1
|
Binder AK, Burns KA, Rodriguez KF, Hamilton K, Pardo-Manuel de Villena F, Korach KS. Postnatal Ovarian Transdifferentiation in the Absence of Estrogen Receptor Signaling Is Dependent on Genetic Background. Endocrinology 2024; 166:bqae157. [PMID: 39576259 PMCID: PMC11630523 DOI: 10.1210/endocr/bqae157] [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: 08/16/2024] [Indexed: 12/12/2024]
Abstract
Normal ovarian function requires the expression of estrogen receptors α (ESR1) and β (ESR2) in distinct cell types within the ovary. The double estrogen receptor knockout (αβERKO) ovary had the appearance of seminiferous tubule-like structures that expressed SOX9; this phenotype was lost when the animals were repeatedly backcrossed to the C57BL/6J genetic background. A new line of ERKO mice, Ex3αβERKO, was developed for targeted disruption on a mixed genetic background. Histological examination of the ovaries in the Ex3αβERKO showed the appearance of seminiferous tubule-like structures in mice aged 6 to 12 months. These dismorphogenic regions have cells that no longer express granulosa cell-specific FOXL2, while other cells express Sertoli cell-specific SOX9 as examined by immunohistochemistry. Whole ovarian gene expression analysis in Ex3αERKO, Ex3βRKO, and Ex3αβERKO found many genes differentially expressed compared to controls with one Esr1 and Esr2 allele. The genes specific to the Ex3αβERKO ovary were compared to other models of postnatal ovarian transdifferentiation, identifying 21 candidate genes. To examine the genetic background contributions, DNA was isolated from αβERKO mice that did not show ovarian transdifferentiation and compared to DNA from Ex3αβERKO using Mouse Diversity Array. A genomic region putatively associated with transdifferentiation was identified on Chr18 (5-15 M) and genes in this region were compared to the genes differentially expressed in models of ovarian transdifferentiation. This work demonstrates the importance of ESRs in maintaining granulosa cell differentiation within the ovary, identifies several potential gene candidates, and suggests that genetic background can be a confounding factor.
Collapse
Affiliation(s)
- April K Binder
- Department of Biological Sciences, Central Washington University, Ellensburg, WA 98926, USA
- Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
- Reproductive & Developmental Biology Laboratory, NIEHS, NIH, Research Triangle Park, NC 27709, USA
| | - Katherine A Burns
- Reproductive & Developmental Biology Laboratory, NIEHS, NIH, Research Triangle Park, NC 27709, USA
- Department of Environmental and Public Health Science, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Karina F Rodriguez
- Reproductive & Developmental Biology Laboratory, NIEHS, NIH, Research Triangle Park, NC 27709, USA
| | - Katherine Hamilton
- Reproductive & Developmental Biology Laboratory, NIEHS, NIH, Research Triangle Park, NC 27709, USA
| | | | - Kenneth S Korach
- Reproductive & Developmental Biology Laboratory, NIEHS, NIH, Research Triangle Park, NC 27709, USA
| |
Collapse
|
2
|
Cai S, Chen M, Xue B, Zhu Z, Wang X, Li J, Wang H, Zeng X, Qiao S, Zeng X. Retinoic acid enhances ovarian steroidogenesis by regulating granulosa cell proliferation and MESP2/STAR/CYP11A1 pathway. J Adv Res 2024; 58:163-173. [PMID: 37315842 PMCID: PMC10982869 DOI: 10.1016/j.jare.2023.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/28/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023] Open
Abstract
INTRODUCTION Ovarian steroidogenesis not only affects the embryonic development and pregnancy outcome, but also associates with many diseases in mammals and women. Exploring the nutrients and mechanisms influencing ovarian steroidogenesis is critical to maintaining the optimal reproductive performance, as well as guaranteeing body health. OBJECTIVES This research aimed to explore the effect of retinol metabolism on ovarian steroidogenesis and the underlying mechanisms. METHODS Comparative transcriptomic analysis of ovaries from normal and low reproductive performance sows were performed to identify the main causes leading to low fertility. The metabolites regulating steroid hormones synthesis were investigated in ovarian granulosa cells. Gene interference, overexpression, dual-luciferase reporter assays, chromatin immunoprecipitation and transcriptome analysis were further conducted to explore the underlying mechanisms of Aldh1a1 mediating ovarian steroidogenesis. RESULTS Transcriptome analysis of ovaries from normal and low reproductive performance sows showed the significant differences in both retinol metabolism and steroid hormones synthesis, indicating retinol metabolism probably influenced steroid hormones synthesis. The related metabolite retinoic acid was furtherly proven a highly active and potent substance strengthening estrogen and progesterone synthesis in ovarian granulosa cells. For the first time, we revealed that retinoic acid synthesis in porcine and human ovarian granulosa cells was dominated by Aldh1a1, and required the assistance of Aldh1a2. Importantly, we demonstrated that Aldh1a1 enhanced the proliferation of ovarian granulosa cells by activating PI3K-Akt-hedgehog signaling pathways. In addition, Aldh1a1 regulated the expression of transcription factor MESP2, which targeted the transcription of Star and Cyp11a1 through binding to corresponding promoter regions. CONCLUSION Our data identified Aldh1a1 modulates ovarian steroidogenesis through enhancing granulosa cell proliferation and MESP2/STAR/CYP11A1 pathway. These findings provide valuable clues for improving ovarian health in mammals.
Collapse
Affiliation(s)
- Shuang Cai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China; Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, PR China
| | - Bangxin Xue
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Zhekun Zhu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Xinyu Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Jie Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China
| | - Huakai Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Xiangzhou Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, PR China; Beijing Biofeed Additive Key Laboratory, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
3
|
Baskin L, Cao M, Askel S, Li Y, Cunha G. Ovotesticular cords and ovotesticular follicles: New markers in a model of human mixed ovotestis. Differentiation 2024; 135:100739. [PMID: 38000942 DOI: 10.1016/j.diff.2023.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/21/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023]
Affiliation(s)
- Laurence Baskin
- Department of Urology, University of California, San Francisco, United States.
| | - Mei Cao
- Department of Urology, University of California, San Francisco, United States
| | - Sena Askel
- Department of Urology, University of California, San Francisco, United States
| | - Yi Li
- Department of Urology, University of California, San Francisco, United States
| | - Gerald Cunha
- Department of Urology, University of California, San Francisco, United States
| |
Collapse
|
4
|
Pei J, Xiong L, Guo S, Wang X, La Y, Chu M, Liang C, Yan P, Guo X. Single-Cell Transcriptomics Analysis Reveals a Cell Atlas and Cell Communication in Yak Ovary. Int J Mol Sci 2023; 24:ijms24031839. [PMID: 36768166 PMCID: PMC9915757 DOI: 10.3390/ijms24031839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/15/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Yaks (Bos grunniens) are the only bovine species that adapt well to the harsh high-altitude environment in the Qinghai-Tibetan plateau. However, the reproductive adaptation to the climate of the high elevation remains to be elucidated. Cell composition and molecular characteristics are the foundation of normal ovary function which determines reproductive performance. So, delineating ovarian characteristics at a cellular molecular level is conducive to elucidating the mechanism underlying the reproductive adaption of yaks. Here, the single-cell RNA-sequencing (scRNA-seq) was employed to depict an atlas containing different cell types with specific molecular signatures in the yak ovary. The cell types were identified on the basis of their specifically expressed genes and biological functions. As a result, a cellular atlas of yak ovary was established successfully containing theca cells, stromal cells, endothelial cells, smooth muscle cells, natural killer cells, macrophages, and proliferating cells. A cell-to-cell communication network between the distinct cell types was constructed. The theca cells were clustered into five subtypes based on their biological functions. Further, CYP11A1 was confirmed as a marker gene for the theca cells by immunofluorescence staining. Our work reveals an ovarian atlas at the cellular molecular level and contributes to providing insights into reproductive adaption in yaks.
Collapse
Affiliation(s)
- Jie Pei
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Yongfu La
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Min Chu
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Chunnian Liang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xian Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Correspondence:
| |
Collapse
|
5
|
Pei J, Xiong L, Guo S, Wang X, Bao P, Wu X, Yan P, Guo X. A single-cell transcriptomic atlas characterizes cell types and their molecular features in yak ovarian cortex. FASEB J 2023; 37:e22718. [PMID: 36527406 DOI: 10.1096/fj.202201176rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The ovary as one of the most dynamic organs produces steroids to orchestrate female secondary sexual characteristics, harbors ovarian reserve for oocytes, releases mature oocytes for fertilization, and maintains pregnancy. Yak (Bos grunniens) is the only bovid animal that can adapt to the harsh climatic conditions on the Qinghai-Tibetan Plateau (altitudes of over 3000 m above sea level). However, the cellular atlas is composed of oocytes and other somatic cells, and their individual molecular characteristics remain to be elucidated in the yak ovary. Here, single-cell RNA sequencing (scRNA-seq) was performed to delineate the molecular signature of various cell types in the yak ovarian cortex. A cellular atlas of yak ovarian cortex was constructed successfully on the basis of the differentially expressed genes (DEGs) from the distinct cell types and their functional enrichment analysis, comprising endothelial cells, nature kill cells, stromal cells, smooth muscle cells, oocytes, macrophages, epithelial cells, and granulosa cells. Meanwhile, the signature genes were determined based on their expression specificity in each cell type. A cell-to-cell communication network was built in light of the differentially overexpressed ligand and receptor genes from each cell type. Further, the oocytes were subdivided into four subtypes based on their individual DEGs and the functional enrichment of the DEGs. FST and TOP2A were identified as maker genes for oocytes by immunostaining in the yak ovarian cortex. The cellular atlas reveals the biological characteristics of the ovarian cortex at the cellular molecular level and provides insights into female reproductive biology via cellular communications in the yak.
Collapse
Affiliation(s)
- Jie Pei
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengjia Bao
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Xiaoyun Wu
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Xian Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| |
Collapse
|
6
|
Nicol B, Estermann MA, Yao HHC, Mellouk N. Becoming female: Ovarian differentiation from an evolutionary perspective. Front Cell Dev Biol 2022; 10:944776. [PMID: 36158204 PMCID: PMC9490121 DOI: 10.3389/fcell.2022.944776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/16/2022] [Indexed: 01/09/2023] Open
Abstract
Differentiation of the bipotential gonadal primordium into ovaries and testes is a common process among vertebrate species. While vertebrate ovaries eventually share the same functions of producing oocytes and estrogens, ovarian differentiation relies on different morphogenetic, cellular, and molecular cues depending on species. The aim of this review is to highlight the conserved and divergent features of ovarian differentiation through an evolutionary perspective. From teleosts to mammals, each clade or species has a different story to tell. For this purpose, this review focuses on three specific aspects of ovarian differentiation: ovarian morphogenesis, the evolution of the role of estrogens on ovarian differentiation and the molecular pathways involved in granulosa cell determination and maintenance.
Collapse
Affiliation(s)
- Barbara Nicol
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States,*Correspondence: Barbara Nicol,
| | - Martin A. Estermann
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Humphrey H-C Yao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Namya Mellouk
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy en Josas, France
| |
Collapse
|
7
|
Kuan KKW, Saunders PTK. Female Reproductive Systems: Hormone Dependence and Receptor Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:21-39. [PMID: 36107311 DOI: 10.1007/978-3-031-11836-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The female reproductive system which consists of the ovaries, uterus (myometrium, endometrium), Fallopian tubes, cervix and vagina is exquisitely sensitive to the actions of steroid hormones. The ovaries play a key role in the synthesis of bioactive steroids (oestrogens, androgens, progestins) that act both within the tissue (intracrine/paracrine) as well as on other reproductive organs following release into the blood stream (endocrine action). Sex steroid receptors encoded by the oestrogen (ESR1, ESR2), progesterone (PR) and androgen (AR) receptor genes, which are members of the superfamily of ligand activated transcription factors are widely expressed within these tissues. These receptors play critical role(s) in regulation of cell proliferation, ovulation, endometrial receptivity, myometrial cell function and inflammatory cell infiltration. Our understanding of their importance has been informed by studies on human tissues and cells, which have employed immunohistochemistry as well as a wide range of molecular and genetic methods to identify which processes are dependent steroid ligand activation. The development of mice with targeted deletions of each of these receptors has provided complementary data that has extended our appreciation of cell-cell interactions in the fine tuning of reproductive tissue function. This large body of work has formed the basis of new and improved therapeutics to treat conditions such as infertility.
Collapse
Affiliation(s)
- Kevin K W Kuan
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
8
|
Shi H, Ru X, Pan S, Jiang D, Huang Y, Zhu C, Li G. Transcriptomic analysis of pituitary in female and male spotted scat (Scatophagus argus) after 17β-estradiol injection. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2021; 41:100949. [PMID: 34942522 DOI: 10.1016/j.cbd.2021.100949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
Spotted scat (Scatophagus argus) is a popular species of marine fish cultured in China. It shows normal sexual growth dimorphism. Female spotted scat grows quicker and bigger than males. Growth and reproduction are the most important traits in aquaculture. In vertebrates, the pituitary gland occupies an important position in the growth and reproduction axis. Estrogen is involved in regulating growth and reproduction in the pituitary gland in an endocrine fashion. Transcriptome sequencing of the pituitary was performed in female and male fish at 6 h after 17β-estradiol injection (4.0 μg E2/g body weight, BW). Compared with the pituitary of female and male groups, 144 and 64 genes [|log2(fold change)| ≥ 1.0 and false discovery rate (FDR) < 0.05] were significantly differentially expressed in E2-injected females and males, respectively (p < 0.05). Of these, 59 and 48 were up-regulated, and 85 and 16 were down-regulated. According to the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathway analyses, DEGs were involved in signal pathways, such as growth, reproduction, oocyte meiosis and steroid biosynthesis. Of these, estrogen affected the expression of some sex steroid synthesis and receptor genes in the pituitary gland through feedback, such as hsd17b7, pgr and cyp19a1b, regulating the reproductive activities. Besides, some growth-related genes, such as gap43, junbb, mstn2 and insm1a responded to estrogen. E2 might affect the expression level of gh mRNA by regulating the expression levels of growth-related genes. Our results provide a theoretical basis for studying the molecular mechanism of growth and reproduction regulation at the pituitary level of spotted scat responded to E2.
Collapse
Affiliation(s)
- Hongjuan Shi
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaoying Ru
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Southern Marine Science and Engineering Guangdong Laboratory-Zhanjiang, Zhanjiang 524088, China
| | - Shuhui Pan
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Dongneng Jiang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yang Huang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chunhua Zhu
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guangli Li
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW The aim of this study was to provide a basic overview on human sex development with a focus on involved genes and pathways, and also to discuss recent advances in the molecular diagnostic approaches applied to clinical workup of individuals with a difference/disorder of sex development (DSD). RECENT FINDINGS Rapid developments in genetic technologies and bioinformatics analyses have helped to identify novel genes and genomic pathways associated with sex development, and have improved diagnostic algorithms to integrate clinical, hormonal and genetic data. Recently, massive parallel sequencing approaches revealed that the phenotype of some DSDs might be only explained by oligogenic inheritance. SUMMARY Typical sex development relies on very complex biological events, which involve specific interactions of a large number of genes and pathways in a defined spatiotemporal sequence. Any perturbation in these genetic and hormonal processes may result in atypical sex development leading to a wide range of DSDs in humans. Despite the huge progress in the understanding of molecular mechanisms underlying DSDs in recent years, in less than 50% of DSD individuals, the genetic cause is currently solved at the molecular level.
Collapse
Affiliation(s)
- Idoia Martinez de LaPiscina
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, UPV/EHU, CIBERER, CIBERDEM, ENDO-ERN, Barakaldo, Spain
| | - Christa E Flück
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
10
|
Major AT, Estermann MA, Roly ZY, Smith CA. An evo-devo perspective of the female reproductive tract. Biol Reprod 2021; 106:9-23. [PMID: 34494091 DOI: 10.1093/biolre/ioab166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/22/2023] Open
Abstract
The vertebrate female reproductive tract has undergone considerable diversification over evolution, having become physiologically adapted to different reproductive strategies. This review considers the female reproductive tract from the perspective of evolutionary developmental biology (evo-devo). Very little is known about how the evolution of this organ system has been driven at the molecular level. In most vertebrates, the female reproductive tract develops from paired embryonic tubes, the Müllerian ducts. We propose that formation of the Müllerian duct is a conserved process that has involved co-option of genes and molecular pathways involved in tubulogenesis in the adjacent mesonephric kidney and Wolffian duct. Downstream of this conservation, genetic regulatory divergence has occurred, generating diversity in duct structure. Plasticity of the Hox gene code and wnt signaling, in particular, may underlie morphological variation of the uterus in mammals, and evolution of the vagina. This developmental plasticity in Hox and Wnt activity may also apply to other vertebrates, generating the morphological diversity of female reproductive tracts evident today.
Collapse
Affiliation(s)
- Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Zahida Y Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| |
Collapse
|
11
|
Telomere associated gene expression as well as TERT protein level and telomerase activity are altered in the ovarian follicles of aged mice. Sci Rep 2021; 11:15569. [PMID: 34330985 PMCID: PMC8324818 DOI: 10.1038/s41598-021-95239-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/05/2021] [Indexed: 01/23/2023] Open
Abstract
Telomeres cap the ends of eukaryotic chromosomes to maintain genomic stability and integrity during an organism’s lifespan. The length of telomeres inevitably shortens due to DNA replication, genotoxic agents, and biological aging. A limited number of cell types, e.g., stem cells, germline cells, and early embryos can elongate shortened telomeres via the enzymatic action of telomerase, which is composed of telomerase reverse transcriptase (TERT) and telomerase RNA component (Terc). Additionally, telomere-associated proteins including telomeric repeat binding factor 1 (TRF1) and 2 (TRF2), as well as protection of telomeres 1a (POT1a), bind to telomeres to maintain their structural integrity and length. During ovarian aging in mammals, telomeres progressively shorten, accompanied by fertility loss; however, the molecular mechanism underlying this attrition during follicle development remains unclear. In this study, the primary, secondary, preantral, and antral follicles were obtained either from 6-week-old adult (n = 19) or 52-week-old aged (n = 12) mice. We revealed that the Tert, Terc, Trf1, Trf2, and Pot1a gene expression (P < 0.001) and TERT protein (P < 0.01) levels significantly decreased in certain ovarian follicles of the aged group when compared to those of the adult group. Also, telomerase activity exhibited remarkable changes in the follicles of both groups. Consequently, altered telomere-associated gene expression and reduced TERT protein levels in the follicles of aged mice may be a determinant of telomere shortening during ovarian aging, and infertility appearing in the later decades of reproductive lifespan. Further investigations are required to determine the molecular mechanisms underlying these alterations in the follicles during ovarian aging.
Collapse
|
12
|
Jiménez R, Burgos M, Barrionuevo FJ. Sex Maintenance in Mammals. Genes (Basel) 2021; 12:genes12070999. [PMID: 34209938 PMCID: PMC8303465 DOI: 10.3390/genes12070999] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/26/2021] [Accepted: 06/26/2021] [Indexed: 12/30/2022] Open
Abstract
The crucial event in mammalian sexual differentiation occurs at the embryonic stage of sex determination, when the bipotential gonads differentiate as either testes or ovaries, according to the sex chromosome constitution of the embryo, XY or XX, respectively. Once differentiated, testes produce sexual hormones that induce the subsequent differentiation of the male reproductive tract. On the other hand, the lack of masculinizing hormones in XX embryos permits the formation of the female reproductive tract. It was long assumed that once the gonad is differentiated, this developmental decision is irreversible. However, several findings in the last decade have shown that this is not the case and that a continuous sex maintenance is needed. Deletion of Foxl2 in the adult ovary lead to ovary-to-testis transdifferentiation and deletion of either Dmrt1 or Sox9/Sox8 in the adult testis induces the opposite process. In both cases, mutant gonads were genetically reprogrammed, showing that both the male program in ovaries and the female program in testes must be actively repressed throughout the individual's life. In addition to these transcription factors, other genes and molecular pathways have also been shown to be involved in this antagonism. The aim of this review is to provide an overview of the genetic basis of sex maintenance once the gonad is already differentiated.
Collapse
|
13
|
Stewart MK, Mattiske DM, Pask AJ. Exogenous Oestrogen Impacts Cell Fate Decision in the Developing Gonads: A Potential Cause of Declining Human Reproductive Health. Int J Mol Sci 2020; 21:E8377. [PMID: 33171657 PMCID: PMC7664701 DOI: 10.3390/ijms21218377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
The increasing incidence of testicular dysgenesis syndrome-related conditions and overall decline in human fertility has been linked to the prevalence of oestrogenic endocrine disrupting chemicals (EDCs) in the environment. Ectopic activation of oestrogen signalling by EDCs in the gonad can impact testis and ovary function and development. Oestrogen is the critical driver of ovarian differentiation in non-mammalian vertebrates, and in its absence a testis will form. In contrast, oestrogen is not required for mammalian ovarian differentiation, but it is essential for its maintenance, illustrating it is necessary for reinforcing ovarian fate. Interestingly, exposure of the bi-potential gonad to exogenous oestrogen can cause XY sex reversal in marsupials and this is mediated by the cytoplasmic retention of the testis-determining factor SOX9 (sex-determining region Y box transcription factor 9). Oestrogen can similarly suppress SOX9 and activate ovarian genes in both humans and mice, demonstrating it plays an essential role in all mammals in mediating gonad somatic cell fate. Here, we review the molecular control of gonad differentiation and explore the mechanisms through which exogenous oestrogen can influence somatic cell fate to disrupt gonad development and function. Understanding these mechanisms is essential for defining the effects of oestrogenic EDCs on the developing gonads and ultimately their impacts on human reproductive health.
Collapse
Affiliation(s)
- Melanie K. Stewart
- School of BioSciences, The University of Melbourne, Melbourne, VIC 3010, Australia; (D.M.M.); (A.J.P.)
| | | | | |
Collapse
|
14
|
The altered expression of telomerase components and telomere-linked proteins may associate with ovarian aging in mouse. Exp Gerontol 2020; 138:110975. [DOI: 10.1016/j.exger.2020.110975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 12/27/2022]
|
15
|
The Dietary Effect of Vitex agnus-castus Hydroalcoholic Extract on Growth Performance, Blood Biochemical Parameters, Carcass Quality, Sex Ratio and Gonad Histology in Zebrafish (Danio rerio). APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10041402] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The main purpose of this study was to consider the impact of Vitex agnus-castus hydroalcoholic extract, containing phytoestrogenic compounds on growth indices, sex ratio and histology of gonads of female Zebrafish. Fish larvae (4-day-old after hatching) were nourished with investigational diets (0 (control group T0), 5 g (T1), 10 g (T2) and 15 g (T3) kg−1 food) from first active feeding for 90 days. The results showed that Condition Factor (CF), Food Conversion Rate (FCR), Body Weight Gain (BWG), and Specific Growth Rate (SGR) were affected meaningfully by hydroalcoholic extract of Vitex agnus-castus (p < 0.05). These factors were significantly higher in compare to T3 treatment (p < 0.05). With increasing concentration of the extract in the diet, the ratio of female to male increased Treatment of T3 resulted in 87.23% feminization. Overall, this study suggests the use of T3 treatment achieves the best reproduction performance and a higher percentage of females in zebrafish as a model for aquaculture species. According to the results, the lowest levels of glucose and cholesterol were observed in T3 treatment and showed a significant changes with the control and treatments (p < 0.05).
Collapse
|
16
|
Retinoic acid signaling in ovarian folliculogenesis and steroidogenesis. Reprod Toxicol 2019; 87:32-41. [PMID: 31059772 DOI: 10.1016/j.reprotox.2019.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/13/2019] [Accepted: 04/30/2019] [Indexed: 12/31/2022]
Abstract
Retinoids are essential for reproduction. Most research has focused on the role of retinoic acid signaling in the regulation of meiosis during early fetal germ cell development. However, less attention has been paid to the possible effects of retinoic acid signaling in adult female gonads. Retinoic acid, its receptors, and the key enzymes required for retinoic acid synthesis are expressed in the ovaries and they are involved in the regulation of folliculogenesis and steroidogenesis. Exposure to compounds that can interfere with normal retinoic acid signaling is associated with adverse ovarian outcomes, including altered steroidogenesis and reduction in indicators of ovarian reserve in women and laboratory animal models. These observations call for more attention to retinoids as regulators of adult ovarian physiology and as possible targets of endocrine disruption by environmental chemicals. In this review, we summarize the current knowledge of retinoids in folliculogenesis and steroidogenesis in post-pubertal mammalian ovaries.
Collapse
|
17
|
Oocyte stage-specific effects of MTOR determine granulosa cell fate and oocyte quality in mice. Proc Natl Acad Sci U S A 2018; 115:E5326-E5333. [PMID: 29784807 PMCID: PMC6003357 DOI: 10.1073/pnas.1800352115] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
MTOR (mechanistic target of rapamycin), an integrator of pathways important for cellular metabolism, proliferation, and differentiation, is expressed at all stages of oocyte development. Primordial oocytes constitute a nonproliferating, nongrowing reserve of potential eggs maintained for the entire reproductive lifespan of mammalian females. Using conditional knockouts, we determined the role of MTOR in both primordial and growing oocytes. MTOR-dependent pathways in primordial oocytes are not needed to sustain the viability of the primordial oocyte pool or their recruitment into the cohort of growing oocytes but are essential later for maintenance of oocyte genomic integrity, sustaining ovarian follicular development, and fertility. In growing oocytes, MTOR-dependent pathways are required for processes that promote completion of meiosis and enable embryonic development. MTOR (mechanistic target of rapamycin) is a widely recognized integrator of signals and pathways key for cellular metabolism, proliferation, and differentiation. Here we show that conditional knockout (cKO) of Mtor in either primordial or growing oocytes caused infertility but differentially affected oocyte quality, granulosa cell fate, and follicular development. cKO of Mtor in nongrowing primordial oocytes caused defective follicular development leading to progressive degeneration of oocytes and loss of granulosa cell identity coincident with the acquisition of immature Sertoli cell-like characteristics. Although Mtor was deleted at the primordial oocyte stage, DNA damage accumulated in oocytes during their later growth, and there was a marked alteration of the transcriptome in the few oocytes that achieved the fully grown stage. Although oocyte quality and fertility were also compromised when Mtor was deleted after oocytes had begun to grow, these occurred without overtly affecting folliculogenesis or the oocyte transcriptome. Nevertheless, there was a significant change in a cohort of proteins in mature oocytes. In particular, down-regulation of PRC1 (protein regulator of cytokinesis 1) impaired completion of the first meiotic division. Therefore, MTOR-dependent pathways in primordial or growing oocytes differentially affected downstream processes including follicular development, sex-specific identity of early granulosa cells, maintenance of oocyte genome integrity, oocyte gene expression, meiosis, and preimplantation developmental competence.
Collapse
|
18
|
Lu H, Cui Y, Jiang L, Ge W. Functional Analysis of Nuclear Estrogen Receptors in Zebrafish Reproduction by Genome Editing Approach. Endocrinology 2017; 158:2292-2308. [PMID: 28398516 DOI: 10.1210/en.2017-00215] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/03/2017] [Indexed: 01/09/2023]
Abstract
Estrogens signal through both nuclear and membrane receptors with most reported effects being mediated via the nuclear estrogen receptors (nERs). Although much work has been reported on nERs in the zebrafish, there is a lack of direct genetic evidence for their functional roles and importance in reproduction. To address this issue, we undertook this study to disrupt all three nERs in the zebrafish, namely esr1 (ERα), esr2a (ERβII), and esr2b (ERβI), by the genome-editing technology clustered regularly interspaced short palindromic repeats and its associated nuclease (CRISPR/Cas9). Using this loss-of-function genetic approach, we successfully created three mutant zebrafish lines with each nER knocked out. In addition, we also generated all possible double and triple knockouts of the three nERs. The phenotypes of these mutants in reproduction were analyzed in all single, double, and triple nER knockouts in both females and males. Surprisingly, all three single nER mutant fish lines display normal reproductive development and function in both females and males, suggesting functional redundancy among these nERs. Further analysis of double and triple knockouts showed that nERs, especially Esr2a and Esr2b, were essential for female reproduction, and loss of these two nERs led to an arrest of folliculogenesis at previtellogenic stage II followed by sex reversal from female to male. In addition, the current study also revealed a unique role for Esr2a in follicle cell proliferation and transdifferentiation, follicle growth, and chorion formation. Taken together, this study provides the most comprehensive genetic analysis for differential functions of esr1, esr2a, and esr2b in fish reproduction.
Collapse
Affiliation(s)
- Huijie Lu
- Centre of Reproduction, Development, and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yong Cui
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Liwen Jiang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wei Ge
- Centre of Reproduction, Development, and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
19
|
Cirelli E, De Domenico E, Botti F, Massoud R, Geremia R, Grimaldi P. Effect Of Microgravity On Aromatase Expression In Sertoli Cells. Sci Rep 2017; 7:3469. [PMID: 28615629 PMCID: PMC5471225 DOI: 10.1038/s41598-017-02018-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/03/2017] [Indexed: 11/09/2022] Open
Abstract
Cytochrome P450-aromatase catalyzes estrogen biosynthesis from C19 steroids. In the testis, Sertoli cells express P450-aromatase and represent the primary source of estrogen during prepuberal age. This study focused on the effect of simulated microgravity (SM) on aromatase expression in primary mouse Sertoli cells. When cultured in Rotary Cell Culture System (RCCS), Sertoli cells, formed multicellular three dimensional spheroids (3D). Biological properties were first analyzed in terms of viability, cell cycle, expression of cytoskeletal components and growth factors in comparison to Sertoli cells cultured in spheroids at unit gravity (G). SM did not affect cell viability and proliferation, nor expression of the main cytoskeleton proteins and of growth factors like Kit Ligand (KL) and glial derived neurotrophic factor (GDNF). On the other hand, SM caused a strong increase in P450 aromatase mRNA and protein expression. Interestingly, P450-aromatase was no more inducible by 8-Br-cAMP. The presence of a functional aromatase was confirmed by enrichment of 17β-estradiol released in the medium by androgen precursors. We concluded that SM causes a significant upregulation of aromatase gene expression in Sertoli cells, leading to a consequent increase in 17β-estradiol secretion. High level of 17β-estradiol in the testis could have potentially adverse effects on male fertility and testicular cancer.
Collapse
Affiliation(s)
- Elisa Cirelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Emanuela De Domenico
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Flavia Botti
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Renato Massoud
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Raffaele Geremia
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Paola Grimaldi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
20
|
Niu X, Tyasi TL, Qin N, Liu D, Zhu H, Chen X, Zhang F, Yuan S, Xu R. Sequence variations in estrogen receptor 1 and 2 genes and their association with egg production traits in Chinese Dagu chickens. J Vet Med Sci 2017; 79:927-934. [PMID: 28420808 PMCID: PMC5447984 DOI: 10.1292/jvms.17-0014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Estrogen receptors α (ESR1) and β (ESR2) play central roles in folliculogenesis and therefore in reproductive biology. In the present study, two single nucleotide polymorphisms (SNPs) were
identified in the ESR1 and ESR2 genes using PCR-single strand conformation polymorphism (PCR-SSCP) and DNA sequencing. One of the identified SNPs, a T1101C transition located within exon 4 of the
ESR1 gene, was significantly associated with hen-housed egg production (HHEP) at 30, 43, 57 and 66 weeks of age (P<0.05), and egg weight (EW) at 30 weeks (P<0.05). Another
SNP, a G1755A transition leading to a non-synonymous substitution (valine 459-to-isoleucine) located within exon 8 of the ESR2 gene, was also markedly correlated with the HHEP at 30, 43, 57 and 66 weeks of age
(P<0.05), and EW at 30 weeks (P<0.05). A greater proportion of the additive variance was explained by the SNPs for most of the associated egg production traits (>1%). Furthermore, the
results of the combined genotype-based association analysis supported the finding that the two SNPs were associated with the traits under a study. Taken together, our findings suggest that the two sequence variations in the
ESR1 and ESR2 genes may provide promising genetic markers for the early selection and prediction of advantageous phenotypes in chicken breeding.
Collapse
Affiliation(s)
- Xiaotian Niu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Thobela Louis Tyasi
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Ning Qin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Dehui Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Hongyan Zhu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Xiaoxia Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Fengwei Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Shuguo Yuan
- Jilin Grain Group Agriculture and Livestock Co., Ltd., Changchun 130062, Jilin, China
| | - Rifu Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin, China
| |
Collapse
|
21
|
Abstract
The hormone estrogen is involved in both female and male reproduction, as well as numerous other biological systems including the neuroendocrine, vascular, skeletal, and immune systems. Therefore, it is also implicated in many different diseases and conditions such as infertility, obesity, osteoporosis, endometriosis, and a variety of cancers. Estrogen works through its two distinct nuclear receptors, estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). Various transcriptional regulation mechanisms have been identified as the mode of action for estrogen, mainly the classical mechanism with direct DNA binding but also a nongenomic mode of action and one using tethered or indirect binding. The expression profiles of ERα and ERβ are unique with the primary sites of ERα expression being the uterus and pituitary gland and the main site of ERβ expression being the granulosa cells of the ovary. Mouse models with knockout or mutation of Esr1 and Esr2 have furthered our understanding of the role of each individual receptor plays in physiology. From these studies, it is known that the primary roles for ERα are in the uterus and neuroendocrine system, as female mice lacking ERα are infertile due to impaired ovarian and uterine function, whereas female mice lacking ERβ are subfertile due to ovarian defects. The development of effective therapies for estrogen-related diseases has relied on an understanding of the physiological roles and mechanistic functionalities of ERα and ERβ in human health and disease.
Collapse
Affiliation(s)
- Katherine J Hamilton
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, United States
| | - Sylvia C Hewitt
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, United States
| | - Yukitomo Arao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, United States
| | - Kenneth S Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, United States.
| |
Collapse
|
22
|
SCA-1 Labels a Subset of Estrogen-Responsive Bipotential Repopulating Cells within the CD24 + CD49f hi Mammary Stem Cell-Enriched Compartment. Stem Cell Reports 2017; 8:417-431. [PMID: 28132885 PMCID: PMC5312257 DOI: 10.1016/j.stemcr.2016.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 12/26/2022] Open
Abstract
Estrogen stimulates breast development during puberty and mammary tumors in adulthood through estrogen receptor-α (ERα). These effects are proposed to occur via ERα+ luminal cells and not the mammary stem cells (MaSCs) that are ERαneg. Since ERα+ luminal cells express stem cell antigen-1 (SCA-1), we sought to determine if SCA-1 could define an ERα+ subset of EpCAM+/CD24+/CD49fhi MaSCs. We show that the MaSC population has a distinct SCA-1+ population that is abundant in pre-pubertal mammary glands. The SCA-1+ MaSCs have less stem cell markers and less in vivo repopulating activity than their SCA-1neg counterparts. However, they express ERα and specifically enter the cell cycle at puberty. Using estrogen-deficient aromatase knockouts (ArKO), we showed that the SCA-1+ MaSC could be directly modulated by estrogen supplementation. Thus, SCA-1 enriches for an ERα+, estrogen-sensitive subpopulation within the CD24+/CD49fhi MaSC population that may be responsible for the hormonal sensitivity of the developing mammary gland. SCA-1+ delineates ER-positive cells in the CD24+ CD49fhi mammary stem population SCA-1+ cells have lower repopulation activity SCA-1+ cells are estrogen responsive
Collapse
|
23
|
Soleimani Mehranjani M, Mansoori T. Stereological study on the effect of vitamin C in preventing the adverse effects of bisphenol A on rat ovary. Int J Reprod Biomed 2016. [DOI: 10.29252/ijrm.14.6.403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
24
|
Abstract
Estrogen receptor alpha (ERα) is a critical player in development and function of the female reproductive system. Perturbations in ERα response can affect wide-ranging aspects of health in humans as well as in livestock and wildlife. Because of its long-known and broad impact, ERα mechanisms of action continue to be the focus on cutting-edge research efforts. Consequently, novel insights have greatly advanced understanding of every aspect of estrogen signaling. In this review, we attempt to briefly outline the current understanding of ERα mediated mechanisms in the context of the female reproductive system.
Collapse
Affiliation(s)
- Sylvia C Hewitt
- Receptor Biology GroupReproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USASchool of Molecular BiosciencesCollege of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| | - Wipawee Winuthayanon
- Receptor Biology GroupReproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USASchool of Molecular BiosciencesCollege of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| | - Kenneth S Korach
- Receptor Biology GroupReproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USASchool of Molecular BiosciencesCollege of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| |
Collapse
|
25
|
Abstract
Current knowledge on gonadal development and sex determination is the product of many decades of research involving a variety of scientific methods from different biological disciplines such as histology, genetics, biochemistry, and molecular biology. The earliest embryological investigations, followed by the invention of microscopy and staining methods, were based on histological examinations. The most robust development of histological staining techniques occurred in the second half of the nineteenth century and resulted in structural descriptions of gonadogenesis. These first studies on gonadal development were conducted on domesticated animals; however, currently the mouse is the most extensively studied species. The next key point in the study of gonadogenesis was the advancement of methods allowing for the in vitro culture of fetal gonads. For instance, this led to the description of the origin of cell lines forming the gonads. Protein detection using antibodies and immunolabeling methods and the use of reporter genes were also invaluable for developmental studies, enabling the visualization of the formation of gonadal structure. Recently, genetic and molecular biology techniques, especially gene expression analysis, have revolutionized studies on gonadogenesis and have provided insight into the molecular mechanisms that govern this process. The successive invention of new methods is reflected in the progress of research on gonadal development.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| |
Collapse
|
26
|
|
27
|
Yu M, Liu W, Wang J, Qin J, Wang Y, Wang Y. Effects of tamoxifen on autosomal genes regulating ovary maintenance in adult mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:20234-20244. [PMID: 26304810 DOI: 10.1007/s11356-015-5245-5] [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: 05/14/2015] [Accepted: 08/13/2015] [Indexed: 06/04/2023]
Abstract
Environmental endocrine-disrupting chemicals (EDCs), known to bind to estrogen/androgen receptors and mimic native estrogens, have been implicated as a main source for increasing human reproductive and developmental deficiencies and diseases. Tamoxifen (TAM) is one of the most well-known antiestrogens with defined adverse effects on the female reproductive tract, but the mechanisms related to autosomal gene regulation governing ovary maintenance in mammals remain unclear. The expression pattern and levels of key genes and proteins involved in maintaining the ovarian phenotype in mice were analyzed. The results showed that TAM induced significant upregulation of Sox9, which is the testis-determining factor gene. The results showed that TAM induced significant upregulation of Sox9, the testis-determining factor gene, and the expression level of Sox9 mRNA in the ovaries of mice exposed to 75 or 225 mg/kg bw TAM was 2- and 10-fold that in the control group, respectively (p < 0.001). Furthermore, the testicular fibroblast growth factor gene, Fgf9, was also elevated in TAM-treated ovaries. Accordingly, expression of the ovary development marker, forkhead transcription factor (FOXL2), and WNT4/FST signaling, were depressed. The levels of protein expression changed consistently with the target genes. Moreover, the detection of platelet/endothelial cell adhesion molecule 1 (PECAM-1) in TAM-treated ovaries suggested the formation of vascular endothelial cells, which is a further evidence for the differentiation of the ovaries to a testis-like phenotype. During this period, the level of 17β-estradiol, progesterone, and luteinizing hormone decreased, while that of testosterone increased by 3.3-fold (p = 0.013). The activation of a testis-specific molecular signaling cascade was a potentially important mechanism contributing to the gender disorder induced by TAM, which resulted in the differentiation of the ovaries to a testis-like phenotype in adult mice. Limited with a relatively higher exposure, the present study provided preliminary molecular insights into the sexual disorder induced by antiestrogens and compounds that interrupted estrogen signaling by other modes of action.
Collapse
Affiliation(s)
- Mingxi Yu
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, No. 2 Linggong Road, 116024, Dalian, Liaoning, China
| | - Wei Liu
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, No. 2 Linggong Road, 116024, Dalian, Liaoning, China
| | - Jingyun Wang
- School of Life Science and Biotechnology, Dalian University of Technology, No. 2 Linggong Road, 116024, Dalian, Liaoning, China.
| | - Junwen Qin
- Department of Developmental and Regenerative Biology, Key Laboratory for Regenerative Medicine (MOE), Jinan University, No. 601 West Huangpu Road, 510632, Guangzhou, Guangdong, China
| | - Yongan Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, No. 2 Linggong Road, 116024, Dalian, Liaoning, China
| | - Yu Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environmental Science and Technology, Dalian University of Technology, No. 2 Linggong Road, 116024, Dalian, Liaoning, China
| |
Collapse
|
28
|
Simão VA, Berloffa Belardin L, Araújo Leite GA, de Almeida Chuffa LG, Camargo ICC. Effects of different doses of nandrolone decanoate on estrous cycle and ovarian tissue of rats after treatment and recovery periods. Int J Exp Pathol 2015; 96:338-49. [PMID: 26575430 DOI: 10.1111/iep.12144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/22/2015] [Indexed: 01/17/2023] Open
Abstract
This study tested the hypothesis that different doses of nandrolone decanoate (ND) will cause changes in the estrous cycle and ovarian tissue of adult rats; and investigated the duration of the recovery period that is sufficient to restore the damage in the animals treated with different doses. Wistar rats were treated with ND at doses of 1.87, 3.75, 7.5 and 15 mg/kg body weight, or received mineral oil (control group) for 15 days, subcutaneously. All animals were divided into three groups according to the treatment periods: (i) ND treatment for 15 days; (ii) ND treatment followed by a 30-day recovery; and (iii) ND treatment followed by a 60-day recovery. Estrous cycle was monitored daily, and at the end of each period, the animals were euthanized for histopathological analysis. During ND treatment and after 30-day recovery, all animals exhibited persistent diestrus. After a 60-day recovery, persistent diestrus was only maintained in the group that had received the highest dose. Ovarian weight was decreased significantly after the 30-day recovery, regardless of ND doses, compared with the control group. There was a reduction (P < 0.05) in the number of corpora lutea and antral and growing follicles, in contrast to an increase (P < 0.05) in atretic follicles in a dose- and time-dependent manner. Remarkable histopathological changes occurred in the ovaries of all ND-treated groups. In conclusion, the different doses of ND caused changes in the estrous cycle and ovarian tissue of rats, and recovery periods (30 and 60 days) were insufficient to completely restore the damage in the animals treated with the highest dose.
Collapse
Affiliation(s)
- Vinícius Augusto Simão
- Department of Biological Sciences, Faculty of Sciences and Letters, Univ Estadual Paulista - UNESP, Assis, Brazil
| | - Larissa Berloffa Belardin
- Department of Biological Sciences, Faculty of Sciences and Letters, Univ Estadual Paulista - UNESP, Assis, Brazil
| | - Gabriel Adan Araújo Leite
- Department of Biological Sciences, Faculty of Sciences and Letters, Univ Estadual Paulista - UNESP, Assis, Brazil
| | | | | |
Collapse
|
29
|
Bazzano M, Torelli C, Pustovrh M, Paz D, Elia E. Obesity induced by cafeteria diet disrupts fertility in the rat by affecting multiple ovarian targets. Reprod Biomed Online 2015; 31:655-67. [DOI: 10.1016/j.rbmo.2015.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/03/2015] [Accepted: 08/06/2015] [Indexed: 11/24/2022]
|
30
|
Bagheri Ziari S, Naji T, Hosseinzadeh Sahafi H. Comparison of the effects of Origanum vulgare with LHRH-A2 and 17β-estradiol on the ultrastructure of gonadotroph cells and ovarian oogenesis in immature Trichogaster trichopterus. Anim Reprod Sci 2015; 161:32-9. [DOI: 10.1016/j.anireprosci.2015.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 11/26/2022]
|
31
|
Toda K, Hayashi Y, Ono M, Saibara T. Co-administration of insulin with a gonadotropin partly improves ovulatory responses of estrogen-deficient mice. Mol Cell Endocrinol 2015; 411:177-86. [PMID: 25957088 DOI: 10.1016/j.mce.2015.04.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/27/2015] [Accepted: 04/29/2015] [Indexed: 12/30/2022]
Abstract
Administration of 17-βestradiol (E2) with pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) can induce ovulation in estrogen-deficient (ArKO) mice; nevertheless, ovulatory efficiency and rate are low. In this study, effects of insulin on the ovulatory responses were investigated. In ArKO ovary, hCG signal was found to be transmitted in an uncoordinated manner when phosphorylation levels of signaling molecules are examined. Co-administration of insulin with hCG improved the transmission of hCG signal as well as the ovulatory efficiency in ArKO mice. It also improved the ovulatory rate but far below the wild-type rate. Gene expression analysis demonstrated that Cyp11a1 and Cyp17a1 mRNAs were significantly induced 4 h after PMSG administration in the wild-type ovary, but not in ArKO ovary. Collectively, these results suggest that insulin improves ovulatory responses of ArKO mice, but it fails to ameliorate follicular dysfunctions caused possibly by an inappropriate intraovarian milieu during follicular maturation.
Collapse
Affiliation(s)
- Katsumi Toda
- Department of Biochemistry, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan.
| | - Yoshihiro Hayashi
- Department of Pathology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Masafumi Ono
- Department of Gastroenterology and Hepatology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Toshiji Saibara
- Department of Gastroenterology and Hepatology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| |
Collapse
|
32
|
Lin YT, Capel B. Cell fate commitment during mammalian sex determination. Curr Opin Genet Dev 2015; 32:144-52. [PMID: 25841206 DOI: 10.1016/j.gde.2015.03.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/24/2015] [Accepted: 03/05/2015] [Indexed: 01/10/2023]
Abstract
The gonads form bilaterally as bipotential organs that can develop as testes or ovaries. All secondary sex characteristics that we associate with 'maleness' or 'femaleness' depend on whether testes or ovaries form. The fate of the gonads depends on a cell fate decision that occurs in a somatic cell referred to as the 'supporting cell lineage'. Once supporting cell progenitors commit to Sertoli (male) or granulosa (female) fate, they propagate this decision to the other cells within the organ. In this review, we will describe what is known about the bipotential state of somatic and germ cell lineages in the gonad and the transcriptional and antagonistic signaling networks that lead to commitment, propagation, and maintenance of testis or ovary fate.
Collapse
Affiliation(s)
- Yi-Tzu Lin
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Blanche Capel
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
33
|
Dezfouli MGZ, Eissazadeh S, Zade SMAS. Histological and histometrical study of the protective role of α-tocopherol against sodium arsenite toxicity in rat ovaries. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2014; 20:1167-1179. [PMID: 24735566 DOI: 10.1017/s1431927614000701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
This study examines histometrical changes induced by sodium arsenite (SA), as an environmental pollutant, and investigates the protective effect of α-tocopherol on ovaries of SA-treated rats during the prenatal stage until sexual maturity. Rats were classified into groups: control, SA (8 ppm/day), α-tocopherol (100 ppm/day), and SA+α-tocopherol. Treatment was performed from pregnancy until maturation when the rats and ovaries were weighed. The Cavalieri method was used to estimate volume of the ovaries, cortex, medulla, and corpus luteum. The mean diameter of oocytes, granulosa cells, and nuclei were measured and volume was estimated using the Nucleator method. The number of oocytes and thickness of the zona pellucida (ZP) were determined using an optical dissector and orthogonal intercept method, respectively. SA reduced the body and ovary weight, the number of secondary, antral and Graafian oocytes, volume of the ovaries, cortex, medulla and corpus luteum, mean diameter and volume of oocytes in primordial and primary follicles, mean diameter and volume of oocyte nuclei in all types of follicles, and mean thickness of the ZP in secondary and antral follicles. Also, the mean diameter and volume of granulosa cells and their nuclei in antral and Graafian follicles decreased significantly. Vacuolization and vascular congestion in the corpus luteum and an increase in the number of atretic oocytes were seen in the SA group. Most of these parameters were unchanged from the control level in the SA+α-tocopherol group. It was concluded that α-tocopherol supplementation reduced the toxic effects of SA exposure on ovarian tissue in rats.
Collapse
Affiliation(s)
- Maryam Ghandi Zadeh Dezfouli
- 1Department of Biology, Faculty of Basic Science,Arak University,Shahid Beheshti Street,Arak,Markazi Province,3945-5-38138Iran
| | - Samira Eissazadeh
- 2Department of Cellular and Molecular Biology, Faculty of Biotechnology and Biomolecular Science,Serding,Selangor,Darul Ehsan,43400Malaysia
| | - Sayed Mohammad Ali Shariat Zade
- 1Department of Biology, Faculty of Basic Science,Arak University,Shahid Beheshti Street,Arak,Markazi Province,3945-5-38138Iran
| |
Collapse
|
34
|
Jorgensen JS. Defining the neighborhoods that escort the oocyte through its early life events and into a functional follicle. Mol Reprod Dev 2013; 80:960-76. [PMID: 24105719 PMCID: PMC3980676 DOI: 10.1002/mrd.22232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/15/2013] [Indexed: 01/19/2023]
Abstract
The ovary functions to chaperone the most precious cargo for female individuals, the oocyte, thereby allowing the passage of genetic material to subsequent generations. Within the ovary, single oocytes are surrounded by a legion of granulosa cells inside each follicle. These two cell types depend upon one another to support follicle formation and oocyte survival. The infrastructure and events that work together to ultimately form these functional follicles within the ovary are unprecedented, given that the oocyte originates as a cell like all other neighboring cells within the embryo prior to gastrulation. This review discusses the journey of the germ cell in the context of the developing female mouse embryo, with a focus on specific signaling events and cell-cell interactions that escort the primordial germ cell as it is specified into the germ cell fate, migrates through the hindgut into the gonad, differentiates into an oocyte, and culminates upon formation of the primordial and then primary follicle.
Collapse
Affiliation(s)
- Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
35
|
Disruption of mitotic arrest precedes precocious differentiation and transdifferentiation of pregranulosa cells in the perinatal Wnt4 mutant ovary. Dev Biol 2013; 383:295-306. [PMID: 24036309 DOI: 10.1016/j.ydbio.2013.08.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 11/21/2022]
Abstract
Mammalian sex determination is controlled by antagonistic pathways that are initially co-expressed in the bipotential gonad and subsequently become male- or female-specific. In XY gonads, testis development is initiated by upregulation of Sox9 by SRY in pre-Sertoli cells. Disruption of either gene leads to complete male-to-female sex reversal. Ovarian development is dependent on canonical Wnt signaling through Wnt4, Rspo1 and β-catenin. However, only a partial female-to-male sex reversal results from disruption of these ovary-promoting genes. In Wnt4 and Rspo1 mutants, there is evidence of pregranulosa cell-to-Sertoli cell transdifferentiation near birth, following a severe decline in germ cells. It is currently unclear why primary sex reversal does not occur at the sex-determining stage, but instead occurs near birth in these mutants. Here we show that Wnt4-null and Rspo1-null pregranulosa cells transition through a differentiated granulosa cell state prior to transdifferentiating towards a Sertoli cell fate. This transition is preceded by a wave of germ cell death that is closely associated with the disruption of pregranulosa cell quiescence. Our results suggest that maintenance of mitotic arrest in pregranulosa cells may preclude upregulation of Sox9 in cases where female sex-determining genes are disrupted. This may explain the lack of complete sex reversal in such mutants at the sex-determining stage.
Collapse
|
36
|
Cutting A, Chue J, Smith CA. Just how conserved is vertebrate sex determination? Dev Dyn 2013; 242:380-7. [PMID: 23390004 DOI: 10.1002/dvdy.23944] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Sex determination in vertebrate embryos has long been equated with gonadal differentiation into testes or ovaries. This view has been challenged over the years by reports of somatic sexual dimorphisms pre-dating gonadal sex differentiation. The recent finding that sex determination in birds is likely to be partly cell autonomous has again called for a broader definition of sex determination. Inherent sexual differentiation in each and every cell may apply widely among vertebrates, and may involve more than one "master sex gene" on a sex chromosome. At the gonadal level, key genes required for proper sexual differentiation are conserved among vertebrates, but their relative positions in the ovarian and testicular cascades differ. RESULTS We illustrate these differences by comparing key sex genes in fishes versus birds and mammals, with emphasis on DM domain genes and the SOX9-AMH pathway in the testis and the FOXL2-Aromatase pathway in the ovary. Such comparisons facilitate the identification of ancient versus derived genes involved in gonadal sex determination. CONCLUSIONS The data indicate that vertebrate sex-determining cascades are not as conserved as once thought.
Collapse
Affiliation(s)
- Andrew Cutting
- Murdoch Childrens Research Institute, Royal Childrens Hospital, Parkville, Australia
| | | | | |
Collapse
|
37
|
Germ cells are not required to establish the female pathway in mouse fetal gonads. PLoS One 2012; 7:e47238. [PMID: 23091613 PMCID: PMC3473035 DOI: 10.1371/journal.pone.0047238] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/10/2012] [Indexed: 01/26/2023] Open
Abstract
The fetal gonad is composed of a mixture of somatic cell lineages and germ cells. The fate of the gonad, male or female, is determined by a population of somatic cells that differentiate into Sertoli or granulosa cells and direct testis or ovary development. It is well established that germ cells are not required for the establishment or maintenance of Sertoli cells or testis cords in the male gonad. However, in the agametic ovary, follicles do not form suggesting that germ cells may influence granulosa cell development. Prior investigations of ovaries in which pre-meiotic germ cells were ablated during fetal life reported no histological changes during stages prior to birth. However, whether granulosa cells underwent normal molecular differentiation was not investigated. In cases where germ cell loss occurred secondary to other mutations, transdifferentiation of granulosa cells towards a Sertoli cell fate was observed, raising questions about whether germ cells play an active role in establishing or maintaining the fate of granulosa cells. We developed a group of molecular markers associated with ovarian development, and show here that the loss of pre-meiotic germ cells does not disrupt the somatic ovarian differentiation program during fetal life, or cause transdifferentiation as defined by expression of Sertoli markers. Since we do not find defects in the ovarian somatic program, the subsequent failure to form follicles at perinatal stages is likely attributable to the absence of germ cells rather than to defects in the somatic cells.
Collapse
|
38
|
Ditewig AC, Yao HHC. Organogenesis of the ovary: a comparative review on vertebrate ovary formation. Organogenesis 2012; 2:36-41. [PMID: 19521565 DOI: 10.4161/org.2.2.2491] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Accepted: 01/10/2006] [Indexed: 11/19/2022] Open
Abstract
The general perspective of ovary organogenesis is that the ovary is the default organ which develops in the absence of testis-promoting factors. Testis formation, on the other hand, is a male-specific event promoted by active components that override the default ovarian process. However, when comparing the sex determination mechanism among different vertebrate species, it is apparent that this default view of ovary formation can only be applied to mammals. In species such as reptiles and birds, ovary formation is an active process stimulated by estrogen. Remnants of this estrogen-dominant pathway are still present in marsupials, a close relative of eutherian mammals, like humans and mice. Although initial formation of the mammalian ovary has become strictly regulated by genetic components and is therefore independent of estrogen, the feminizing effect of estrogen regains its command in adult ovaries. When estrogen production, or its signaling, is inhibited, transdifferentiation of ovarian tissues to testis structures occur in adult females. Taken together, these observations prompt us to reconsider the process of ovary organogenesis as the default organ and question if testis development is actually the default pathway.
Collapse
Affiliation(s)
- Amy C Ditewig
- Department of Veterinary Biosciences; University of Illinois; Urbana, Illinois USA
| | | |
Collapse
|
39
|
Mork L, Maatouk DM, McMahon JA, Guo JJ, Zhang P, McMahon AP, Capel B. Temporal differences in granulosa cell specification in the ovary reflect distinct follicle fates in mice. Biol Reprod 2012; 86:37. [PMID: 21976597 DOI: 10.1095/biolreprod.111.095208] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The embryonic origins of ovarian granulosa cells have been a subject of debate for decades. By tamoxifen-induced lineage tracing of Foxl2-expressing cells, we show that descendants of the bipotential supporting cell precursors in the early gonad contribute granulosa cells to a specific population of follicles in the medulla of the ovary that begin to grow immediately after birth. These precursor cells arise from the proliferative ovarian surface epithelium and enter mitotic arrest prior to upregulating Foxl2. Granulosa cells that populate the cortical primordial follicles activated in adult life derive from the surface epithelium perinatally, and enter mitotic arrest at that stage. Ingression from the surface epithelium dropped to undetectable levels by Postnatal Day 7, when most surviving oocytes were individually encapsulated by granulosa cells. These findings add complexity to the standard model of sex determination in which the Sertoli and granulosa cells of the adult testis and ovary directly stem from the supporting cell precursors of the bipotential gonad.
Collapse
Affiliation(s)
- Lindsey Mork
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Toda K, Hayashi Y, Ono M, Saibara T. Impact of ovarian sex steroids on ovulation and ovulatory gene induction in aromatase-null mice. Endocrinology 2012; 153:386-94. [PMID: 22147013 DOI: 10.1210/en.2011-1462] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Female mice deficient in the aromatase gene [aromatase knockout (ArKO)] fail to ovulate owing to an inability to produce estrogens. Here, we demonstrated that sequential administration of adequate amounts of 17β-estradiol (E2), pregnant mare serum gonadotropin, and human chorionic gonadotropin could induce ovulation in immature ArKO mice; nevertheless, significantly fewer oocytes were released into the oviducts in ArKO mice than in wild-type mice. Analysis of ovarian steroids by liquid chromatography coupled with electrospray ionization-tandem mass spectrometry identified a trace amount of E2 in the untreated immature ArKO ovary. The analysis further detected significant increases and decreases in progesterone and testosterone contents, respectively, in addition to an increase of E2 in the ovulation-induced ArKO ovaries compared with the levels in untreated ArKO ovaries. Gene expression analysis demonstrated marked elevation in the mRNA levels of members of the epidermal growth factor family and extracellular matrix-related proteins at 4 h after human chorionic gonadotropin injection in the ovaries of ArKO mice treated for ovulation, as observed in the ovulation-induced wild-type ovaries. Collectively, these findings suggest the vital contribution of the intraovarian milieu of sex steroids to ovulatory regulation in vivo.
Collapse
Affiliation(s)
- Katsumi Toda
- Department of Biochemistry, Kochi University, School of Medicine, Nankoku, Kochi 783-8505, Japan.
| | | | | | | |
Collapse
|
41
|
|
42
|
Pannetier M, Elzaiat M, Thépot D, Pailhoux E. Telling the story of XX sex reversal in the goat: highlighting the sex-crossroad in domestic mammals. Sex Dev 2011; 6:33-45. [PMID: 22094227 DOI: 10.1159/000334056] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The conditions for sex reversal in vertebrate species have been studied extensively and have highlighted numerous key factors involved in sex differentiation. We review here the history of the development of knowledge, referring to one example of complete female-to-male XX sex reversal associated with a polled phenotype in the goat. The results and hypotheses concerning this polled intersex syndrome (PIS) are then presented, firstly with respect to the transcriptional regulatory effects of the PIS mutation, and secondly regarding the role of the main ovarian-differentiating factor in this PIS locus, the FOXL2 gene.
Collapse
Affiliation(s)
- M Pannetier
- INRA, UMR 1198, ENVA, Biologie du Développement et de la Reproduction, Jouy-en-Josas, France
| | | | | | | |
Collapse
|
43
|
Nicholls C, Li H, Wang JQ, Liu JP. Molecular regulation of telomerase activity in aging. Protein Cell 2011; 2:726-38. [PMID: 21976062 DOI: 10.1007/s13238-011-1093-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Accepted: 08/30/2011] [Indexed: 11/25/2022] Open
Abstract
The process of aging is mitigated by the maintenance and repair of chromosome ends (telomeres), resulting in extended lifespan. This review examines the molecular mechanisms underlying the actions and regulation of the enzyme telomerase reverse transcriptase (TERT), which functions as the primary mechanism of telomere maintenance and regulates cellular life expectancy. Underpinning increased cell proliferation, telomerase is also a key factor in facilitating cancer cell immortalization. The review focuses on aspects of hormonal regulations of telomerase, and the intracellular pathways that converge to regulate telomerase activity with an emphasis on molecular interactions at protein and gene levels. In addition, the basic structure and function of two key telomerase enzyme components-the catalytic subunit TERT and the template RNA (TERC) are discussed briefly.
Collapse
Affiliation(s)
- Craig Nicholls
- Molecular Signalling Laboratory, Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia
| | | | | | | |
Collapse
|
44
|
Bayne S, Li H, Jones MEE, Pinto AR, van Sinderen M, Drummond A, Simpson ER, Liu JP. Estrogen deficiency reversibly induces telomere shortening in mouse granulosa cells and ovarian aging in vivo. Protein Cell 2011; 2:333-46. [PMID: 21574023 DOI: 10.1007/s13238-011-1033-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 03/21/2011] [Indexed: 12/31/2022] Open
Abstract
Estrogen is implicated as playing an important role in aging and tumorigenesis of estrogen responsive tissues; however the mechanisms underlying the mitogenic actions of estrogen are not fully understood. Here we report that estrogen deficiency in mice caused by targeted disruption of the aromatase gene results in a significant inhibition of telomerase maintenance of telomeres in mouse ovaries in a tissue-specific manner. The inhibition entails a significant shortening of telomeres and compromised proliferation in the follicular granulosa cell compartment of ovary. Gene expression analysis showed decreased levels of proto-oncogene c-Myc and the telomerase catalytic subunit, telomerase reverse transcriptase (TERT), in response to estrogen deficiency. Estrogen replacement therapy led to increases in TERT gene expression, telomerase activity, telomere length and ovarian tissue growth, thereby reinstating ovary development to normal in four weeks. Our data demonstrate for the first time that telomere maintenance is the primary mechanism mediating the mitogenic effect of estrogen on ovarian granulosa cell proliferation by upregulating the genes of c-Myc and TERT in vivo. Estrogen deficiency or over-activity may cause ovarian tissue aging or tumorigenesis, respectively, through estrogen regulation of telomere remodeling.
Collapse
Affiliation(s)
- Sharyn Bayne
- Department of Immunology, Central Eastern Clinical School, Monash University, Victoria 3800, Australia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Liew SH, Sarraj MA, Drummond AE, Findlay JK. Estrogen-dependent gene expression in the mouse ovary. PLoS One 2011; 6:e14672. [PMID: 21347412 PMCID: PMC3036593 DOI: 10.1371/journal.pone.0014672] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Accepted: 10/29/2010] [Indexed: 01/01/2023] Open
Abstract
Estrogen (E) plays a pivotal role in regulating the female reproductive system, particularly the ovary. However, the number and type of ovarian genes influenced by estrogen remain to be fully elucidated. In this study, we have utilized wild-type (WT) and aromatase knockout (ArKO; estrogen free) mouse ovaries as an in vivo model to profile estrogen dependent genes. RNA from each individual ovary (n = 3) was analyzed by a microarray-based screen using Illumina Sentrix Mouse WG-6 BeadChip (45,281 transcripts). Comparative analysis (GeneSpring) showed differential expression profiles of 450 genes influenced by E, with 291 genes up-regulated and 159 down-regulated by 2-fold or greater in the ArKO ovary compared to WT. Genes previously reported to be E regulated in ArKO ovaries were confirmed, in addition to novel genes not previously reported to be expressed or regulated by E in the ovary. Of genes involved in 5 diverse functional processes (hormonal processes, reproduction, sex differentiation and determination, apoptosis and cellular processes) 78 had estrogen-responsive elements (ERE). These analyses define the transcriptome regulated by E in the mouse ovary. Further analysis and investigation will increase our knowledge pertaining to how E influences follicular development and other ovarian functions.
Collapse
Affiliation(s)
- Seng H Liew
- Prince Henry's Institute of Medical Research, Clayton, Melbourne, Australia.
| | | | | | | |
Collapse
|
46
|
Pask AJ, Calatayud NE, Shaw G, Wood WM, Renfree MB. Oestrogen blocks the nuclear entry of SOX9 in the developing gonad of a marsupial mammal. BMC Biol 2010; 8:113. [PMID: 20807406 PMCID: PMC2940779 DOI: 10.1186/1741-7007-8-113] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 08/31/2010] [Indexed: 11/10/2022] Open
Abstract
Background Hormones are critical for early gonadal development in nonmammalian vertebrates, and oestrogen is required for normal ovarian development. In contrast, mammals determine sex by the presence or absence of the SRY gene, and hormones are not thought to play a role in early gonadal development. Despite an XY sex-determining system in marsupial mammals, exposure to oestrogen can override SRY and induce ovarian development of XY gonads if administered early enough. Here we assess the effect of exogenous oestrogen on the molecular pathways of mammalian gonadal development. Results We examined the expression of key testicular (SRY, SOX9, AMH and FGF9) and ovarian (WNT4, RSPO1, FOXL2 and FST) markers during gonadal development in the marsupial tammar wallaby (Macropus eugenii) and used these data to determine the effect of oestrogen exposure on gonadal fate. During normal development, we observed male specific upregulation of AMH and SOX9 as in the mouse and human testis, but this upregulation was initiated before the peak in SRY expression and 4 days before testicular cord formation. Similarly, key genes for ovarian development in mouse and human were also upregulated during ovarian differentiation in the tammar. In particular, there was early sexually dimorphic expression of FOXL2 and WNT4, suggesting that these genes are key regulators of ovarian development in all therian mammals. We next examined the effect of exogenous oestrogen on the development of the mammalian XY gonad. Despite the presence of SRY, exogenous oestrogen blocked the key male transcription factor SOX9 from entering the nuclei of male somatic cells, preventing activation of the testicular pathway and permitting upregulation of key female genes, resulting in ovarian development of the XY gonad. Conclusions We have uncovered a mechanism by which oestrogen can regulate gonadal development through the nucleocytoplasmic shuttling of SOX9. This may represent an underlying ancestral mechanism by which oestrogen promotes ovarian development in the gonads of nonmammalian vertebrates. Furthermore, oestrogen may retain this function in adult female mammals to maintain granulosa cell fate in the differentiated ovary by suppressing nuclear translocation of the SOX9 protein. See commentary: http://www.biomedcentral.com/1741-7007/8/110
Collapse
Affiliation(s)
- Andrew J Pask
- Department of Molecular and Cellular Biology, University of Connecticut, Storrs, CT 06260, USA.
| | | | | | | | | |
Collapse
|
47
|
Guiguen Y, Fostier A, Piferrer F, Chang CF. Ovarian aromatase and estrogens: a pivotal role for gonadal sex differentiation and sex change in fish. Gen Comp Endocrinol 2010; 165:352-66. [PMID: 19289125 DOI: 10.1016/j.ygcen.2009.03.002] [Citation(s) in RCA: 433] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 02/23/2009] [Accepted: 03/03/2009] [Indexed: 10/21/2022]
Abstract
The present review focuses on the roles of estrogens and aromatase (Cyp19a1a), the enzyme needed for their synthesis, in fish gonadal sex differentiation. Based on the recent literature, we extend the already well accepted hypothesis of an implication of estrogens and Cyp19a1a in ovarian differentiation to a broader hypothesis that would place estrogens and Cyp19a1a in a pivotal position to control not only ovarian, but also testicular differentiation, in both gonochoristic and hermaphrodite fish species. This working hypothesis states that cyp19a1a up-regulation is needed not only for triggering but also for maintaining ovarian differentiation and that cyp19a1a down-regulation is the only necessary step for inducing a testicular differentiation pathway. When considering arguments for and against, most of the information available for fish supports this hypothesis since either suppression of cyp19a1a gene expression, inhibition of Cyp19a1a enzymatic activity, or blockage of estrogen receptivity are invariably associated with masculinization. This is also consistent with reports on normal gonadal differentiation, and steroid-modulated masculinization with either androgens, aromatase inhibitors or estrogen receptor antagonists, temperature-induced masculinization and protogynous sex change in hermaphrodite species. Concerning the regulation of fish cyp19a1a during gonadal differentiation, the transcription factor foxl2 has been characterized as an ovarian specific upstream regulator of a cyp19a1a promoter that would co-activate cyp19a1a expression, along with some additional partners such as nr5a1 (sf1) or cAMP. In contrast, upstream factors potentially down-regulating cyp19a1a during testicular differentiation are still hypothetical, such as the dmrt1 gene, but their definitive characterization as testicular repressors of cyp19a1a would strongly strengthen the hypothesis that early testicular differentiation would need active repression of cyp19a1a expression.
Collapse
Affiliation(s)
- Yann Guiguen
- INRA, UR1037 SCRIBE, IFR140, Ouest-Genopole, F-35000 Rennes, France.
| | | | | | | |
Collapse
|
48
|
Sarraj MA, Escalona RM, Umbers A, Chua HK, Small C, Griswold M, Loveland K, Findlay JK, Stenvers KL. Fetal testis dysgenesis and compromised Leydig cell function in Tgfbr3 (beta glycan) knockout mice. Biol Reprod 2009; 82:153-62. [PMID: 19696014 DOI: 10.1095/biolreprod.109.078766] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Betaglycan (Tgfbr3) is a coreceptor for transforming growth factor-beta (TGFB) superfamily ligands. In the current study, a defect in seminiferous cord formation was detected in 12.5-13.5 days postcoitum (dpc) beta glycan null murine testis. Immunohistochemistry with antibodies against cell-specific markers revealed defects in somatic cell populations. To confirm these data, quantitative real-time PCR was performed to determine changes in the expression levels of genes involved in fetal testis cell differentiation and function. The expression levels of the Leydig cell markers Insl3, Cyp17a1, Cyp11a1, Star, and Hsd3b1 were reduced in knockout testis compared to wild-type testis, beginning at 12.5 dpc. Whole mount in situ hybridization confirmed that Cyp11a1 expression was reduced in the null testis, but its distribution pattern was unchanged. Apoptosis was not affected by the loss of beta glycan, but proliferation within the interstitium was reduced at 14.5 dpc. However, morphometric analysis showed no changes in Leydig cell counts between the wild-type and the knockout testes at 14.5 dpc, indicating that fetal Leydig function, rather than number, was affected by the loss of beta glycan. The expression levels of Sertoli cell markers Dhh, Sox9, and Amh were also reduced in the knockout testis at 14.5 dpc. However, the expression of fetal germ cell markers Pou5f1 and DDX4 were not changed across the genotypes at any age examined. Our data show that the presence of beta glycan is required for normal cord formation, normal fetal Leydig cell development, and the establishment of fetal testis endocrine function, thus implicating TGFB superfamily members as regulators of early fetal testis structure and function.
Collapse
Affiliation(s)
- Mai A Sarraj
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
1. Oestrogen plays an important role in ageing and ageing-related development. Lack of oestrogen prompts endocrine cell ageing of the ovary, whereas oestrogen overflow impacts on epithelial cell neoplastic development. 2. Recent studies indicate that oestrogen regulates cell proliferative fates by a mechanism of reprogramming the size of telomeres (ends of chromosomes) in the oestrogen target cells. This is achieved by upregulating the telomerase reverse transcriptase (TERT) gene in a temporal and spatial manner. 3. Currently, the relationship between oestrogen and telomerase activity in regulating productive cell development and function remains elusive. A number of lines of evidence suggest that telomerase is a downstream target of oestrogen in oestrogen-dependent reproductive ageing and neoplastic development. 4. The present minireview discusses our current understanding of the mechanisms by which telomerase maintenance of telomere homeostasis mediates oestrogen-induced ageing and tumourigenesis in the ovary under physiological and pathological conditions.
Collapse
Affiliation(s)
- He Li
- Department of Immunology, Molecular Signalling Laboratory, Monash University Central Clinical School, AMREP, Melbourne, Victoria 3004, Australia.
| | | | | |
Collapse
|
50
|
Abstract
Stem cells have enormous potential for therapeutic application because of their ability to self-renew and differentiate into different cell types. Gonads, which consist of somatic cells and germ cells, are the only organs capable of transmitting genetic materials to the offspring. Germ-line stem cells and somatic stem cells have been found in the testis; however, the presence of stem cells in the ovary remains controversial. In this review, we discuss studies focusing on whether stem cell properties are present in the different cell types of male and female gonads and their implications on stem cell research.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign
| | - Ivraym Barsoum
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign
| | - Rupesh Gupta
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign
| | - Marie-Claude Hofmann
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign
| | | |
Collapse
|