchestrated to achieve fertility is unknown. Here, we have tested the hypothesis that AVPV and ARC neurons have different sensitivities to E2 to coordinate changes in LH secretion. Cycling and ovariectomized rats with low and high E2 levels were used. As an index of E2 responsiveness, progesterone receptor (PR) was expressed only in the AVPV of rats with high E2, showing the preovulatory LH surge. On the other hand, kisspeptin neurons in the ARC responded to low E2 levels sufficient to suppress LH release. Notably, the Esr1/Esr2 ratio of gene expression was higher in the ARC than AVPV, regardless of E2 levels. Accordingly, the selective pharmacological activation of estrogen receptor α (ERα) required lower doses to induce PR in the ARC. The activation of ERβ, in turn, amplified E2-induced PR expression in the AVPV and the LH surge. Thus, ARC and AVPV neurons are differently responsive to E2. Lower E2 levels activate ERα in the ARC, whereas ERβ potentiates the E2 positive feedback in the AVPV, which appears related to the differential Esr1/Esr2 ratio in these 2 brain areas. Our findings provide evidence that the distinct expression of ER isoforms in the AVPV and ARC plays a key role in the control of periodic secretion of LH required for fertility in females.
Collapse
Affiliation(s)
- Patricia C Henriques
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Nayara S S Aquino
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Ana C Campideli-Santana
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Juneo F Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Roberta Araujo-Lopes
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Celso R Franci
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirao Preto, 14049-900, Universidade de São Paulo, Ribeirao Preto, Brazil
| | - Candido C Coimbra
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Raphael E Szawka
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Ma Y, Awe O, Radovick S, Yang X, Divall S, Wolfe A, Wu S. Lower FSH With Normal Fertility in Male Mice Lacking Gonadotroph Kisspeptin Receptor. Front Physiol 2022; 13:868593. [PMID: 35557961 PMCID: PMC9089166 DOI: 10.3389/fphys.2022.868593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/31/2022] [Indexed: 12/03/2022] Open
Abstract
The kisspeptin receptor, crucial for hypothalamic control of puberty and reproduction, is also present in the pituitary gland. Its role in the pituitary gland is not defined. Kisspeptin signaling via the Kiss1r could potentially regulate reproductive function at the level of pituitary gonadotrope. Using Cre/Lox technology, we deleted the Kiss1r gene in pituitary gonadotropes (PKiRKO). PKiRKO males have normal genital development (anogenital distance WT: 19.1 ± 0.4 vs. PKiRKO: 18.5 ± 0.4 mm), puberty onset, testes cell structure on gross histology, normal testes size, and fertility. PKiRKO males showed significantly decreased serum FSH levels compared to WT males (5.6 ± 1.9 vs. 10.2 ± 1.8 ng/ml) with comparable LH (1.1 ± 0.2 vs. 1.8 ± 0.4 ng/ml) and testosterone levels (351.8 ± 213.0 vs. 342.2 ± 183.0 ng/dl). PKiRKO females have normal puberty onset, cyclicity, LH and FSH levels and fertility. Overall, these findings indicate that absence of pituitary Kiss1r reduces FSH levels in male mice without affecting testis function. PKiRKO mice have normal reproductive function in both males and females.
Collapse
Affiliation(s)
- Yaping Ma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Olubusayo Awe
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sally Radovick
- Department of Pediatrics, Rutgers University Medical School, New Brunswick, NJ, United States
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Sara Divall
- Department of Pediatrics, University of Washington, Seattle's Children's Hospital, Seattle, United States
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
12
|
Xu XL, Huang ZY, Yu K, Li J, Fu XW, Deng SL. Estrogen Biosynthesis and Signal Transduction in Ovarian Disease. Front Endocrinol (Lausanne) 2022; 13:827032. [PMID: 35299973 PMCID: PMC8921451 DOI: 10.3389/fendo.2022.827032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/03/2022] [Indexed: 12/01/2022] Open
Abstract
Estrogen mainly binds to estrogen receptors (ERs) to regulate menstrual cycles and reproduction. The expression of ERalpha (ERα), ERbeta (ERβ), and G-protein-coupled estrogen receptor (GPER) mRNA could be detected in ovary, suggesting that they play an important role in estrogen signal transduction in ovary. And many studies have revealed that abnormal expression of estrogen and its receptors is closely related to ovarian disease or malignant tumors. With the continuous development and research of animal models, tissue-specific roles of both ERα and ERβ have been demonstrated in animals, which enable people to have a deeper understanding of the potential role of ER in regulating female reproductive diseases. Nevertheless, our current understanding of ERs expression and function in ovarian disease is, however, incomplete. To elucidate the biological mechanism behind ERs in the ovary, this review will focus on the role of ERα and ERβ in polycystic ovary syndrome (PCOS), ovarian cancer and premature ovarian failure (POF) and discuss the major challenges of existing therapies to provide a reference for the treatment of estrogen target tissue ovarian diseases.
Collapse
Affiliation(s)
- Xue-Ling Xu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Yuan Huang
- Department of Metabolism, Digestion and Reproduction, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Kun Yu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jun Li
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Wei Fu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shou-Long Deng
- National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Ma H, Ishida K, Xu C, Takahashi K, Li Y, Zhang C, Kang Q, Jia Y, Hu W, Matsumaru D, Nakanishi T, Hu J. Triphenyl phosphate delayed pubertal timing and induced decline of ovarian reserve in mice as an estrogen receptor antagonist. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 290:118096. [PMID: 34488164 DOI: 10.1016/j.envpol.2021.118096] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
Although concerns have been raised about the adverse effects of triphenyl phosphate (TPhP) on female fertility, its risk to ovarian functioning remains unknown. In this study, female C57BL/6 mice at postnatal day 21 were exposed on a daily basis to TPhP dose of 2, 10, and 50 mg/kg for 40 days. A significant delay in pubertal timing was observed in the mice exposed to 50 mg/kg of TPhP. An estrogen-responsive reporter transgenic mice assay demonstrated that TPhP significantly downregulated the estrogen receptor (ER) signaling by 45.1% in the whole body in the 50 mg/kg group, and by 14.7-43.7% in the uterus for all exposure groups compared with the control. This strong antagonistic activity of TPhP toward ER explained the delay in pubertal timing. A significant reduction in the number of follicles in all stages was observed in mice after being exposed to TPhP for 40 days at concentrations of 10 and 50 mg/kg, resulting in a decline of the ovarian reserve. The elevation of the follicle-stimulating hormone concentration may have contributed to this phenomenon, as controlled by the antagonistic activity of TPhP toward ER in the brain. The toxic effects of TPhP on ovarian functioning highlight this chemical as a potential risk factor for female fertility.
Collapse
Affiliation(s)
- Haojia Ma
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Keishi Ishida
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Chenke Xu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Kyosuke Takahashi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Yu Li
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Chenhao Zhang
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Qiyue Kang
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Yingting Jia
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Wenxin Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Daisuke Matsumaru
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Jianying Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
14
|
Ohara T, Nakamura TJ, Nakamura W, Tokuda IT. Modeling circadian regulation of ovulation timing: age-related disruption of estrous cyclicity. Sci Rep 2020; 10:16767. [PMID: 33028871 PMCID: PMC7541497 DOI: 10.1038/s41598-020-73669-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/14/2020] [Indexed: 11/21/2022] Open
Abstract
The circadian clocks within the hypothalamic–pituitary–gonadal axis control estrous cycles in female rodents. The suprachiasmatic nucleus (SCN), where the central clock is located, generates daily signals to trigger surge release of luteinizing hormone (LH), which in turn induces ovulation. It has been observed in aged rodents that output from the SCN such as neuronal firing activity is declined, and estrous cycles become irregular and finally stop. Circadian clock mutants display accelerated reproductive aging, suggesting the complicated interplay between the circadian system and the endocrine system. To investigate such circadian regulation of estrous cycles, we construct a mathematical model that describes dynamics of key hormones such as LH and of circadian clocks in the SCN and in the ovary, and simulate estrous cycles for various parameter values. Our simulation results demonstrate that reduction of the amplitude of the SCN signal, which is a symptom of aging, makes estrous cycles irregular. We also show that variation in the phase of the SCN signal and changes in the period of ovarian circadian clocks exacerbates the aging effect on estrous cyclicity. Our study suggests that misalignment between the SCN and ovarian circadian oscillations is one of the primary causes of the irregular estrous cycles.
Collapse
Affiliation(s)
- Takayuki Ohara
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany.
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Tokyo, Japan
| | - Wataru Nakamura
- Department of Oral-Chrono Physiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Isao T Tokuda
- Department of Mechanical Engineering, Ritsumeikan University, Kyoto, Japan.
| |
Collapse
|
15
|
SIX1 cooperates with RUNX1 and SMAD4 in cell fate commitment of Müllerian duct epithelium. Cell Death Differ 2020; 27:3307-3320. [PMID: 32572167 PMCID: PMC7852590 DOI: 10.1038/s41418-020-0579-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
During female mammal reproductive tract development, epithelial cells of the lower Müllerian duct are committed to become stratified squamous epithelium of the vagina and ectocervix, when the expression of ΔNp63 transcription factor is induced by mesenchymal cells. The absence of ΔNp63 expression leads to adenosis, the putative precursor of vaginal adenocarcinoma. Our previous studies with genetically engineered mouse models have established that fibroblast growth factor (FGF)/mitogen-activated protein kinase (MAPK), bone morphogenetic protein (BMP)/SMAD, and activin A/runt-related transcription factor 1 (RUNX1) signaling pathways are independently required for ΔNp63 expression in Müllerian duct epithelium (MDE). Here, we report that sine oculis homeobox homolog 1 (SIX1) plays a critical role in the activation of ΔNp63 locus in MDE as a downstream transcription factor of mesenchymal signals. In the developing mouse reproductive tract, SIX1 expression was restricted to MDE within the future cervix and vagina. SIX1 expression was totally absent in SMAD4 null MDE and was reduced in RUNX1 null and FGFR2 null MDE, indicating that SIX1 is under the control of vaginal mesenchymal factors: BMP4, activin A and FGF7/10. Furthermore, Six1, Runx1, and Smad4 gene-dose-dependently activated ΔNp63 expression in MDE within the vaginal fornix. Using a mouse model of diethylstilbestrol (DES)-associated vaginal adenosis, we found DES action through epithelial estrogen receptor α (ESR1) inhibits activation of ΔNp63 locus in MDE by transcriptionally repressing SIX1 and RUNX1 in the vaginal fornix.
Collapse
|
16
|
Li J, Ge W. Zebrafish as a model for studying ovarian development: Recent advances from targeted gene knockout studies. Mol Cell Endocrinol 2020; 507:110778. [PMID: 32142861 DOI: 10.1016/j.mce.2020.110778] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022]
Abstract
Ovarian development is a complex process controlled by precise coordination of multiple factors. The targeted gene knockout technique is a powerful tool to study the functions of these factors. The successful application of this technique in mice in the past three decades has significantly enhanced our understanding on the molecular mechanism of ovarian development. Recently, with the advent of genome editing techniques, targeted gene knockout research can be carried out in many species. Zebrafish has emerged as an excellent model system to study the control of ovarian development. Dozens of genes related to ovarian development have been knocked out in zebrafish in recent years. Much new information and perspectives on the molecular mechanism of ovarian development have been obtained from these mutant zebrafish. Some findings have challenged conventional views. Several genes have been identified for the first time in vertebrates to control ovarian development. Focusing on ovarian development, the purpose of this review is to briefly summarize recent findings using these gene knockout zebrafish models, and compare these findings with mammalian models. These established mutants and rapid development of gene knockout techniques have prompted zebrafish as an ideal animal model for studying ovarian development.
Collapse
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China, 730070.
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
17
|
Chen Z, Pan X, Kong Y, Jiang Y, Zhong Y, Zhang H, Zhang Z, Yuan X, Li J. Pituitary-Derived Circular RNAs Expression and Regulatory Network Prediction During the Onset of Puberty in Landrace × Yorkshire Crossbred Pigs. Front Genet 2020; 11:135. [PMID: 32180798 PMCID: PMC7059797 DOI: 10.3389/fgene.2020.00135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/04/2020] [Indexed: 01/25/2023] Open
Abstract
Being the center of the hypothalamus-pituitary-ovary (HPO) axis, the pituitary plays a key role in the onset of puberty. Recent studies show that circular RNAs (circRNAs) can perform as miRNA sponges to regulate development in animals. However, the function of pituitary-derived circRNAs in first estrus remains unclear in pigs. In this study, we performed a genome-wide identification and characterization of circRNAs using pituitaries from Landrace × Yorkshire crossbred pigs at three stages: pre-, in-, and post-puberty, to describe such pituitary-derived circRNAs in pigs. A total of 5148 circRNAs were found in the gilts' pituitaries, averaging 18 682 bp in genomic distance, which consisted of approximately 91% exonic, 6% intergenic, and 3% intronic circRNAs. Furthermore, 158 novel circRNAs were identified for the first time and classified as putative pituitary-specific circRNAs. Their expression levels during the onset of puberty, significantly exceeded those of the other circRNAs, and the parental genes of these putative pituitary-specific circRNAs were enriched in "ssc04917: prolactin signaling pathway," "ssc04080: neuroactive ligand-receptor interaction," and "ssc04728: dopaminergic synapse" pathways, all of which were consistent with pituitary functioning. Additionally, 17 differentially regulated circRNAs were found and investigated for their potential interaction with miRNAs, along with genes, by constructing a circRNA-targeted miRNA-gene network. Taken together, these results provide new insight into the circRNA-mediated timing of puberty in gilts at the pituitary level.
Collapse
Affiliation(s)
- Zitao Chen
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiangchun Pan
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yaru Kong
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yao Jiang
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yuyi Zhong
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hao Zhang
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhe Zhang
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaolong Yuan
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiaqi Li
- National Engineering Research Centre for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
18
|
Arao Y, Hamilton KJ, Wu SP, Tsai MJ, DeMayo FJ, Korach KS. Dysregulation of hypothalamic-pituitary estrogen receptor α-mediated signaling causes episodic LH secretion and cystic ovary. FASEB J 2019; 33:7375-7386. [PMID: 30866655 DOI: 10.1096/fj.201802653rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a hypothalamic-pituitary-gonadal (HPG) axis disorder. PCOS symptoms most likely result from a disturbance in the complex feedback regulation system of the HPG axis, which involves gonadotrophic hormones and ovarian steroid hormones. However, the nature of this complex and interconnecting feedback regulation makes it difficult to dissect the molecular mechanisms responsible for PCOS phenotypes. Global estrogen receptor α (ERα) knockout (KO) mice exhibit a disruption of the HPG axis, resulting in hormonal dysregulation in which female ERα KO mice have elevated levels of serum estradiol (E2), testosterone, and LH. The ERα KO females are anovulatory and develop cystic hemorrhagic ovaries that are thought to be due to persistently high circulating levels of LH from the pituitary. However, the role of ERα in the pituitary is still controversial because of the varied phenotypes reported in pituitary-specific ERα KO mouse models. Therefore, we developed a mouse model where ERα is reintroduced to be exclusively expressed in the pituitary on the background of a global ERα-null (PitERtgKO) mouse. Serum E2 and LH levels were normalized in PitERtgKO females and were comparable to wild-type serum levels. However, the ovaries of PitERtgKO adult mice displayed a more overt cystic and hemorrhagic phenotype when compared with ERα KO littermates. We determined that anomalous sporadic LH secretion caused the severe ovarian phenotype of PitERtgKO females. Our observations suggest that pituitary ERα is involved in the estrogen negative feedback regulation, whereas hypothalamic ERα is necessary for the precise control of LH secretion. Uncontrolled, irregular LH secretion may be the root cause of the cystic ovarian phenotype with similarities to PCOS.-Arao, Y., Hamilton, K. J., Wu, S.-P., Tsai, M.-J., DeMayo, F. J., Korach, K. S. Dysregulation of hypothalamic-pituitary estrogen receptor α-mediated signaling causes episodic LH secretion and cystic ovary.
Collapse
Affiliation(s)
- Yukitomo Arao
- Receptor Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Katherine J Hamilton
- Receptor Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - San-Pin Wu
- Pregnancy and Female Reproduction Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA; and
| | | | - Francesco J DeMayo
- Pregnancy and Female Reproduction Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA; and
| | - Kenneth S Korach
- Receptor Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
19
|
Novaira HJ, Negron AL, Graceli JB, Capellino S, Schoeffield A, Hoffman GE, Levine JE, Wolfe A, Wondisford FE, Radovick S. Impairments in the reproductive axis of female mice lacking estrogen receptor β in GnRH neurons. Am J Physiol Endocrinol Metab 2018; 315:E1019-E1033. [PMID: 30040478 PMCID: PMC6293171 DOI: 10.1152/ajpendo.00173.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/02/2018] [Accepted: 07/21/2018] [Indexed: 12/24/2022]
Abstract
The effect of estrogen on the differentiation and maintenance of reproductive tissues is mediated by two nuclear estrogen receptors (ERs), ERα, and ERβ. Lack of functional ERα and ERβ genes in vivo significantly affects reproductive function; however, the target tissues and signaling pathways in the hypothalamus are not clearly defined. Here, we describe the generation and reproductive characterization of a complete-ERβ KO (CERβKO) and a GnRH neuron-specific ERβKO (GERβKO) mouse models. Both ERβKO mouse models displayed a delay in vaginal opening and first estrus. Hypothalamic gonadotropin-releasing hormone (GnRH) mRNA expression levels in both ERβKO mice were similar to control mice; however female CERβKO and GERβKO mice had lower basal and surge serum gonadotropin levels. Although a GnRH stimulation test in both female ERβKO models showed preserved gonadotropic function in the same animals, a kisspeptin stimulation test revealed an attenuated response by GnRH neurons, suggesting a role for ERβ in normal GnRH neuron function. No alteration in estrogen-negative feedback was observed in either ERβKO mouse models after ovariectomy and estrogen replacement. Further, abnormal development of ovarian follicles with low serum estradiol levels and impairment of fertility were observed in both ERβKO mouse models. In male ERβKO mice, no differences in the timing of pubertal onset or serum luteinizing hormone and follicle-stimulating hormone levels were observed as compared with controls. Taken together, these data provide in vivo evidence for a role of ERβ in GnRH neurons in modulating puberty and reproduction, specifically through kisspeptin responsiveness in the female hypothalamic-pituitary-gonadal axis.
Collapse
Affiliation(s)
- Horacio J Novaira
- Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| | - Ariel L Negron
- Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| | - Jones B Graceli
- Department of Morphology, Federal University of Espirito Santo , Vitoria , Brazil
| | - Silvia Capellino
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, Department of Immunology , Dortmund , Germany
| | | | - Gloria E Hoffman
- Department of Biology, Morgan State University , Baltimore, Maryland
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin , Madison, Wisconsin
| | - Andrew Wolfe
- Department of Pediatrics, Division of Endocrinology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Fredric E Wondisford
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| | - Sally Radovick
- Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| |
Collapse
|
20
|
Tao Z, Song W, Zhu C, Xu W, Liu H, Zhang S, Huifang L. Comparative transcriptomic analysis of high and low egg-producing duck ovaries. Poult Sci 2018; 96:4378-4388. [PMID: 29053813 DOI: 10.3382/ps/pex229] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/14/2017] [Indexed: 01/03/2023] Open
Abstract
The egg-laying rate is an important indicator of egg production of laying ducks. Egg production directly impacts the economic benefits of the duck industry. In order to obtain better insight into the molecular mechanisms associated with the process of egg production, comparative transcriptomic analysis of the ovaries of Jinding ducks with high and low egg production was performed using the Illumina HiSeq 2500 system. A total of 843 differentially expressed genes (DEGs) was identified, 367 that were down-regulated and 476 that were up-regulated in high egg production (HEP) ovaries, as compared with low egg production (LEP) ovaries. Some genes, such as MC5R, APOD, ORAI1, and DYRK4, were more active in HEP ovaries, indicating that these genes may play important roles in regulation of egg production. Among these 843 DEGs, 685 were assigned to gene ontology (GO) categories. Of these, 25 genes were related to reproduction, and 30 were related to the reproductive process, including some associated with ovarian follicle development, circadian regulation of gene expression, circadian rhythm, and estrogen receptor binding. Furthermore, some important functional pathways were revealed, such as the steroid biosynthesis pathway, the endocrine and other factor-regulated calcium reabsorption pathways, circadian rhythm, the neuroactive ligand-receptor interaction pathway, fatty acid biosynthesis, and the calcium-signaling pathway, which appear to be much more active in the HEP group, as compared to those of the LEP group. The results of this study provide very useful information that may contribute to future functional studies of genes involved in bird reproduction.
Collapse
Affiliation(s)
- Z Tao
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| | - W Song
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| | - C Zhu
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| | - W Xu
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| | - H Liu
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| | - S Zhang
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| | - Li Huifang
- Department of waterfowl breeding and production, Jiangsu Institute of Poultry Sciences, Yangzhou, 225125, China
| |
Collapse
|
21
|
Chapple RH, Hu T, Tseng YJ, Liu L, Kitano A, Luu V, Hoegenauer KA, Iwawaki T, Li Q, Nakada D. ERα promotes murine hematopoietic regeneration through the Ire1α-mediated unfolded protein response. eLife 2018; 7:31159. [PMID: 29451493 PMCID: PMC5829925 DOI: 10.7554/elife.31159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 02/15/2018] [Indexed: 01/08/2023] Open
Abstract
Activation of the unfolded protein response (UPR) sustains protein homeostasis (proteostasis) and plays a fundamental role in tissue maintenance and longevity of organisms. Long-range control of UPR activation has been demonstrated in invertebrates, but such mechanisms in mammals remain elusive. Here, we show that the female sex hormone estrogen regulates the UPR in hematopoietic stem cells (HSCs). Estrogen treatment increases the capacity of HSCs to regenerate the hematopoietic system upon transplantation and accelerates regeneration after irradiation. We found that estrogen signals through estrogen receptor α (ERα) expressed in hematopoietic cells to activate the protective Ire1α-Xbp1 branch of the UPR. Further, ERα-mediated activation of the Ire1α-Xbp1 pathway confers HSCs with resistance against proteotoxic stress and promotes regeneration. Our findings reveal a systemic mechanism through which HSC function is augmented for hematopoietic regeneration.
Collapse
Affiliation(s)
- Richard H Chapple
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Tianyuan Hu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Yu-Jung Tseng
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| | - Lu Liu
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, United States
| | - Ayumi Kitano
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Victor Luu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Kevin A Hoegenauer
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Qing Li
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, United States
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| |
Collapse
|
22
|
Li G, Tang H, Chen Y, Yin Y, Ogawa S, Liu M, Guo Y, Qi X, Liu Y, Parhar IS, Liu X, Lin H. Estrogen directly stimulates LHb expression at the pituitary level during puberty in female zebrafish. Mol Cell Endocrinol 2018; 461:1-11. [PMID: 28801227 DOI: 10.1016/j.mce.2017.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/05/2017] [Accepted: 08/06/2017] [Indexed: 11/16/2022]
Abstract
The LHb expression is up-regulated during puberty in female zebrafish. However, the molecular mechanism underlying how LHb expression is regulated during puberty remains largely unknown. In this study, we found that the mRNA expression levels of lhb, fshb and cyp19a1b were up-regulated along with the puberty onset in zebrafish. Among the three nuclear estrogen receptors (nERs), the esr2b is the only type whose expression is significantly up-regulated during puberty onset in the pituitary. However, in situ hybridization results revealed that lhb mRNA was colocalized with esr1 and esr2a but not esr2b. Exposure to estradiol (E2) significantly stimulates LHb expression in both wild-type and kiss1-/-;kiss2-/-;gnrh3-/- triple knockout pubertal zebrafish. Moreover, exposure of cultured pituitary cells to E2 increased the LHb expression, indicating that the estrogenic effect on LHb expression could be acted at the pituitary level. Finally, we cloned and analyzed the promoter of lhb by luciferase assay. Our results indicated that the E2 responsive regions of lhb promoter for ERα and ERβ2 are identical, suggesting that ERα and ERβ2 could bind to the same half ERE region of the promoter of lhb, exhibiting a classical ERE-dependent pathway. In summary, we demonstrate that E2 could directly act on the pituitary level to stimulate LHb transcription during puberty in zebrafish.
Collapse
Affiliation(s)
- Gaofei Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Haipei Tang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu Chen
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yike Yin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Satoshi Ogawa
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Meifeng Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yin Guo
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xin Qi
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Ishwar S Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| |
Collapse
|
23
|
Chen Y, Tang H, Wang L, He J, Guo Y, Liu Y, Liu X, Lin H. Fertility Enhancement but Premature Ovarian Failure in esr1-Deficient Female Zebrafish. Front Endocrinol (Lausanne) 2018; 9:567. [PMID: 30319547 PMCID: PMC6165881 DOI: 10.3389/fendo.2018.00567] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/05/2018] [Indexed: 12/12/2022] Open
Abstract
It is well established that estrogens regulate female reproduction through estrogen receptors (ERs) in the ovary. However, the precise physiological role of estrogen/ER signaling in reproduction processes remains poorly defined in zebrafish. In this study, we successfully generated an ERα (esr1) mutant line in zebrafish via transcription activator-like effectors nucleases (TALENs). It was found in the mutant females that the fertility was enhanced and the ovarian histology was normal at 90 days post-fertilization (dpf). However, the number of fertile females decreased with age. By 180 dpf, esr1 mutant females were infertile with degenerated ovaries, while the age-matched wild-type females were still fertile. Additionally, few large vitellogenic granules can be found in full grown (FG) follicles at 90 dpf and the expression of vtg genes were down-regulated at both 90 and 180 dpf in esr1 mutant zebrafish. Moreover, steroidogenesis pathway and mTOR signaling pathway were over-activated at 90 dpf, but declined prematurely in esr1 mutant zebrafish by 180 dpf. Collectively, the present study provides evidence that esr1 is fundamental for ovarian maintenance in zebrafish.
Collapse
|
24
|
Qin X, Xiao Y, Ye C, Jia J, Liu X, Liang H, Zou G, Hu G. Pituitary Action of E2 in Prepubertal Grass Carp: Receptor Specificity and Signal Transduction for Luteinizing Hormone and Follicle-Stimulating Hormone Regulation. Front Endocrinol (Lausanne) 2018; 9:308. [PMID: 29937753 PMCID: PMC6002485 DOI: 10.3389/fendo.2018.00308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/23/2018] [Indexed: 12/29/2022] Open
Abstract
17β-estradiol (E2) is an important sex steroid produced by ovary and brain. In mammals, E2 plays an important role in hypothalamus-pituitary-gonad axis to regulate puberty onset, however, little is known about the functional role of E2 in teleost pituitary. Using prepubertal grass carp as model, three nuclear estrogen receptors (nERs: estrogen receptor alpha, estrogen receptor beta 1, and estrogen receptor beta 2) and two G protein-coupled estrogen receptors (GPER1: GPER1a and GPER1b) were isolated from grass carp pituitary. Tissue distribution analysis indicated that both nERs and GPERs were highly detected in grass carp pituitary, which suggested that E2 should play an important role in grass carp pituitary. Using primary cultured grass carp pituitary cells as model, high-throughput RNA-seq was used to examine the E2-induced differentially expressed genes (DEGs). Transcriptomic analysis showed that E2 could significantly upregulate the expression of 28 genes in grass carp pituitary cells, which were characterized into different functions including reproduction, gonad development, and central nervous system development. Further studies confirmed that E2 could induce luteinizing hormone (LH) and follicle-stimulating hormone (FSH) secretion and mRNA expression in prepubertal grass carp pituitary in vivo and in vitro. In the pituitary, LH and FSH regulation by E2 were mediated by both ERβ and GPER1. Apparently, E2-induced LHβ and FSHβ mRNA expression were mediated by adenylyl cyclase/cAMP/protein kinase A, phospholipase C/inositol 1,4,5-triphosphate/protein kinase C, and Ca2+/calmodulin/CaM-dependent protein kinase II pathways. In addition to LH and FSH, E2 could also induce growth regulation by estrogen in breast cancer 1 (a novel regulator for pituitary development) mRNA expression in grass carp pituitary cells. These results, as a whole, suggested that E2 could play an important role in gonadotropin hormone release and pituitary development in prepubertal grass carp.
Collapse
Affiliation(s)
- Xiangfeng Qin
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Yaqian Xiao
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Cheng Ye
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jingyi Jia
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Xiangjiang Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Hongwei Liang
- Key Laboratory of Freshwater Biodiversity Conservation Ministry of Agriculture, Yangtze River Fisheries Research Institute, The Chinese Academy of Fishery Sciences, Wuhan, China
| | - Guiwei Zou
- Key Laboratory of Freshwater Biodiversity Conservation Ministry of Agriculture, Yangtze River Fisheries Research Institute, The Chinese Academy of Fishery Sciences, Wuhan, China
| | - Guangfu Hu
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Guangfu Hu,
| |
Collapse
|
25
|
Cho SH, Choi Y, Kim SH, Kim SJ, Chang J. Urinary bisphenol A versus serum bisphenol A concentration and ovarian reproductive outcomes among IVF patients: Which is a better biomarker of BPA exposure? Mol Cell Toxicol 2017. [DOI: 10.1007/s13273-017-0039-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
26
|
Park CJ, Chen G, Koo Y, Lin PCP, Cacioppo JA, Prohaska H, Ko CJ. Generation and characterization of an estrogen receptor alpha-iCre knock-in mouse. Genesis 2017; 55. [PMID: 29115049 DOI: 10.1002/dvg.23084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 01/11/2023]
Abstract
Two estrogen receptors, ESR1 and ESR2, are responsible for the classical actions of estrogens in mammalian species. They display different spatiotemporal expression patterns and nonoverlapping functions in various tissues and physiological conditions. In this study, a novel knock-in mouse line that expresses codon-improved Cre recombinase (iCre) under regulation of the natural Esr1 promoter (Esr1-iCre) was developed. Functional characterization of iCre expression by crossing them with reporter lines (ROSA26-lacZ or Ai9-RFP) showed that iCre is faithfully expressed in Esr1-lineage cells. This novel transgenic mouse line will be a useful animal model for lineage-tracing Esr1-expressing cells, selective gene ablation in the Esr1-lineage cells and for generating global Esr1 knockout mice.
Collapse
Affiliation(s)
- Chan Jin Park
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Guanglin Chen
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Yongbum Koo
- School of Biological Sciences, Inje University, Gimhae, South Korea
| | - Po-Ching P Lin
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Joseph A Cacioppo
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Hailey Prohaska
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - CheMyong J Ko
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| |
Collapse
|
27
|
E6/E7 oncogenes in epithelial suprabasal layers and estradiol promote cervical growth and ear regeneration. Oncogenesis 2017; 6:e374. [PMID: 28846079 PMCID: PMC5608921 DOI: 10.1038/oncsis.2017.73] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 06/10/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023] Open
Abstract
Tissue growth is a common characteristic of carcinogenesis and regeneration. Here we show that suprabasal expression of human papillomavirus (HPV)16 E6/E7 oncogenes in Tg(K6b-E6/E7) mice, similar to that observed in HPV-infected human tissue, and estradiol increased cervical epithelium growth and ear-hole closure efficiency. Oncogenes in combination with estradiol had a significant contribution to the proliferation of suprabasal cells of cervical epithelium that correlated with an increased expression of keratin genes. Remarkably, long-term treatments with estradiol resulted in evident cellular and tissue abnormalities indicative of a precancerous phenotype. Regenerating ear epithelium of transgenic mice also showed increased suprabasal cell proliferation and expression of keratin genes. Unexpectedly, we observed higher ear regeneration efficiency in adult than in young female mice, which was further increased by E6/E7 oncogenes. Supporting a role of estradiol in this phenomenon, ovariectomy and treatment with an estrogen receptor inhibitor caused a significant reduction in regenerative capacity. Our data suggest that Tg(K6b-E6/E7) mice are unique to mimic the initial stages of HPV-mediated cervical carcinogenesis, and ear regeneration could facilitate the elucidation of mechanisms involved.
Collapse
|
28
|
Maguire CA, Song YB, Wu M, León S, Carroll RS, Alreja M, Kaiser UB, Navarro VM. Tac1 Signaling Is Required for Sexual Maturation and Responsiveness of GnRH Neurons to Kisspeptin in the Male Mouse. Endocrinology 2017; 158:2319-2329. [PMID: 28444173 PMCID: PMC5505212 DOI: 10.1210/en.2016-1807] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/18/2017] [Indexed: 11/19/2022]
Abstract
The tachykinins substance P (SP) and neurokinin A (Tac1) have emerged as novel regulators of kisspeptin/GnRH release. Recently, we documented that SP modulates reproductive function in the female mouse. Here, we extended this characterization to the male mouse. Tac1-/- male mice showed delayed puberty onset. They also presented significantly decreased expression levels of Pdyn (dynorphin) and Nos1 (nitric oxide synthase) in the mediobasal hypothalamus and elevated Gnrh1 levels. Unexpectedly, the response of Tac1-/- mice to central kisspeptin or senktide (neurokinin B receptor-agonist) administration was significantly decreased compared with controls, despite the preserved ability of GnRH neurons to stimulate luteinizing hormone release as demonstrated by central N-methyl-D-aspartate receptor administration, suggesting a deficit at the GnRH neuron level. Importantly, we demonstrated that kisspeptin receptor and SP receptor (NK1R) heterodimerize, indicating that changes in the SP tone could alter the responsiveness of GnRH neurons to kisspeptin. Finally, electrophysiological recordings from arcuate Kiss1 neurons showed that, although virtually all Kiss1 neurons responded to NKB and senktide, only half responded to an NK1R agonist and none to the neurokinin A receptor agonist at a 1-μM dose. In summary, we provide compelling evidence for a role of Tac1 in the control of reproductive function in the male mouse, suggesting a predominant central action that may involve a change in the balance of neural factors that control GnRH expression.
Collapse
Affiliation(s)
- Caroline A. Maguire
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Yong Bhum Song
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Min Wu
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06508
| | - Silvia León
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Rona S. Carroll
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Meenakshi Alreja
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06508
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Víctor M. Navarro
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
29
|
Qiu S, Vazquez JT, Boulger E, Liu H, Xue P, Hussain MA, Wolfe A. Hepatic estrogen receptor α is critical for regulation of gluconeogenesis and lipid metabolism in males. Sci Rep 2017; 7:1661. [PMID: 28490809 PMCID: PMC5431852 DOI: 10.1038/s41598-017-01937-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/06/2017] [Indexed: 12/19/2022] Open
Abstract
Impaired estrogens action is associated with features of the metabolic syndrome in animal models and humans. We sought to determine whether disruption of hepatic estrogens action in adult male mice could recapitulate aspects of the metabolic syndrome to understand the mechanistic basis for the phenotype. We found 17β-estradiol (E2) inhibited hepatic gluconeogenic genes such as phosphoenolpyruvate carboxykinase 1 (Pck-1) and glucose 6-phosphatase (G6Pase) and this effect was absent in mice lacking liver estrogen receptor α (Esr1) (LERKO mice). Male LERKO mice displayed elevated hepatic gluconeogenic activity and fasting hyperglycemia. We also observed increased liver lipid deposits and triglyceride levels in male LERKO mice, resulting from increased hepatic lipogenesis as reflected by increased mRNA levels of fatty acid synthase (Fas) and acetyl-CoA carboxylase (Acc1). ChIP assay demonstrated estradiol (E2) induced ESR1 binding to Pck-1, G6Pase, Fas and Acc1 promoters. Metabolic phenotyping demonstrated both basal metabolic rate and feeding were lower for the LERKO mice as compared to Controls. Furthermore, the respiratory exchange rate was significantly lower in LERKO mice than in Controls, suggesting an increase in lipid oxidation. Our data indicate that hepatic E2/ESR1 signaling plays a key role in the maintenance of gluconeogenesis and lipid metabolism in males.
Collapse
Affiliation(s)
- Shuiqing Qiu
- Division of Metabolism and Pediatric Endocrinology, Departments of Medicine, Pediatrics, Biological Chemistry and Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Erin Boulger
- School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ping Xue
- Division of Metabolism and Pediatric Endocrinology, Departments of Medicine, Pediatrics, Biological Chemistry and Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mehboob Ali Hussain
- Division of Metabolism and Pediatric Endocrinology, Departments of Medicine, Pediatrics, Biological Chemistry and Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew Wolfe
- Division of Metabolism and Pediatric Endocrinology, Departments of Medicine, Pediatrics, Biological Chemistry and Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
30
|
Stallings CE, Kapali J, Ellsworth BS. Mouse Models of Gonadotrope Development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:1-48. [PMID: 27697200 DOI: 10.1016/bs.pmbts.2016.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotrope is central to reproductive function. Gonadotropes develop in a systematic process dependent on signaling factors secreted from surrounding tissues and those produced within the pituitary gland itself. These signaling pathways are important for stimulating specific transcription factors that ultimately regulate the expression of genes and define gonadotrope identity. Proper gonadotrope development and ultimately gonadotrope function are essential for normal sexual maturation and fertility. Understanding the mechanisms governing differentiation programs of gonadotropes is important to improve treatment and molecular diagnoses for patients with gonadotrope abnormalities. Much of what is known about gonadotrope development has been elucidated from mouse models in which important factors contributing to gonadotrope development and function have been deleted, ectopically expressed, or modified. This chapter will focus on many of these mouse models and their contribution to our current understanding of gonadotrope development.
Collapse
Affiliation(s)
- C E Stallings
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - J Kapali
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - B S Ellsworth
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States.
| |
Collapse
|
31
|
Zhang R, Hu Y, Wang H, Yan P, Zhou Y, Wu R, Wu X. Molecular cloning, characterization, tissue distribution and mRNA expression changes during the hibernation and reproductive periods of estrogen receptor alpha (ESR1) in Chinese alligator, Alligator sinensis. Comp Biochem Physiol B Biochem Mol Biol 2016; 200:28-35. [PMID: 27212643 DOI: 10.1016/j.cbpb.2016.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023]
Abstract
Chinese alligator, Alligator sinensis, is a critically endangered reptile species unique to China. Little is known about the mechanism of growth- and reproduction-related hormones gene expression in Chinese alligator. Estrogens play important roles in regulating multiple reproduction- and non-reproduction-related functions by binding to their corresponding receptors. Here, the full-length cDNA of estrogen receptor alpha (ERα/ESR1) was cloned and sequenced from Chinese alligator for the first time, which comprises 1764bp nucleotides and encodes a predicted protein of 587 amino acids. Phylogenetic analysis of ESR1 showed that crocodilians and turtles were the sister-group of birds. The results of real-time quantitative PCR indicated that the ESR1 mRNA was widely expressed in the brain and peripheral tissues. In the brain and pituitary gland, ESR1 was most highly transcribed in the cerebellum. But in other peripheral tissues, ESR1 mRNA expression level was the highest in the ovary. Compared with hibernation period, ESR1 mRNA expression levels were increased significantly in the reproductive period (P<0.05) in cerebellum, pituitary gland, liver, spleen, lung, kidney and ovary, while no significant change in other examined tissues (P>0.05). The ESR1 mRNA expression levels changes during the two periods of different tissues suggested that ESR1 might play an important role in mediation of estrogenic multiple reproductive effects in Chinese alligator. Furthermore, it was the first time to quantify ESR1 mRNA level in the brain of crocodilians, and the distribution and expression of ESR1 mRNA in the midbrain, cerebellum and medulla oblongata was also reported for the first time in reptiles.
Collapse
Affiliation(s)
- Ruidong Zhang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Yuehong Hu
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Huan Wang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Peng Yan
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Yongkang Zhou
- Alligator Research Center of Anhui Province, Xuanzhou 242000, People's Republic of China
| | - Rong Wu
- Alligator Research Center of Anhui Province, Xuanzhou 242000, People's Republic of China
| | - Xiaobing Wu
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China.
| |
Collapse
|
32
|
Dubois SL, Wolfe A, Radovick S, Boehm U, Levine JE. Estradiol Restrains Prepubertal Gonadotropin Secretion in Female Mice via Activation of ERα in Kisspeptin Neurons. Endocrinology 2016; 157:1546-54. [PMID: 26824364 PMCID: PMC4816723 DOI: 10.1210/en.2015-1923] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/15/2016] [Indexed: 11/19/2022]
Abstract
Elimination of estrogen receptorα (ERα) from kisspeptin (Kiss1) neurons results in premature LH release and pubertal onset, implicating these receptors in 17β-estradiol (E2)-mediated negative feedback regulation of GnRH release during the prepubertal period. Here, we tested the dependency of prepubertal negative feedback on ERα in Kiss1 neurons. Prepubertal (postnatal d 14) and peripubertal (postnatal d 34) wild-type (WT) and Kiss1 cell-specific ERα knockout (KERαKO) female mice were sham operated or ovariectomized and treated with either vehicle- or E2-containing capsules. Plasma and tissues were collected 2 days after surgery for analysis. Ovariectomy increased LH and FSH levels, and E2 treatments completely prevented these increases in WT mice of both ages. However, in prepubertal KERαKO mice, basal LH levels were elevated vs WT, and both LH and FSH levels were not further increased by ovariectomy or affected by E2 treatment. Similarly, Kiss1 mRNA levels in the medial basal hypothalamus, which includes the arcuate nucleus, were suppressed with E2 treatment in ovariectomized prepubertal WT mice but remained unaffected by any treatment in KERαKO mice. In peripubertal KERαKO mice, basal LH and FSH levels were not elevated vs WT and were unaffected by ovariectomy or E2. In contrast to our previous findings in adult animals, these results demonstrate that suppression of gonadotropins and Kiss1 mRNA by E2 in prepubertal animals depends upon ERα activation in Kiss1 neurons. Our observations are consistent with the hypothesis that these receptors play a critical role in restraining GnRH release before the onset and completion of puberty.
Collapse
Affiliation(s)
- Sharon L Dubois
- Neuroscience Training Program (S.L.D.) and Department of Neuroscience (S.L.D., J.E.L.), University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Pediatrics (A.W., S.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Pharmacology and Toxicology (U.B.), University of Saarland School of Medicine, Homburg D-66421, Germany; and Wisconsin National Primate Research Center (J.E.L.), Madison, Wisconsin 53715
| | - Andrew Wolfe
- Neuroscience Training Program (S.L.D.) and Department of Neuroscience (S.L.D., J.E.L.), University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Pediatrics (A.W., S.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Pharmacology and Toxicology (U.B.), University of Saarland School of Medicine, Homburg D-66421, Germany; and Wisconsin National Primate Research Center (J.E.L.), Madison, Wisconsin 53715
| | - Sally Radovick
- Neuroscience Training Program (S.L.D.) and Department of Neuroscience (S.L.D., J.E.L.), University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Pediatrics (A.W., S.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Pharmacology and Toxicology (U.B.), University of Saarland School of Medicine, Homburg D-66421, Germany; and Wisconsin National Primate Research Center (J.E.L.), Madison, Wisconsin 53715
| | - Ulrich Boehm
- Neuroscience Training Program (S.L.D.) and Department of Neuroscience (S.L.D., J.E.L.), University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Pediatrics (A.W., S.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Pharmacology and Toxicology (U.B.), University of Saarland School of Medicine, Homburg D-66421, Germany; and Wisconsin National Primate Research Center (J.E.L.), Madison, Wisconsin 53715
| | - Jon E Levine
- Neuroscience Training Program (S.L.D.) and Department of Neuroscience (S.L.D., J.E.L.), University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Pediatrics (A.W., S.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; Department of Pharmacology and Toxicology (U.B.), University of Saarland School of Medicine, Homburg D-66421, Germany; and Wisconsin National Primate Research Center (J.E.L.), Madison, Wisconsin 53715
| |
Collapse
|
33
|
Kadam KM, Mande PV, Gawas N, Ahire S, Khole LVV. Autoantibodies to Heat-Shock Protein, HSPA5, and Epitope Spreading: High-Dose Dexamethasone Therapy Rescues Ovarian Function in Experimental Autoimmune Ovarian Insufficiency Mouse Model. Am J Reprod Immunol 2016; 75:580-93. [PMID: 26840828 DOI: 10.1111/aji.12494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 01/05/2016] [Indexed: 11/27/2022] Open
Abstract
PROBLEM Role of autoantibodies to heat-shock protein 70 isoform, HSPA5, both alone or in combination with other antigenic peptides in epitope spreading and effect of high-dose dexamethasone to overcome this. METHOD OF STUDY Experimental autoimmune premature ovarian insufficiency mouse model generated by immunization with immunodominant epitopes of HSPA5 alone or in combination with other antigenic peptides. Two doses of dexamethasone treatment are given to the latter group. Immunosorbent assay and Western blot analysis were undertaken to detect cross-reactivity. Hormonal estimations, histological evaluation, and fertility studies were performed to assess treatment efficacy. RESULTS One of the immunodominant epitopes of HSPA5 led to epitope spreading. Of the two doses, 100 mg was more effective in rescuing fertility. CONCLUSIONS We postulate that the shared immunodominant peptide could be included in a peptide array to detect both HSAP5 and HSP90β autoantibodies for early diagnosis or prognosis of aPOI and customized glucocorticoid therapy for such subjects.
Collapse
Affiliation(s)
- Kaushiki M Kadam
- Department of Gamete Immunobiology, National Institute for Research in Reproductive Health, Mumbai, India
| | - Purvi V Mande
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Nilesh Gawas
- Department of Gamete Immunobiology, National Institute for Research in Reproductive Health, Mumbai, India
| | - Sarika Ahire
- Department of Gamete Immunobiology, National Institute for Research in Reproductive Health, Mumbai, India
| | - Late Vrinda V Khole
- Department of Gamete Immunobiology, National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
34
|
GnRH Neuron-Specific Ablation of Gαq/11 Results in Only Partial Inactivation of the Neuroendocrine-Reproductive Axis in Both Male and Female Mice: In Vivo Evidence for Kiss1r-Coupled Gαq/11-Independent GnRH Secretion. J Neurosci 2016; 35:12903-16. [PMID: 26377475 DOI: 10.1523/jneurosci.0041-15.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility and kisspeptin (KP) is a potent trigger of GnRH secretion from GnRH neurons. KP signals via KISS1R, a Gαq/11-coupled receptor, and mice bearing a global deletion of Kiss1r (Kiss1r(-/-)) or a GnRH neuron-specific deletion of Kiss1r (Kiss1r(d/d)) display hypogonadotropic hypogonadism and infertility. KISS1R also signals via β-arrestin, and in mice lacking β-arrestin-1 or -2, KP-triggered GnRH secretion is significantly diminished. Based on these findings, we hypothesized that ablation of Gαq/11 in GnRH neurons would diminish but not completely block KP-triggered GnRH secretion and that Gαq/11-independent GnRH secretion would be sufficient to maintain fertility. To test this, Gnaq (encodes Gαq) was selectively inactivated in the GnRH neurons of global Gna11 (encodes Gα11)-null mice by crossing Gnrh-Cre and Gnaq(fl/fl);Gna11(-/-) mice. Experimental Gnaq(fl/fl);Gna11(-/-);Gnrh-Cre (Gnaq(d/d)) and control Gnaq(fl/fl);Gna11(-/-) (Gnaq(fl/fl)) littermate mice were generated and subjected to reproductive profiling. This process revealed that testicular development and spermatogenesis, preputial separation, and anogenital distance in males and day of vaginal opening and of first estrus in females were significantly less affected in Gnaq(d/d) mice than in previously characterized Kiss1r(-/-) or Kiss1r(d/d) mice. Additionally, Gnaq(d/d) males were subfertile, and although Gnaq(d/d) females did not ovulate spontaneously, they responded efficiently to a single dose of gonadotropins. Finally, KP stimulation triggered a significant increase in gonadotropins and testosterone levels in Gnaq(d/d) mice. We therefore conclude that the milder reproductive phenotypes and maintained responsiveness to KP and gonadotropins reflect Gαq/11-independent GnRH secretion and activation of the neuroendocrine-reproductive axis in Gnaq(d/d) mice. SIGNIFICANCE STATEMENT The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility. Over the last decade, several studies have established that the KISS1 receptor, KISS1R, is a potent trigger of GnRH secretion and inactivation of KISS1R on the GnRH neuron results in infertility. While KISS1R is best understood as a Gαq/11-coupled receptor, we previously demonstrated that it could couple to and signal via non-Gαq/11-coupled pathways. The present study confirms these findings and, more importantly, while it establishes Gαq/11-coupled signaling as a major conduit of GnRH secretion, it also uncovers a significant role for non-Gαq/11-coupled signaling in potentiating reproductive development and function. This study further suggests that by augmenting signaling via these pathways, GnRH secretion can be enhanced to treat some forms of infertility.
Collapse
|
35
|
Abstract
Tissue specific knockout mice are valuable tools to study gene function in vivo. The method uses the Cre/loxP system in which loxP sites are cloned into the genome surrounding one or more exons of a gene and the targeted exon(s) are deleted when the Cre enzyme is expressed. Mouse lines that are prepared for the generation of knockout ERα mice have been developed independently by many research groups and the number of available transgenic mouse lines that express Cre under tissue specific promoters is large. Here, we describe how tissue specific ERα knockout mice are generated.
Collapse
|
36
|
Al-Safi ZA, Liu H, Carlson NE, Chosich J, Lesh J, Robledo C, Bradford AP, Gee NA, Phang T, Santoro N, Kohrt W, Polotsky AJ. Estradiol Priming Improves Gonadotrope Sensitivity and Pro-Inflammatory Cytokines in Obese Women. J Clin Endocrinol Metab 2015; 100:4372-81. [PMID: 26425884 PMCID: PMC4702462 DOI: 10.1210/jc.2015-1946] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Obesity is associated with a pro-inflammatory state and relative hypogonadotropic hypogonadism. Estrogen (E2) is a potential link between these phenomena because it exhibits negative feedback on gonadotropin secretion and also inhibits production of pro-inflammatory cytokines. OBJECTIVE We sought to examine the effect of estrogen priming on the hypothalamic-pituitary-ovarian axis in obesity. DESIGN, SETTING, AND PARTICIPANTS This was an interventional study at an academic center of 11 obese and 10 normal-weight (NW) women. INTERVENTION A frequent blood-sampling study and one month of daily urinary collection were performed before and after administration of transdermal estradiol 0.1 mg/d for one entire menstrual cycle. MAIN OUTCOME MEASURES Serum LH and FSH before and after GnRH stimulation, and urinary estrogen and progesterone metabolites were measured. RESULTS E2 increased LH pulse amplitude and FSH response to GnRH (P = .048, and P < .03, respectively) in obese but not NW women. After E2 priming, ovulatory obese but not NW women had a 25% increase in luteal progesterone (P = .01). Obese women had significantly higher baseline IL-6, IL-10, TGF-β, and IL-12 compared with NW (all P < .05); these levels were reduced after E2 (-6% for IL-1β, -21% for IL-8, -5% for TGF-β, -5% for IL-12; all P < .05) in obese but not in NW women. CONCLUSIONS E2 priming seems to improve hypothalamic-pituitary-ovarian axis function and systemic inflammation in ovulatory, obese women. Reducing chronic inflammation at the pituitary level may decrease the burden of obesity on fertility.
Collapse
Affiliation(s)
- Zain A Al-Safi
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Huayu Liu
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nichole E Carlson
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Justin Chosich
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Jennifer Lesh
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Celeste Robledo
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Andrew P Bradford
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nancy A Gee
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Tzu Phang
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nanette Santoro
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Wendy Kohrt
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Alex J Polotsky
- Department of Obstetrics and Gynecology (Z.A.A., J.C., J.L., C.R., A.P.B., N.S., W.K., A.J.P.), University of Colorado School of Medicine, Aurora, Colorado 80045; Department of Biostatistics and Informatics (H.L., N.E.C., T.P.), Colorado School of Public Health, Aurora, Colorado 80045; Center for Health and the Environment and California National Primate Research Center (N.A.G.), University of California, Davis, California 95616; and Department of Medicine (W.K.), University of Colorado School of Medicine, Aurora, Colorado 80045
| |
Collapse
|
37
|
O’Hara L, Curley M, Tedim Ferreira M, Cruickshanks L, Milne L, Smith LB. Pituitary androgen receptor signalling regulates prolactin but not gonadotrophins in the male mouse. PLoS One 2015; 10:e0121657. [PMID: 25799562 PMCID: PMC4370825 DOI: 10.1371/journal.pone.0121657] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 02/11/2015] [Indexed: 12/02/2022] Open
Abstract
Production of the androgen testosterone is controlled by a negative feedback loop within the hypothalamic-pituitary-gonadal (HPG) axis. Stimulation of testicular Leydig cells by pituitary luteinising hormone (LH) is under the control of hypothalamic gonadotrophin releasing hormone (GnRH), while suppression of LH secretion by the pituitary is controlled by circulating testosterone. Exactly how androgens exert their feedback control of gonadotrophin secretion (and whether this is at the level of the pituitary), as well as the role of AR in other pituitary cell types remains unclear. To investigate these questions, we exploited a transgenic mouse line (Foxg1Cre/+; ARfl/y) which lacks androgen receptor in the pituitary gland. Both circulating testosterone and gonadotrophins are unchanged in adulthood, demonstrating that AR signalling is dispensable in the male mouse pituitary for testosterone-dependent regulation of LH secretion. In contrast, Foxg1Cre/+; ARfl/y males have a significant increase in circulating prolactin, suggesting that, rather than controlling gonadotrophins, AR-signalling in the pituitary acts to suppress aberrant prolactin production in males.
Collapse
Affiliation(s)
- Laura O’Hara
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Curley
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Maria Tedim Ferreira
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Lyndsey Cruickshanks
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Laura Milne
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Lee B. Smith
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Navarro VM, Bosch MA, León S, Simavli S, True C, Pinilla L, Carroll RS, Seminara SB, Tena-Sempere M, Rønnekleiv OK, Kaiser UB. The integrated hypothalamic tachykinin-kisspeptin system as a central coordinator for reproduction. Endocrinology 2015; 156:627-37. [PMID: 25422875 PMCID: PMC4298326 DOI: 10.1210/en.2014-1651] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tachykinins are comprised of the family of related peptides, substance P (SP), neurokinin A (NKA), and neurokinin B (NKB). NKB has emerged as regulator of kisspeptin release in the arcuate nucleus (ARC), whereas the roles of SP and NKA in reproduction remain unknown. This work explores the roles of SP and NKA in the central regulation of GnRH release. First, central infusion of specific agonists for the receptors of SP (neurokinin receptor 1, NK1R), NKA (NK2R) and NKB (NK3R) each induced gonadotropin release in adult male and ovariectomized, estradiol-replaced female mice, which was absent in Kiss1r(-/-) mice, indicating a kisspeptin-dependent action. The NK2R agonist, however, decreased LH release in ovariectomized-sham replaced females, as documented for NK3R agonists but in contrast to the NK1R agonist, which further increased LH release. Second, Tac1 (encoding SP and NKA) expression in the ARC and ventromedial nucleus was inhibited by circulating estradiol but did not colocalize with Kiss1 mRNA. Third, about half of isolated ARC Kiss1 neurons expressed Tacr1 (NK1R) and 100% Tacr3 (NK3R); for anteroventral-periventricular Kiss1 neurons and GnRH neurons, approximately one-fourth expressed Tacr1 and one-tenth Tacr3; Tacr2 (NK2R) expression was absent in all cases. Overall, these results identify a potent regulation of gonadotropin release by the SP/NK1R and NKA/NK2R systems in the presence of kisspeptin-Kiss1r signaling, indicating that they may, along with NKB/NK3R, control GnRH release, at least in part through actions on Kiss1 neurons.
Collapse
Affiliation(s)
- Víctor M Navarro
- Division of Endocrinology, Diabetes, and Hypertension (V.M.N., S.S., R.S.C., U.B.K.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115; Department of Physiology and Pharmacology (M.A.B., O.K.R.), Oregon Health and Science University, Portland, Oregon 97239; Department of Cell Biology, Physiology, and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (S.L., L.P., M.T.-S.), Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas and Hospital Universitario Reina Sofia (S.L., L.P., M.T.-S.), 14004 Córdoba, Spain; Department of Obstetrics and Gynecology (S.S.), Pamukkale University School of Medicine, Denizli, 20020 Turkey; and Massachusetts General Hospital and Harvard Medical School (C.T., S.B.S.), Boston, Massachusetts 02114
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Simonneaux V, Bahougne T. A Multi-Oscillatory Circadian System Times Female Reproduction. Front Endocrinol (Lausanne) 2015; 6:157. [PMID: 26539161 PMCID: PMC4611855 DOI: 10.3389/fendo.2015.00157] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/21/2015] [Indexed: 01/14/2023] Open
Abstract
Rhythms in female reproduction are critical to insure that timing of ovulation coincides with oocyte maturation and optimal sexual arousal. This fine tuning of female reproduction involves both the estradiol feedback as an indicator of oocyte maturation, and the master circadian clock of the suprachiasmatic nuclei (SCN) as an indicator of the time of the day. Herein, we are providing an overview of the state of knowledge regarding the differential inhibitory and stimulatory effects of estradiol at different stages of the reproductive axis, and the mechanisms through which the two main neurotransmitters of the SCN, arginine vasopressin, and vasoactive intestinal peptide, convey daily time cues to the reproductive axis. In addition, we will report the most recent findings on the putative functions of peripheral clocks located throughout the reproductive axis [kisspeptin (Kp) neurons, gonadotropin-releasing hormone neurons, gonadotropic cells, the ovary, and the uterus]. This review will point to the critical position of the Kp neurons of the anteroventral periventricular nucleus, which integrate both the stimulatory estradiol signal, and the daily arginine vasopressinergic signal, while displaying a circadian clock. Finally, given the critical role of the light/dark cycle in the synchronization of female reproduction, we will discuss the impact of circadian disruptions observed during shift-work conditions on female reproductive performance and fertility in both animal model and humans.
Collapse
Affiliation(s)
- Valérie Simonneaux
- Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), Strasbourg, France
- *Correspondence: Valérie Simonneaux, Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), 5 rue Blaise Pascal, Strasbourg 67084, France,
| | - Thibault Bahougne
- Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), Strasbourg, France
- Service d’Endocrinologie et Diabète, Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
40
|
Zhang S, Zhang Y, Chen W, Wu Y, Ge W, Zhang L, Zhang W. Aromatase (Cyp19a1b) in the pituitary is dynamically involved in the upregulation of lhb but not fshb in the vitellogenic female ricefield eel Monopterus albus. Endocrinology 2014; 155:4531-41. [PMID: 25105781 DOI: 10.1210/en.2014-1069] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aromatase, encoded by Cyp19a1, is expressed in the pituitary of vertebrates; however, its physiological relevance remains poorly defined. In teleosts, the duplicated cyp19a1b is preferentially expressed in the pituitary where LH and FSH are synthesized in distinct gonadotropes. Our present study demonstrated that Cyp19a1b is colocalized with Lhb, but not Fshb, during vitellogenesis in female ricefield eels. The immunoreactive levels of Cyp19a1b and Lhb, as well as their colocalization frequency, increased during vitellogenesis toward maturation. The expression of lhb but not fshb in the pituitary fragments of female ricefield eels was induced by both estradiol (E2) and testosterone (T). In agreement, the promoter of lhb but not fshb was activated by both E2 and T. T is more potent than E2 in inducing lhb expression, whereas E2 is much more effective in activating the lhb promoter. T-induced lhb expression in the pituitary fragments was abolished by the estrogen receptor (Esr) antagonist fulvestrant and suppressed by the aromatase inhibitor letrozole, suggesting that the effect of T on lhb expression at the pituitary is largely mediated by E2. Furthermore, Lhb was shown to colocalize with Esr1 but not Esr2a. Taken together, results of the present study suggest that Cyp19a1b in LH cells may greatly upregulate lhb expression during vitellogenesis, possibly via E2 and Esr1 in an intracrine manner. The absence of Cyp19a1b in FSH cells and the insensitivity of fshb to sex steroids may contribute to the differential expression of lhb and fshb in ricefield eels and possibly other vertebrates as well.
Collapse
Affiliation(s)
- Shen Zhang
- School of Life Sciences (S.Z., Y.Z., W.C., Y.W., L.Z., W.Z.), Sun Yat-sen University, Guangzhou 510275, People's Republic of China; and Faculty of Heath Sciences (W.G.), University of Macau, Taipa, Macau Special Administrative Region (SAR), China, and School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Wolfe A, Divall S, Wu S. The regulation of reproductive neuroendocrine function by insulin and insulin-like growth factor-1 (IGF-1). Front Neuroendocrinol 2014; 35:558-72. [PMID: 24929098 PMCID: PMC4175134 DOI: 10.1016/j.yfrne.2014.05.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/01/2014] [Accepted: 05/27/2014] [Indexed: 12/27/2022]
Abstract
The mammalian reproductive hormone axis regulates gonadal steroid hormone levels and gonadal function essential for reproduction. The neuroendocrine control of the axis integrates signals from a wide array of inputs. The regulatory pathways important for mediating these inputs have been the subject of numerous studies. One class of proteins that have been shown to mediate metabolic and growth signals to the CNS includes Insulin and IGF-1. These proteins are structurally related and can exert endocrine and growth factor like action via related receptor tyrosine kinases. The role that insulin and IGF-1 play in controlling the hypothalamus and pituitary and their role in regulating puberty and nutritional control of reproduction has been studied extensively. This review summarizes the in vitro and in vivo models that have been used to study these neuroendocrine structures and the influence of these growth factors on neuroendocrine control of reproduction.
Collapse
Affiliation(s)
- Andrew Wolfe
- Johns Hopkins University School of Medicine, Department of Pediatrics, Division of Endocrinology, Baltimore, MD 21287, United States.
| | - Sara Divall
- Johns Hopkins University School of Medicine, Department of Pediatrics, Division of Endocrinology, Baltimore, MD 21287, United States
| | - Sheng Wu
- Johns Hopkins University School of Medicine, Department of Pediatrics, Division of Endocrinology, Baltimore, MD 21287, United States
| |
Collapse
|
42
|
Qin F, Wang X, Liu S, Zheng Y, Li M, Zhang Y, Wang Z. Gene expression profiling of key genes in hypothalamus-pituitary-gonad axis of rare minnow Gobiocypris rarus in response to EE2. Gene 2014; 552:8-17. [PMID: 25194895 DOI: 10.1016/j.gene.2014.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 11/18/2022]
Abstract
The 17α-ethinylestradiol (EE2), which could induce estrogenic effects, is found in different aquatic systems. The current study aimed to assess in vivo effects of short-term EE2 exposure on the transcriptional activity of genes in the brain and gonad tissues in order to characterize the mode of action of EE2 on the hypothalamus-pituitary-gonad axis in rare minnow (Gobiocypris rarus). The full length cDNAs of fshβ, lhβ, fshr and lhr were first characterized in G. rarus. The homology and phylogenetic analyses of the amino acid sequences revealed that these four genes share high identity in cyprinid fish. The tissue distribution analysis by qRT-PCR showed that fshβ and lhβ were mainly expressed in the brain and fshr and lhr were mainly expressed in gonads. Adult G. rarus was exposed to EE2 at 1, 5, 25 and 125 ng/L for 3 and 6 days and the expression of brain cyp19a1b, fshβ and lhβ, estrogen receptors (esr1, esr2a, and esr2b) and gonadal fshr, lhr and cyp19a1a were assessed. Cyp19a1b was significantly up-regulated in the brains of female exposed to EE2 at 1-125 ng/L for 6 days. The brain lhβ, but not fshβ was strongly suppressed in most EE2 exposure groups of both sexes. The brain esr2b was inhibited in both sexes exposed to EE2 at all of the four concentrations for 6 days. Esr2a was up-regulated in the females by 6-day EE2 treatment at 1 and 25 ng/L. The high responsiveness of brain lhβ and esr2s to EE2 and their significant correlation in both sexes suggested that the transcriptional activity of Esr2s could play key roles in modulation of lhβ expression via direct action on gonadotropic cells in response to EE2. In gonads, fshr was strongly inhibited by EE2 in males, while lhr was significantly stimulated by EE2 in females. Cyp19a1a was inhibited by EE2 in both sexes. The positive correlations of gene expressions of both fshr and lhr with cyp19a1a in testes suggest that the suppression of 17α-estradiol (E2) synthesis in testis by exogenous estrogen could mediate via both Fsh/Fshr and Lh/Lhr signaling in male G. rarus.
Collapse
Affiliation(s)
- Fang Qin
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Xueqin Wang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Shaozhen Liu
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Yao Zheng
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Meng Li
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Yingying Zhang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Zaizhao Wang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China.
| |
Collapse
|
43
|
Yeo SH, Herbison AE. Estrogen-negative feedback and estrous cyclicity are critically dependent upon estrogen receptor-α expression in the arcuate nucleus of adult female mice. Endocrinology 2014; 155:2986-95. [PMID: 24905671 DOI: 10.1210/en.2014-1128] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The location and characteristics of cells within the brain that suppress GnRH neuron activity to contribute to the estrogen-negative feedback mechanism are poorly understood. Using adeno-associated virus (AAV)-mediated Cre-LoxP recombination in estrogen receptor-α (ERα) floxed mice (ERα(flox/flox)), we aimed to examine the role of ERα-expressing neurons located in the arcuate nucleus (ARN) in the estrogen-negative feedback mechanism. Bilateral injection of AAV-Cre into the ARN of ERα(flox/flox) mice (n = 14) resulted in the time-dependent ablation of up to 99% of ERα-immunoreactive cell numbers throughout the rostrocaudal length of the ARN. These mice were all acyclic by 5 weeks after AAV-Cre injections with most mice in constant estrous. Control wild-type mice injected with AAV-Cre (n = 13) were normal. Body weight was not altered in ERα(flox/flox) mice. After ovariectomy, a significant increment in LH secretion was observed in all genotypes, although its magnitude was reduced in ERα(flox/flox) mice. Acute and chronic estrogen-negative feedback were assessed by administering 17β-estradiol to mice as a bolus (LH measured 3 h later) or SILASTIC brand capsule implant (LH measured 5 d later). This demonstrated that chronic estrogen feedback was absent in ERα(flox/flox) mice, whereas the acute feedback was normal. These results reveal a critical role for ERα-expressing cells within the ARN in both estrous cyclicity and the chronic estrogen negative feedback mechanism in female mice. This suggests that ARN cells provide a key indirect, transsynpatic route through which estradiol suppresses the activity of GnRH neurons.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | | |
Collapse
|
44
|
Leptin-responsive GABAergic neurons regulate fertility through pathways that result in reduced kisspeptinergic tone. J Neurosci 2014; 34:6047-56. [PMID: 24760864 DOI: 10.1523/jneurosci.3003-13.2014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The adipocyte-derived hormone leptin plays a critical role in the central transmission of energy balance to modulate reproductive function. However, the neurocircuitry underlying this interaction remains elusive, in part due to incomplete knowledge of first-order leptin-responsive neurons. To address this gap, we explored the contribution of predominantly inhibitory (GABAergic) neurons versus excitatory (glutamatergic) neurons in the female mouse by selective ablation of the leptin receptor in each neuronal population: Vgat-Cre;Lepr(lox/lox) and Vglut2-Cre;Lepr(lox/lox) mice, respectively. Female Vgat-Cre;Lepr(lox/lox) but not Vglut2-Cre;Lepr(lox/lox) mice were obese. Vgat-Cre;Lepr(lox/lox) mice had delayed or absent vaginal opening, persistent diestrus, and atrophic reproductive tracts with absent corpora lutea. In contrast, Vglut2-Cre;Lepr(lox/lox) females exhibited reproductive maturation and function comparable to Lepr(lox/lox) control mice. Intracerebroventricular administration of kisspeptin-10 to Vgat-Cre;Lepr(lox/lox) female mice elicited robust gonadotropin responses, suggesting normal gonadotropin-releasing hormone neuronal and gonadotrope function. However, adult ovariectomized Vgat-Cre;Lepr(lox/lox) mice displayed significantly reduced levels of Kiss1 (but not Tac2) mRNA in the arcuate nucleus, and a reduced compensatory luteinizing hormone increase compared with control animals. Estradiol replacement after ovariectomy inhibited gonadotropin release to a similar extent in both groups. These animals also exhibited a compromised positive feedback response to sex steroids, as shown by significantly lower Kiss1 mRNA levels in the AVPV, compared with Lepr(lox/lox) mice. We conclude that leptin-responsive GABAergic neurons, but not glutamatergic neurons, act as metabolic sensors to regulate fertility, at least in part through modulatory effects on kisspeptin neurons.
Collapse
|
45
|
Fortin J, Boehm U, Deng CX, Treier M, Bernard DJ. Follicle-stimulating hormone synthesis and fertility depend on SMAD4 and FOXL2. FASEB J 2014; 28:3396-410. [PMID: 24739304 DOI: 10.1096/fj.14-249532] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Follicle-stimulating hormone (FSH) is an essential regulator of gonadal function and fertility. Loss-of-function mutations in the FSHB/Fshb gene cause hypogonadotropic hypogonadism in humans and mice. Both gonadotropin-releasing hormone (GnRH) and activins, members of the transforming growth factor β (TGFβ) superfamily, stimulate FSH synthesis; yet, their relative roles and mechanisms of action in vivo are unknown. Here, using conditional gene-targeting, we show that the canonical mediator of TGFβ superfamily signaling, SMAD4, is absolutely required for normal FSH synthesis in both male and female mice. Moreover, when the Smad4 gene is ablated in combination with its DNA binding cofactor Foxl2 in gonadotrope cells, mice make essentially no FSH and females are sterile. Indeed, the phenotype of these animals is remarkably similar to that of Fshb-knockout mice. Not only do these results establish SMAD4 and FOXL2 as essential master regulators of Fshb transcription in vivo, they also suggest that activins, or related ligands, could play more important roles in FSH synthesis than GnRH.
Collapse
Affiliation(s)
- Jérôme Fortin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada;
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, Homburg, Germany
| | - Chu-Xia Deng
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA; and
| | - Mathias Treier
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada;
| |
Collapse
|
46
|
Wu S, Divall S, Nwaopara A, Radovick S, Wondisford F, Ko C, Wolfe A. Obesity-induced infertility and hyperandrogenism are corrected by deletion of the insulin receptor in the ovarian theca cell. Diabetes 2014; 63:1270-82. [PMID: 24379345 PMCID: PMC3964497 DOI: 10.2337/db13-1514] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/16/2013] [Indexed: 01/21/2023]
Abstract
Women with polycystic ovary syndrome (PCOS) exhibit elevated androgen levels, oligoanovulation, infertility, and insulin resistance in metabolic tissues. The aims of these studies were to determine the role of insulin signaling in the development and function of ovarian theca cells and the pathophysiologic effects of hyperinsulinism on ovarian function in obesity. We disrupted the insulin receptor (IR) gene specifically in the theca-interstitial (TI) cells of the ovaries (Cyp17IRKO). No changes in reproductive development or function were observed in lean Cyp17IRKO female mice, suggesting that insulin signaling in TI cell is not essential for reproduction. However, when females were fed a high-fat diet, diet-induced obesity (DIO) wild-type (DIO-WT) mice were infertile and experienced increased circulating testosterone levels, whereas DIO-Cyp17IRKO mice exhibited improved fertility and testosterone levels comparable to those found in lean mice. The levels of phosphorylated IRS1 and CYP17 protein were higher in the ovary of DIO-WT compared with DIO-Cyp17IRKO or lean mice. Ex vivo studies using a whole ovary culture model demonstrated that insulin acts independently or additively with human chorionic gonadotropin to enhance androstenedione secretion. These studies reveal the causal pathway linking hyperinsulinism with ovarian hyperandrogenism and the infertility of obesity.
Collapse
Affiliation(s)
- Sheng Wu
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sara Divall
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Amanda Nwaopara
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sally Radovick
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Fredric Wondisford
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - CheMyong Ko
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Andrew Wolfe
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
47
|
Cheong RY, Porteous R, Chambon P, Abrahám I, Herbison AE. Effects of neuron-specific estrogen receptor (ER) α and ERβ deletion on the acute estrogen negative feedback mechanism in adult female mice. Endocrinology 2014; 155:1418-27. [PMID: 24476134 DOI: 10.1210/en.2013-1943] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The negative feedback mechanism through which 17β-estradiol (E2) acts to suppress the activity of the GnRH neurons remains unclear. Using inducible and cell-specific genetic mouse models, we examined the estrogen receptor (ER) isoforms expressed by neurons that mediate acute estrogen negative feedback. Adult female mutant mice in which ERα was deleted from all neurons in the neonatal period failed to exhibit estrous cycles or negative feedback. Adult mutant female mice with neonatal neuronal ERβ deletion exhibited normal estrous cycles, but a failure of E2 to suppress LH secretion was seen in ovariectomized mice. Mutant mice with a GnRH neuron-selective deletion of ERβ exhibited normal cycles and negative feedback, suggesting no critical role for ERβ in GnRH neurons in acute negative feedback. To examine the adult roles of neurons expressing ERα, an inducible tamoxifen-based Cre-LoxP approach was used to ablate ERα from neurons that express calmodulin kinase IIα in adults. This resulted in mice with no estrous cycles, a normal increase in LH after ovariectomy, but an inability of E2 to suppress LH secretion. Finally, acute administration of ERα- and ERβ-selective agonists to adult ovariectomized wild-type mice revealed that activation of ERα suppressed LH secretion, whereas ERβ agonists had no effect. This study highlights the differences in adult reproductive phenotypes that result from neonatal vs adult ablation of ERα in the brain. Together, these experiments expand previous global knockout studies by demonstrating that neurons expressing ERα are essential and probably sufficient for the acute estrogen negative feedback mechanism in female mice.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Centre for Neuroendocrinology (R.Y.C., R.P., I.A., A.E.H.), Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand; and Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), 67400 Illkirch, France
| | | | | | | | | |
Collapse
|
48
|
Novaira HJ, Sonko ML, Hoffman G, Koo Y, Ko C, Wolfe A, Radovick S. Disrupted kisspeptin signaling in GnRH neurons leads to hypogonadotrophic hypogonadism. Mol Endocrinol 2014; 28:225-38. [PMID: 24422632 DOI: 10.1210/me.2013-1319] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Landmark studies have shown that mutations in kisspeptin and the kisspeptin receptor (Kiss1r) result in reproductive dysfunction in humans and genetically altered mouse models. However, because kisspeptin and its receptor are present in target cells of the central and peripheral reproductive axis, the precise location(s) for the pathogenic signal is unknown. The study described herein shows that the kisspeptin-Kiss1r signaling pathway in the GnRH neuron is singularly critical for both the onset of puberty as well as the attainment of normal reproductive function. In this study, we directly test the hypothesis that kisspeptin neurons regulate GnRH secretion through the activation of Kiss1r on the plasma membrane of GnRH neurons. A GnRH neuron-specific Kiss1r knockout mouse model (GKirKO) was generated, and reproductive development and phenotype were assessed. Both female and male GKirKO mice were infertile, having low serum LH and FSH levels. External abnormalities such as microphallus and decreased anogenital distance associated with failure of preputial gland separation were present in GKirKO males. A delay in pubertal onset and abnormal estrous cyclicity were observed in female GKirKO mice. Taken together, these data provide in vivo evidence that Kiss1r in GnRH neurons is critical for reproductive development and fertility.
Collapse
Affiliation(s)
- Horacio J Novaira
- Department of Pediatrics (H.J.N., M.L.S., A.W., S.R.), Division of Endocrinology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; Department of Biology (G.H.), Morgan State University, Baltimore, Maryland 21251; School of Biological Sciences (Y.K.), Inje University, Gimhae, 621-749, South Korea; and University of Illinois at Champaign-Urbana (C.K.), Champaign, Illinois 61820
| | | | | | | | | | | | | |
Collapse
|
49
|
Avtanski D, Novaira HJ, Wu S, Romero CJ, Kineman R, Luque RM, Wondisford F, Radovick S. Both estrogen receptor α and β stimulate pituitary GH gene expression. Mol Endocrinol 2014; 28:40-52. [PMID: 24284820 PMCID: PMC3874459 DOI: 10.1210/me.2013-1245] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 11/18/2013] [Indexed: 11/19/2022] Open
Abstract
Although sex steroids have been implicated in the control of mammalian growth, their direct effect on GH synthesis is less clear. The aim of this study was to establish whether estradiol (E2) directly affects GH synthesis in somatotrophs. Somatotroph GH3 and MtT/S cells were used as in vitro models. At physiological doses of E2 stimulation, GH mRNA levels were increased and the ER antagonist ICI 182,780 completely abolished this effect. Estrogen receptor (ER) α- and ERβ-selective agonists, propylpyrazole triol (PPT), and 2,3-bis(4-hydroxyphenyl) propionitrile (DPN), respectively, augmented GH mRNA expression and secretion, whereas E2 and PPT, but not DPN increased prolactin (PRL) mRNA levels. E2, PPT, and DPN stimulated expression of the pituitary transcription factor Pou1f1 and increased its binding to the GH promoter. In vivo evidence of E2 effects on GH synthesis was obtained from the generation of the somatotroph-specific ERα knockout (sERα-KO) mouse model. Basal pituitary GH, PRL, POU1F1, and ERα mRNA expression levels were lower in sERα-KO mice compared with those in controls; whereas ERβ mRNA levels remained unchanged. E2 and DPN stimulated pituitary GH mRNA expression and serum GH levels in control and sERα-KO ovariectomized mice; however, serum GH levels were unchanged in PPT-treated ovariectomized sERα-KO mice. In these animal models, PRL mRNA levels increased after either E2 or PPT, but an increase was not seen after DPN treatment. Thus, we propose a mechanism by which estrogen directly regulates somatotroph GH synthesis at a pretranslational level. In contrast to the predominant effect of ERα in the lactotroph, these results support a role for both ERα and ERβ in the transcriptional control of Gh in the somatotroph and illustrate important differences in ER isoform specificity in the anterior pituitary gland.
Collapse
Affiliation(s)
| | | | - Sheng Wu
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Christopher J. Romero
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Rhonda Kineman
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Raul M. Luque
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Fredric Wondisford
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Sally Radovick
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| |
Collapse
|
50
|
Hamilton KJ, Arao Y, Korach KS. Estrogen hormone physiology: reproductive findings from estrogen receptor mutant mice. Reprod Biol 2013; 14:3-8. [PMID: 24607249 DOI: 10.1016/j.repbio.2013.12.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 11/08/2013] [Accepted: 12/10/2013] [Indexed: 11/16/2022]
Abstract
Estrogen receptors (ERs) play a crucial role in reproduction and normal physiology. The two sub-types of ER (ERα and β) are expressed in various levels in different tissues and selective cell types. Gene targeting technology allowed us to produce lines of mice with disrupted ERα (αERKO) and ERβ genes (βERKO) as well as a compound αβERKO in the whole body. Male and female αERKO mice are infertile. Estrogen, EGF and IGF-1 treatments failed to induce uterine growth and DNA synthesis in αERKO uteri. αERKO females are infertile due to hypoplastic uteri and hyperemic ovaries with no corpora lutea due to persistent LH stimulation from loss of negative feedback. αERKO males are infertile, with testicular atrophy and seminiferous tubule dysmorphogenesis producing decreased spermatogenesis and inactive sperm. βERKO females show arrested folliculogenesis and subfertility. Ovarian analyses indicate differential gene expression related to ovulatory stimulation deficits including lack of LH, PR, Cyp19 and Cox2 expression. A unique ovarian phenotype is found only in αβERKO females showing transdifferentiation of granulosa cells to Sertoli cells. We describe here several novel mouse models which possess ERα gene modification. To understand ERα function in uterine endometrial epithelial cells, we generated a tissue selective ERα gene disrupted mouse model, the uterine epithelial-specific ERα knockout (UtEpiαERKO). To understand the physiological role of ERα functional domains, we generated a mouse model with a mutation in the ligand dependent transcription activation domain of ERα (AF2ERKI). Findings from the ERα mutant mice suggest that the absence of functional ERα is not lethal and results in significant endocrine effects and altered physiological processes.
Collapse
Affiliation(s)
- Katherine J Hamilton
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC 27709, United States
| | - Yukitomo Arao
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC 27709, United States
| | - Kenneth S Korach
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC 27709, United States.
| |
Collapse
|