1
|
Jiang K, Jorgensen JS. Fetal Leydig cells: What we know and what we don't. Mol Reprod Dev 2024; 91:e23739. [PMID: 38480999 PMCID: PMC11135463 DOI: 10.1002/mrd.23739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/24/2024] [Indexed: 05/24/2024]
Abstract
During male fetal development, testosterone plays an essential role in the differentiation and maturation of the male reproductive system. Deficient fetal testosterone production can result in variations of sex differentiation that may cause infertility and even increased tumor incidence later in life. Fetal Leydig cells in the fetal testis are the major androgen source in mammals. Although fetal and adult Leydig cells are similar in their functions, they are two distinct cell types, and therefore, the knowledge of adult Leydig cells cannot be directly applied to understanding fetal Leydig cells. This review summarizes our current knowledge of fetal Leydig cells regarding their cell biology, developmental biology, and androgen production regulation in rodents and human. Fetal Leydig cells are present in basement membrane-enclosed clusters in between testis cords. They originate from the mesonephros mesenchyme and the coelomic epithelium and start to differentiate upon receiving a Desert Hedgehog signal from Sertoli cells or being released from a NOTCH signal from endothelial cells. Mature fetal Leydig cells produce androgens. Human fetal Leydig cell steroidogenesis is LHCGR (Luteinizing Hormone Chronic Gonadotropin Receptor) dependent, while rodents are not, although other Gαs -protein coupled receptors might be involved in rodent steroidogenesis regulation. Fetal steroidogenesis ceases after sex differentiation is completed, and some fetal Leydig cells dedifferentiate to serve as stem cells for adult testicular cell types. Significant gaps are acknowledged: (1) Why are adult and fetal Leydig cells different? (2) What are bona fide progenitor and fetal Leydig cell markers? (3) Which signaling pathways and transcription factors regulate fetal Leydig cell steroidogenesis? It is critical to discover answers to these questions so that we can understand vulnerable targets in fetal Leydig cells and the mechanisms for androgen production that when disrupted, leads to variations in sex differentiation that range from subtle to complete sex reversal.
Collapse
Affiliation(s)
- Keer Jiang
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joan S. Jorgensen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Bhattacharya I, Dey S. Emerging concepts on Leydig cell development in fetal and adult testis. Front Endocrinol (Lausanne) 2023; 13:1086276. [PMID: 36686449 PMCID: PMC9851038 DOI: 10.3389/fendo.2022.1086276] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Leydig cells (Lc) reside in the interstitial compartment of the testis and are the target of Luteinising hormone (LH) for Testosterone (T) production, thus critically regulates male fertility. Classical histological studies have identified two morphologically different populations of Lc during testicular development [fetal (FLc) and adult (ALc)]. Recent progress in ex vivo cell/organ culture, genome-wide analysis, genetically manipulated mouse models, lineage tracing, and single-cell RNA-seq experiments have revealed the diverse cellular origins with differential transcriptomic and distinct steroidogenic outputs of these populations. FLc originates from both coelomic epithelium and notch-active Nestin-positive perivascular cells located at the gonad-mesonephros borders, and get specified as Nr5a1 (previously known as Ad4BP/SF-1) expressing cells by embryonic age (E) 12.5 days in fetal mouse testes. These cells produce androstenedione (precursor of T, due to lack of HSD17β3 enzyme) and play critical a role in initial virilization and patterning of the male external genitalia. However, in neonatal testis, FLc undergoes massive regression/dedifferentiation and gradually gets replaced by T-producing ALc. Very recent studies suggest a small fraction (5-20%) of FLc still persists in adult testis. Both Nestin-positive perivascular cells and FLc are considered to be the progenitor populations for ALc. This minireview article summarizes the current understanding of Lc development in fetal and adult testes highlighting their common or diverse cellular (progenitor/stem) origins with respective functional significance in both rodents and primates. (227 words).
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Periye, Kerala, India
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
3
|
Role of mesonephric contribution to mouse testicular development revisited. Differentiation 2023; 129:109-119. [PMID: 35000816 DOI: 10.1016/j.diff.2021.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 01/25/2023]
Abstract
The role of the mesonephros in testicular development was re-evaluated by growing embryonic day 11.5 (E11.5) mouse testes devoid of mesonephros for 8-21 days in vivo under the renal capsule of castrated male athymic nude mice. This method provides improved growth conditions relative to previous studies based upon short-term (4-7 days) organ culture. Meticulous controls involved wholemount examination of dissected E11.5 mouse testes as well as serial sections of dissected E11.5 mouse testes which were indeed shown to be devoid of mesonephros. As expected, grafts of E11.5 mouse testes with mesonephros attached formed seminiferous tubules and also contained mesonephric derivatives. Grafts of E11.5 mouse testes without associated mesonephros also formed seminiferous tubules and never contained mesonephric derivatives. The consistent absence of mesonephric derivatives in grafts of E11.5 mouse testes grafted alone is further proof of the complete removal of the mesonephros from the E11.5 mouse testes. The testicular tissues that developed in grafts of E11.5 mouse testes alone contained canalized seminiferous tubules composed of Sox9-positive Sertoli cells as well as GENA-positive germ cells. The seminiferous tubules were surrounded by α-actin-positive myoid cells, and the interstitial space contained 3βHSD-1-positive Leydig cells. Grafts of E11.5 GFP mouse testes into wild-type hosts developed GFP-positive vasculature indicating that E11.5 mouse testes contain vascular precursors. These results indicate that the E11.5 mouse testis contains precursor cells for Sertoli cells, Leydig cells, myoid cells and vasculature whose development and differentiation are independent of cells migrating from the E11.5 mesonephros.
Collapse
|
4
|
Wang Y, Liu X, Xie X, He J, Gao Y. Adult‑onset X‑linked adrenal hypoplasia congenita caused by a novel mutation in DAX1/NR0B1: A case report and literature review. Exp Ther Med 2022; 24:628. [PMID: 36160878 PMCID: PMC9468782 DOI: 10.3892/etm.2022.11565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022] Open
Abstract
Adrenal hypoplasia congenita (AHC) is a rare X-linked recessive disease caused by mutations in the nuclear receptor subfamily 0, group B, member 1 (NR0B1) gene, which is also referred to as dosage-sensitive sex-reversal, adrenal hypoplasia congenita, in the critical region of the X chromosome, gene 1 (DAX1). This gene is expressed in the hypothalamus, anterior pituitary and steroidogenic tissues, including the gonads and adrenal cortex. Adult-onset forms of X-linked AHC are a significant cause of concern. In the present study, the case of a 21-year-old male who exhibited adrenal insufficiency and hypogonadotropic hypogonadism was described. The patient initially presented with nausea, vomiting, fatigue and dizziness. The laboratory results demonstrated that the patient had hyponatremia, a low basal cortisol concentration and increased adrenocorticotropic hormone levels. Molecular genetic examination revealed a novel frameshift mutation (c.1005delC, p.V336Cfs*36). Following steroid supplementation, the patient's vomiting, fatigue and dizziness rapidly improved. To the best of our knowledge, the present study was the first case report of adult-onset X-linked AHC with this novel frameshift mutation. Furthermore, the present study highlighted differences in the clinical presentation of adult-onset forms of X-linked AHC. This may therefore alert medical professionals to the need to perform genetic analysis for DAX1 mutations in adolescents and adults with primary adrenal insufficiency and hypogonadotropic hypogonadism.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiufen Liu
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaona Xie
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jingjing He
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ying Gao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
5
|
A patched1 gene homologue participates in female differentiation of Cynoglossus semilaevis. Gene Expr Patterns 2022; 45:119265. [DOI: 10.1016/j.gep.2022.119265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/21/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022]
|
6
|
Mo JY, Yan YS, Lin ZL, Liu R, Liu XQ, Wu HY, Yu JE, Huang YT, Sheng JZ, Huang HF. Gestational diabetes mellitus suppresses fetal testis development in mice. Biol Reprod 2022; 107:148-156. [PMID: 35774031 DOI: 10.1093/biolre/ioac138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/10/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
The prevalence of Gestational diabetes mellitus (GDM) is increasing rapidly. In addition to the metabolic disease risks, GDM might increase the risks of cryptorchidism in children. However, its mechanism involved in abnormalities of the male reproductive system is still unclear. The purpose of this study was to study the effects of GDM on the development of mouse fetal Leydig and Sertoli cells. Pregnant mice were treated on gestational day (GD) 6.5 and 12.5 with streptozotocin (STZ, 100 mg/kg) or vehicle (sodium citrate buffer). Leydig and Sertoli cell development and functions were evaluated by investigating serum testosterone levels, cell number and distribution, genes, and protein expression. GDM decreased serum testosterone levels, the anogenital distance, and the level of DHH in Sertoli cells of testes of male offspring. Fetal Leydig cell number was also decreased in testes of GDM offspring by delaying the commitment of stem Leydig cells into the Leydig cell lineage. RNA-seq showed that FOXL2, RSPO1/β-Catenin signaling was activated and Gsk3β signaling was inhibited in GDM offspring testis. In conclusion, GDM disrupted reproductive tract and testis development in mouse male offspring via altering genes related to development.
Collapse
Affiliation(s)
- Jia-Ying Mo
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Yi-Shang Yan
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Zhong-Liang Lin
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Rui Liu
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Xuan-Qi Liu
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Hai-Yan Wu
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Jia-En Yu
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Yu-Tong Huang
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - Jian-Zhong Sheng
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| | - He-Feng Huang
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang university school of medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Mayère C, Regard V, Perea-Gomez A, Bunce C, Neirijnck Y, Djari C, Bellido-Carreras N, Sararols P, Reeves R, Greenaway S, Simon M, Siggers P, Condrea D, Kühne F, Gantar I, Tang F, Stévant I, Batti L, Ghyselinck NB, Wilhelm D, Greenfield A, Capel B, Chaboissier MC, Nef S. Origin, specification and differentiation of a rare supporting-like lineage in the developing mouse gonad. SCIENCE ADVANCES 2022; 8:eabm0972. [PMID: 35613264 PMCID: PMC10942771 DOI: 10.1126/sciadv.abm0972] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
Gonadal sex determination represents a unique model for studying cell fate decisions. However, a complete understanding of the different cell lineages forming the developing testis and ovary remains elusive. Here, we investigated the origin, specification, and subsequent sex-specific differentiation of a previously uncharacterized population of supporting-like cells (SLCs) in the developing mouse gonads. The SLC lineage is closely related to the coelomic epithelium and specified as early as E10.5, making it the first somatic lineage to be specified in the bipotential gonad. SLC progenitors are localized within the genital ridge at the interface with the mesonephros and initially coexpress Wnt4 and Sox9. SLCs become sexually dimorphic around E12.5, progressively acquire a more Sertoli- or pregranulosa-like identity and contribute to the formation of the rete testis and rete ovarii. Last, we found that WNT4 is a crucial regulator of the SLC lineage and is required for normal development of the rete testis.
Collapse
Affiliation(s)
- Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Switzerland
| | - Violaine Regard
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Switzerland
| | - Aitana Perea-Gomez
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Corey Bunce
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Cyril Djari
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | | | - Pauline Sararols
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Richard Reeves
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Simon Greenaway
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Michelle Simon
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Diana Condrea
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP1014267404 ILLKIRCH CEDEX, France
| | - Françoise Kühne
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Ivana Gantar
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Furong Tang
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Laura Batti
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Norbert B. Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP1014267404 ILLKIRCH CEDEX, France
| | - Dagmar Wilhelm
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Switzerland
| |
Collapse
|
8
|
Shi H, Ru X, Mustapha UF, Jiang D, Huang Y, Pan S, Zhu C, Li G. Characterization, expression, and regulatory effects of nr0b1a and nr0b1b in spotted scat (Scatophagus argus). Comp Biochem Physiol B Biochem Mol Biol 2021; 256:110644. [PMID: 34224854 DOI: 10.1016/j.cbpb.2021.110644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022]
Abstract
Nuclear receptor subfamily 0 group B member 1 (Nr0b1) belongs to the nuclear receptor (NR) superfamily. It plays critical roles in sex determination, sex differentiation, and gonadal development in mammals. In this study, the duplicated genes nr0b1a and nr0b1b were identified in spotted scat (Scatophagus argus). Phylogenetic and synteny analyses revealed that, unlike nr0b1a, nr0b1b was retained in several species of teleosts after an nr0b1 gene duplication event but was secondarily lost in other fish species, amphibians, reptiles, birds, and mammals. In a sequence analysis, only 1.5 LXXLL-related repeat motifs were identified in spotted scat Nr0b1a, Nr0b1b, and non-mammalian Nr0b1a/Nr0b1, different from the 3.5 repeat motifs in mammalian Nr0b1. By qPCR, nr0b1a and nr0b1b were highly expressed in testes from stages IV to V and in ovaries from stages II to IV, respectively. Male-to-female sex reversal was induced in XY spotted scat by the administration of exogenous E2. A qPCR analysis showed that nr0b1b mRNA expression was higher in sex-reversed XY fish than in control XY fish, with no difference in nr0b1a. A luciferase assay showed that spotted scat Nr0b1a and Nr0b1b did not individually activate cyp19a1a gene transcription. As in mammals, spotted scat Nr0b1a suppressed Nr5a1-mediated cyp19a1a expression, despite containing only 1.5 LXXLL-related repeat motifs in its N-terminal region, while Nr0b1b stimulated Nr5a1-mediated cyp19a1a transcription. These results demonstrated that nr0b1a and nr0b1b in spotted scat have distinct expression patterns and regulatory effects and further indicate that nr0b1b might be involved in ovarian development by regulating Nr5a1-mediated cyp19a1a expression.
Collapse
Affiliation(s)
- Hongjuan Shi
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaoying Ru
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Umar Farouk Mustapha
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Dongneng Jiang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yang Huang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Shuhui Pan
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chunhua Zhu
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guangli Li
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Fisheries College, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
9
|
The conditional deletion of steroidogenic factor 1 (Nr5a1) in Sox9-Cre mice compromises testis differentiation. Sci Rep 2021; 11:4486. [PMID: 33627800 PMCID: PMC7904858 DOI: 10.1038/s41598-021-84095-y] [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: 07/04/2020] [Accepted: 02/12/2021] [Indexed: 12/25/2022] Open
Abstract
Steroidogenic factor 1 (NR5A1) is essential for gonadal development. To study the importance of NR5A1 during early gonadal sex differentiation, we generated Sox9-Cre-Nr5a1 conditional knockout (cKO) mice: Sox9-Cre;Nr5a1flox/flox and Sox9-Cre;Nr5a1flox/− mice. Double-immunostaining for NR5A1 and AMH revealed silenced NR5A1 in Sertoli cells and reduced AMH+ cells in the gonads of XY Sox9-Cre-Nr5a1 cKO mice between embryonic days 12.5 (E12.5) and E14.5. Double-immunostaining for SOX9 and FOXL2 further indicated an early block in Sertoli cells and ectopic granulosa cell differentiation. The number of cells expressing the Leydig cell marker 3βHSD obviously reduced in the gonads of XY Sox9-Cre;Nr5a1flox/− but not Sox9-Cre;Nr5a1flox/flox mice at E15.5. The presence of STRA8+ cells indicated that germ cells entered meiosis in the gonads of XY Sox9-Cre-Nr5a1 cKO mice. The results of qRT-PCR revealed remarkably reduced and elevated levels of testis and ovary markers, respectively, in the gonads of XY Sox9-Cre-Nr5a1 cKO mice at E12.5‒E13.5. These data suggested that the loss of Nr5a1 abrogates the testicular pathway and induces the ectopic ovarian pathway, resulting in postnatal partial/complete male-to-female gonadal sex reversal. Our findings provide evidence for the critical role of NR5A1 in murine gonadal sex determination in vivo.
Collapse
|
10
|
Picard JY, Josso N. Persistent Müllerian duct syndrome: an update. Reprod Fertil Dev 2020; 31:1240-1245. [PMID: 32172781 DOI: 10.1071/rd17501] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/06/2018] [Indexed: 11/23/2022] Open
Abstract
Male sex differentiation is driven by two hormones, testosterone and anti-Müllerian hormone (AMH), responsible for regression of Müllerian ducts in male fetuses. Mutations inactivating AMH or AMH receptor type 2 (AMHR2) are responsible for persistent Müllerian duct syndrome (PMDS) in otherwise normally virilised 46,XY males. This review is based on published cases, including 157 personal ones. PMDS can present in one of three ways: bilateral cryptorchidism, unilateral cryptorchidism with contralateral hernia and transverse testicular ectopia. Abnormalities of male excretory ducts are frequent. Testicular malignant degeneration occurs in 33% of adults with PMDS. Cancer of Müllerian derivatives is less frequent. Fertility is rare but possible if at least one testis is scrotal and its excretory ducts are intact. Up to January 2019, 81 families with 65 different mutations of the AMH gene, mostly in exons 1, 2 and 5, have been identified. AMHR2 gene mutations comprising 64 different alleles have been discovered in 79 families. The most common mutation, a 27-bp deletion in the kinase domain, was found in 30 patients of mostly Northern European origin. In 12% of cases, no mutation of AMH or AMHR2 has been detected, suggesting a disruption of other pathways involved in Müllerian regression.
Collapse
Affiliation(s)
- Jean-Yves Picard
- INSERM UMRS 938, Centre de Recherche Saint Antoine, 27, rue Chaligny, 75571 Paris Cedex 12, France; and Faculté de Médecine Sorbonne Université, 27, rue Chaligny, 75571 Paris Cedex 12, France; and Corresponding author.
| | - Nathalie Josso
- INSERM UMRS 938, Centre de Recherche Saint Antoine, 27, rue Chaligny, 75571 Paris Cedex 12, France; and Faculté de Médecine Sorbonne Université, 27, rue Chaligny, 75571 Paris Cedex 12, France
| |
Collapse
|
11
|
Stévant I, Nef S. Single cell transcriptome sequencing: A new approach for the study of mammalian sex determination. Mol Cell Endocrinol 2018; 468:11-18. [PMID: 29371022 DOI: 10.1016/j.mce.2018.01.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/21/2018] [Accepted: 01/21/2018] [Indexed: 10/18/2022]
Abstract
Mammalian sex determination is a highly complex developmental process that is particularly difficult to study due to the limited number of gonadal cells present at the bipotential stage, the large cellular heterogeneity in both testis and ovaries and the rapid sex-dependent differentiation processes. Single-cell RNA-sequencing (scRNA-seq) circumvents the averaging artifacts associated with methods traditionally used to profile bulk populations of cells. It is a powerful tool that allows the identification and classification of cell populations in a comprehensive and unbiased manner. In particular, scRNA-seq enables the tracing of cells along developmental trajectories and characterization of the transcriptional dynamics controlling their differentiation. In this review, we describe the current state-of-the-art experimental methods used for scRNA-seq and discuss their strengths and limitations. Additionally, we summarize the multiple key insights that scRNA-seq has provided to the understanding of mammalian sex determination. Finally, we briefly discuss the future of this technology, as well as complementary applications in single cell -omics in the context of mammalian sex determination.
Collapse
Affiliation(s)
- Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland; SIB, Swiss Institute of Bioinformatics, University of Geneva, 1211 Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
12
|
Shima Y, Morohashi KI. Leydig progenitor cells in fetal testis. Mol Cell Endocrinol 2017; 445:55-64. [PMID: 27940302 DOI: 10.1016/j.mce.2016.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/18/2016] [Accepted: 12/07/2016] [Indexed: 12/21/2022]
Abstract
Testicular Leydig cells play pivotal roles in masculinization of organisms by producing androgens. At least two distinct Leydig cell populations sequentially emerge in the mammalian testis. Leydig cells in the fetal testis (fetal Leydig cells) appear just after initial sex differentiation and induce masculinization of male fetuses. Although there has been a debate on the fate of fetal Leydig cells in the postnatal testis, it has been generally believed that fetal Leydig cells regress and are completely replaced by another Leydig cell population, adult Leydig cells. Recent studies revealed that gene expression patterns are different between fetal and adult Leydig cells and that the androgens produced in fetal Leydig cells are different from those in adult Leydig cells in mice. Although these results suggested that fetal and adult Leydig cells have distinct origins, several recent studies of mouse models support the hypothesis that fetal and adult Leydig cells arise from a common progenitor pool. In this review, we first provide an overview of previous knowledge, mainly from mouse studies, focusing on the cellular origins of fetal Leydig cells and the regulatory mechanisms underlying fetal Leydig cell differentiation. In addition, we will briefly discuss the functional differences of fetal Leydig cells between human and rodents. We will also discuss recent studies with mouse models that give clues for understanding how the progenitor cells in the fetal testis are subsequently destined to become fetal or adult Leydig cells.
Collapse
Affiliation(s)
- Yuichi Shima
- Department of Anatomy, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan.
| | - Ken-Ichirou Morohashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
13
|
Song S, Yu H, Li Q. Genome survey and characterization of reproduction-related genes in the Pacific oyster. INVERTEBR REPROD DEV 2017. [DOI: 10.1080/07924259.2017.1287780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Shanshan Song
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Hong Yu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Qi Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| |
Collapse
|
14
|
Abstract
Since its discovery nearly 30 years ago, the Hedgehog (Hh) signaling pathway has been shown to be pivotal in many developmental and pathophysiological processes in several steroidogenic tissues, including the testis, ovary, adrenal cortex, and placenta. New evidence links the evolutionarily conserved Hh pathway to the steroidogenic organs, demonstrating how Hh signaling can influence their development and homeostasis and can act in concert with steroids to mediate physiological functions. In this review, we highlight the role of the components of the Hh signaling pathway in steroidogenesis of endocrine tissues.
Collapse
Affiliation(s)
- Isabella Finco
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan 48109; , ,
| | | | | | | |
Collapse
|
15
|
Karpova T, Ravichandiran K, Insisienmay L, Rice D, Agbor V, Heckert LL. Steroidogenic factor 1 differentially regulates fetal and adult leydig cell development in male mice. Biol Reprod 2015; 93:83. [PMID: 26269506 DOI: 10.1095/biolreprod.115.131193] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/05/2015] [Indexed: 12/17/2022] Open
Abstract
The nuclear receptor steroidogenic factor 1 (SF-1, AD4BP, NR5A1) is a key regulator of the endocrine axes and is essential for adrenal and gonad development. Partial rescue of Nr5a1(-/-) mice with an SF-1-expressing transgene caused a hypomorphic phenotype that revealed its roles in Leydig cell development. In contrast to controls, all male rescue mice (Nr5a1(-/-);tg(+/0)) showed varying signs of androgen deficiency, including spermatogenic arrest, cryptorchidism, and poor virilization. Expression of various Leydig cell markers measured by immunohistochemistry, Western blot analysis, and RT-PCR indicated fetal and adult Leydig cell development were differentially impaired. Whereas fetal Leydig cell development was delayed in Nr5a1(-/-);tg(+/0) embryos, it recovered to control levels by birth. In contrast, Sult1e1, Vcam1, and Hsd3b6 transcript levels in adult rescue testes indicated complete blockage in adult Leydig cell development. In addition, between Postnatal Days 8 and 12, peritubular cells expressing PTCH1, SF-1, and CYP11A1 were observed in control testes but not in rescue testes, indicating SF-1 is needed for either survival or differentiation of adult Leydig cell progenitors. Cultured prepubertal rat peritubular cells also expressed SF-1 and PTCH1, but Cyp11a1 was expressed only after treatment with cAMP and retinoic acid. Together, data show SF-1 is needed for proper development of fetal and adult Leydig cells but with distinct primary functions; in fetal Leydig cells, it regulates differentiation, whereas in adult Leydig cells it regulates progenitor cell formation and/or survival.
Collapse
Affiliation(s)
- Tatiana Karpova
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Kumarasamy Ravichandiran
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lovella Insisienmay
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Daren Rice
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Valentine Agbor
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Leslie L Heckert
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
16
|
Hussain S, Amar A, Najeeb MN, Khaliq S. Two novel mutations in theNR5A1gene as a cause of disorders of sex development in a Pakistani cohort of 46,XY patients. Andrologia 2015; 48:509-17. [DOI: 10.1111/and.12470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2015] [Indexed: 11/29/2022] Open
Affiliation(s)
- S. Hussain
- Department of Biochemistry; University of Health Sciences; Lahore Pakistan
| | - A. Amar
- Department Human Genetics & Molecular Biology; University of Health Sciences; Lahore Pakistan
| | - M. N. Najeeb
- Department of Biochemistry; Quaid-e-Azam Medical Collage; Bahawalpur Pakistan
| | - S. Khaliq
- Department Human Genetics & Molecular Biology; University of Health Sciences; Lahore Pakistan
| |
Collapse
|
17
|
Nistal M, Paniagua R, González-Peramato P, Reyes-Múgica M. Perspectives in pediatric pathology, chapter 1. Normal development of testicular structures: from the bipotential gonad to the fetal testis. Pediatr Dev Pathol 2015; 18:88-102. [PMID: 25119266 DOI: 10.2350/12-04-1184-pb.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Manuel Nistal
- 1 Pathology, Hospital La Paz, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo #2, Madrid 28029, Spain
| | | | | | | |
Collapse
|
18
|
Teerds KJ, Huhtaniemi IT. Morphological and functional maturation of Leydig cells: from rodent models to primates. Hum Reprod Update 2015; 21:310-28. [PMID: 25724971 DOI: 10.1093/humupd/dmv008] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 01/15/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Leydig cells (LC) are the sites of testicular androgen production. Development of LC occurs in the testes of most mammalian species as two distinct growth phases, i.e. as fetal and pubertal/adult populations. In primates there are indications of a third neonatal growth phase. LC androgen production begins in embryonic life and is crucial for the intrauterine masculinization of the male fetal genital tract and brain, and continues until birth after which it rapidly declines. A short post-natal phase of LC activity in primates (including human) termed 'mini-puberty' precedes the period of juvenile quiescence. The adult population of LC evolves, depending on species, in mid- to late-prepuberty upon reawakening of the hypothalamic-pituitary-testicular axis, and these cells are responsible for testicular androgen production in adult life, which continues with a slight gradual decline until senescence. This review is an updated comparative analysis of the functional and morphological maturation of LC in model species with special reference to rodents and primates. METHODS Pubmed, Scopus, Web of Science and Google Scholar databases were searched between December 2012 and October 2014. Studies published in languages other than English or German were excluded, as were data in abstract form only. Studies available on primates were primarily examined and compared with available data from specific animal models with emphasis on rodents. RESULTS Expression of different marker genes in rodents provides evidence that at least two distinct progenitor lineages give rise to the fetal LC (FLC) population, one arising from the coelomic epithelium and the other from specialized vascular-associated cells along the gonad-mesonephros border. There is general agreement that the formation and functioning of the FLC population in rodents is gonadotrophin-responsive but not gonadotrophin-dependent. In contrast, although there is in primates some controversy on the role of gonadotrophins in the formation of the FLC population, there is consensus about the essential role of gonadotrophins in testosterone production. Like the FLC population, adult Leydig cells (ALC) in rodents arise from stem cells, which have their origin in the fetal testis. In contrast, in primates the ALC population is thought to originate from FLC, which undergo several cycles of regression and redifferentiation before giving rise to the mature ALC population, as well as from differentiation of stem cells/precursor cells. Despite this difference in origin, both in primates and rodents the formation of the mature and functionally active ALC population is critically dependent on the pituitary gonadotrophin, LH. From studies on rodents considerable knowledge has emerged on factors that are involved besides LH in the regulation of this developmental process. Whether the same factors also play a role in the development of the mature primate LC population awaits further investigation. CONCLUSION Distinct populations of LC develop along the life span of males, including fetal, neonatal (primates) and ALC. Despite differences in the LC lineages of rodents and primates, the end product is a mature population of LC with the main function to provide androgens necessary for the maintenance of spermatogenesis and extra-gonadal androgen actions.
Collapse
Affiliation(s)
- Katja J Teerds
- Human and Animal Physiology, Wageningen University, De Elst 1, 6709 WD, Wageningen, The Netherlands
| | - Ilpo T Huhtaniemi
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, Du Cane Road, W12 0NN London, UK Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| |
Collapse
|
19
|
Orekhova AS, Rubtsov PM. DAX1, an unusual member of the nuclear receptor superfamily with diverse functions. Mol Biol 2015. [DOI: 10.1134/s0026893315010124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Tang C, Pan Y, Luo H, Xiong W, Zhu H, Ruan H, Wang J, Zou C, Tang L, Iguchi T, Long F, Wu X. Hedgehog signaling stimulates the conversion of cholesterol to steroids. Cell Signal 2015; 27:487-97. [PMID: 25582983 DOI: 10.1016/j.cellsig.2015.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 12/19/2014] [Accepted: 01/05/2015] [Indexed: 12/28/2022]
Abstract
Cholesterol modification of Hedgehog (Hh) ligands is fundamental for the activity of Hh signaling, and cholesterol biosynthesis is also required for intracellular Hh signaling transduction. Here, we investigated the roles and underlying mechanism of Hh signaling in metabolism of cholesterol. The main components of the Hh pathway are abundantly expressed in both human cytotrophoblasts and trophoblast-like cells. Activation of Hh signaling induces the conversion of cholesterol to progesterone (P4) and estradiol (E2) through up-regulating the expression of steroidogenic enzymes including P450 cholesterol side chain cleavage enzyme (P450scc), 3β-hydroxysteroid dehydrogenase type 1 (3β-HSD1), and aromatase. Moreover, inhibition of Hh signaling attenuates not only Hh-induced expression of steroidogenic enzymes but also the conversion of cholesterol to P4 and E2. Whereas Gli3 is required for Hh-induced P450scc expression, Gli2 mediates the induction of 3β-HSD1 and aromatase. Finally, in ovariectomized nude mice, systemic inhibition of Hh signaling by cyclopamine suppresses circulating P4 and E2 levels derived from a trophoblast-like choricarcinoma xenograft, and attenuates uterine response to P4 and E2. Together these results uncover a hitherto uncharacterized role of Hh signaling in metabolism of cholesterol.
Collapse
Affiliation(s)
- Chao Tang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yibin Pan
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Huan Luo
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Wenyi Xiong
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Haibin Zhu
- The Affiliated First Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hongfeng Ruan
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jirong Wang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Chaochun Zou
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Lanfang Tang
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Takuma Iguchi
- Department of Toxicology, Osaka University, Suita, Osaka 565-0871, Japan
| | - Fanxin Long
- Departments of Orthopaedic Surgery, Medicine and Developmental Biology Washington University in St. Louis, MO, 63110, USA
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China; Departments of Orthopaedic Surgery, Medicine and Developmental Biology Washington University in St. Louis, MO, 63110, USA.
| |
Collapse
|
21
|
Abstract
Sex determination refers to the developmental decision that directs the bipotential genital ridge to develop as a testis or an ovary. Genetic studies on mice and humans have led to crucial advances in understanding the molecular fundamentals of sex determination and the mutually antagonistic signaling pathway. In this review, we summarize the current molecular mechanisms of sex determination by focusing on the known critical sex determining genes and their related signaling pathways in mammalian vertebrates from mice to humans. We also discuss the underlying delicate balance between testis and ovary sex determination pathways, concentrating on the antagonisms between major sex determining genes.
Collapse
Affiliation(s)
- Zhen-Yu She
- The Sperm LaboratoryCollege of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm LaboratoryCollege of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| |
Collapse
|
22
|
Role of Orphan Nuclear Receptor DAX-1/NR0B1 in Development, Physiology, and Disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/582749] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DAX-1/NR0B1 is an unusual orphan receptor that has a pivotal role in the development and function of steroidogenic tissues and of the reproductive axis. Recent studies have also indicated that this transcription factor has an important function in stem cell biology and in several types of cancer. Here I critically review the most important findings on the role of DAX-1 in development, physiology, and disease of endocrine tissues since the cloning of its gene twenty years ago.
Collapse
|
23
|
Ravel C, Hyon C, Siffroi JP, Christin-Maitre S. Are human male patients with DAX1/NR0B1 mutations infertile? ANNALES D'ENDOCRINOLOGIE 2014; 75:126-7. [DOI: 10.1016/j.ando.2014.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/20/2014] [Indexed: 11/30/2022]
|
24
|
Three-step method for proliferation and differentiation of human embryonic stem cell (hESC)-derived male germ cells. PLoS One 2014; 9:e90454. [PMID: 24690677 PMCID: PMC3972183 DOI: 10.1371/journal.pone.0090454] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/03/2014] [Indexed: 12/26/2022] Open
Abstract
The low efficiency of differentiation into male germ cell (GC)-like cells and haploid germ cells from human embryonic stem cells (hESCs) reflects the culture method employed in the two-dimensional (2D)-microenvironment. In this study, we applied a three-step media and calcium alginate-based 3D-culture system for enhancing the differentiation of hESCs into male germ stem cell (GSC)-like cells and haploid germ cells. In the first step, embryoid bodies (EBs) were derived from hESCs cultured in EB medium for 3 days and re-cultured for 4 additional days in EB medium with BMP4 and RA to specify GSC-like cells. In the second step, the resultant cells were cultured in GC-proliferation medium for 7 days. The GSC-like cells were then propagated after selection using GFR-α1 and were further cultured in GC-proliferation medium for 3 weeks. In the final step, a 3D-co-culture system using calcium alginate encapsulation and testicular somatic cells was applied to induce differentiation into haploid germ cells, and a culture containing approximately 3% male haploid germ cells was obtained after 2 weeks of culture. These results demonstrated that this culture system could be used to efficiently induce GSC-like cells in an EB population and to promote the differentiation of ESCs into haploid male germ cells.
Collapse
|
25
|
Flood DEK, Fernandino JI, Langlois VS. Thyroid hormones in male reproductive development: evidence for direct crosstalk between the androgen and thyroid hormone axes. Gen Comp Endocrinol 2013; 192:2-14. [PMID: 23524004 DOI: 10.1016/j.ygcen.2013.02.038] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/24/2013] [Accepted: 02/26/2013] [Indexed: 01/06/2023]
Abstract
Thyroid hormones (THs) exert a broad range of effects on development in vertebrate species, demonstrating connections in nearly every biological endocrine system. In particular, studies have shown that THs play a role in sexual differentiation and gonadal development in mammalian and non-mammalian species. There is considerable evidence that the effects of THs on reproductive development are mediated through the female hormonal axis; however, recent findings suggest a more direct crosstalk between THs and the androgen axis. These findings demonstrate that THs have considerable influence in the sexual ontogeny of male vertebrates, through direct interactions with select sex-determining-genes and regulation of gonadotropin production in the hypothalamus-pituitary-gonad axis. THs also regulate androgen biosynthesis and signaling through direct and indirect regulation of steroidogenic enzyme expression and activity. Novel promoter analysis presented in this work demonstrates the potential for direct and vertebrate wide crosstalk at the transcriptional level in mice (Mus musculus), Western clawed frogs (Silurana tropicalis) and medaka (Oryzias latipes). Cumulative evidence from previous studies; coupled with novel promoter analysis suggests mechanisms for a more direct crosstalk between the TH and male reproductive axes across vertebrate species.
Collapse
Affiliation(s)
- Diana E K Flood
- Department of Chemistry and Chemical Engineering, Royal Military College of Canada, ON, Canada; Biology Department, Queen's University, Kingston, ON, Canada.
| | | | | |
Collapse
|
26
|
DAX1 regulatory networks unveil conserved and potentially new functions. Gene 2013; 530:66-74. [PMID: 23954228 DOI: 10.1016/j.gene.2013.07.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/09/2013] [Indexed: 11/22/2022]
Abstract
DAX1 is an orphan nuclear receptor with actions in mammalian sex determination, regulation of steroidogenesis, embryonic development and neural differentiation. Conserved patterns of DAX1 gene expression from mammals to fish have been taken to suggest conserved function. In the present study, the European sea bass, Dicentrarchus labrax, DAX1 promoter was isolated and its conserved features compared to other fish and mammalian DAX1 promoters in order to derive common regulators and functional gene networks. Fish and mammalian DAX1 promoters share common sets of transcription factor frameworks which were also present in the promoter region of another 127 genes. Pathway analysis clustered these into candidate gene networks associated with the fish and mammalian DAX1. The networks identified are concordant with described functions for DAX1 in embryogenesis, regulation of transcription, endocrine development and steroid production. Novel candidate gene network partners were also identified, which implicate DAX1 in ion homeostasis and transport, lipid transport and skeletal development. Experimental evidence is provided supporting roles for DAX1 in steroid signalling and osmoregulation in fish. These results highlight the usefulness of the in silico comparative approach to analyse gene regulation for hypothesis generation. Conserved promoter architecture can be used also to predict potentially new gene functions. The approach reported can be applied to genes from model and non-model species.
Collapse
|
27
|
Gonzales KAU, Ng HH. Driving pluripotency and reprogramming: nuclear receptors at the helm. Semin Cell Dev Biol 2013; 24:670-8. [PMID: 23916717 DOI: 10.1016/j.semcdb.2013.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 07/16/2013] [Accepted: 07/21/2013] [Indexed: 01/30/2023]
Abstract
The identity of a cell is determined by the concerted interplay of multiple molecular modulators such as transcription factors, chromatin modifiers and signalling mediators. Among these, the transcriptional circuitry holds great influence on the specification and maintenance of a cellular state, and its perturbation can trigger a transition to another cell state. This is particularly striking in the field of pluripotency, where tempering the expression levels of one or few transcription factors is sufficient to induce the loss or acquisition of the pluripotent state. Recently, nuclear receptors, a class of transcription factors, have emerged as major players in the molecular network governing pluripotency. In this review, we discuss the importance of nuclear receptors in embryonic stem cell self-renewal, differentiation and cellular reprogramming, highlighting recent discoveries as well as providing an outlook in stem cell and nuclear receptor research.
Collapse
Affiliation(s)
- Kevin Andrew Uy Gonzales
- Gene Regulation Laboratory, Genome Institute of Singapore, Singapore 138672, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456, Singapore.
| | | |
Collapse
|
28
|
Morohashi K, Baba T, Tanaka M. Steroid Hormones and the Development of Reproductive Organs. Sex Dev 2013; 7:61-79. [DOI: 10.1159/000342272] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
29
|
Ehrlund A, Treuter E. Ligand-independent actions of the orphan receptors/corepressors DAX-1 and SHP in metabolism, reproduction and disease. J Steroid Biochem Mol Biol 2012; 130:169-79. [PMID: 21550402 DOI: 10.1016/j.jsbmb.2011.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 03/11/2011] [Accepted: 04/21/2011] [Indexed: 12/11/2022]
Abstract
DAX-1 and SHP are two closely related atypical orphan members of the nuclear receptor (NR) family that make up the NR0B subfamily. They combine properties of typical NRs and of NR-associated coregulators: both carry the characteristic NR ligand-binding domain but instead of a NR DNA-binding domain they have unique N-terminal regions that contain LxxLL-related NR-binding motifs often found in coregulators. Recent structural data indicate that DAX-1 lacks a ligand-binding pocket and thus should rely on ligand-independent mechanisms of regulation. This might be true, but remains to be proven, for SHP as well. DAX-1 and SHP have in common that they act as transcriptional corepressors of cholesterol metabolism pathways that are related on a molecular level. However, the expression patterns of the two NRs are largely different, with some notable exceptions, and so are the physiological processes they regulate. DAX-1 is mainly involved in steroidogenesis and reproductive development, while SHP plays major roles in maintaining cholesterol and glucose homeostasis. This review highlights the key similarities and differences between DAX-1 and SHP with regard to structure, function and biology and considers what can be learnt from recent research advances in the field. This article is part of a Special Issue entitled 'Orphan Receptors'.
Collapse
Affiliation(s)
- Anna Ehrlund
- Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institutet, S-14183 Huddinge/Stockholm, Sweden
| | | |
Collapse
|
30
|
Shima Y, Miyabayashi K, Baba T, Otake H, Katsura Y, Oka S, Zubair M, Morohashi KI. Identification of an enhancer in the Ad4BP/SF-1 gene specific for fetal Leydig cells. Endocrinology 2012; 153:417-25. [PMID: 22128023 DOI: 10.1210/en.2011-1407] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adrenal 4 binding protein/steroidogenic factor 1 (Ad4BP/SF-1) (Nr5a1) is a nuclear receptor essential for reproductive tissue development and endocrine regulation. This factor is expressed in steroidogenic tissues (e.g. adrenal glands and gonads), and expression of this factor is tightly regulated in a tissue and cell type-specific manner. Our previous studies have identified tissue and cell type-specific enhancers in the introns of the Ad4BP/SF-1 gene in fetal adrenal glands, ventromedial hypothalamus, and pituitary gonadotrope. Characterization of the enhancers had provided new insights into tissue and cell development. However, these studies have failed to identify any gonad-specific enhancer. Here, we identified a fetal Leydig cell-specific enhancer in the upstream region of the mouse Ad4BP/SF-1 gene using transgenic mouse assays. Alignment of the upstream regions among vertebrate animal species demonstrated that the enhancer consisted of three conserved regions, whereby the most highly conserved region contained an Ad4BP/SF-1 binding sequence and an E-box. Mutation of each sequence abolished the enhancer activity and led to a loss of reporter gene expression. These results suggested that Ad4BP/SF-1 gene expression in the fetal Leydig cell is regulated by a yet unidentified E-box binding protein(s) and by an autoregulatory loop formed by Ad4BP/SF-1. Although fetal Leydig cells have been thought to play crucial roles for masculinization of various fetal tissues through androgen production, other functions have remained elusive. Our identification of a fetal Leydig cell-specific enhancer in the Ad4BP/SF-1 gene would be a powerful tool to address these gaps in the knowledge base.
Collapse
Affiliation(s)
- Yuichi Shima
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Wu CM, Zhang HB, Zhou Q, Wan L, Jin J, Ni L, Pan YJ, Wu XY, Ruan LY. Two novel DAX1 gene mutations in Chinese patients with X-linked adrenal hypoplasia congenita: clinical, hormonal and genetic analysis. J Endocrinol Invest 2011; 34:e235-9. [PMID: 21270512 DOI: 10.3275/7484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mutations in the DAX1 gene result in X-linked congenital adrenal hypoplasia (AHC). Affected boys usually present with primary adrenal failure in early infancy or childhood and hypogonadotropic hypogonadism (HH) at puberty. This paper describes the clinical, hormonal, radiological, and genetic characteristics of 2 Chinese patients with X-linked AHC. Primary adrenal insufficiency occurred in the 2 patients during their childhood and HH was recognized at puberty. Genomic DNA was extracted from their peripheral blood leukocytes and coding sequence abnormalities of the DAX1 gene were assessed by PCR and direct sequencing analysis. Genetic analysis of the DAX1 gene revealed 2 novel mutations c.572-575 dupGGGC, p.Thr193Gly,fs,205X and c.773- 774 dupCC, p.Ser259Pro,fs,264X in exon 1, causing frameshifts and yeilding premature stop codons at 205 and 264, respectively. This study identifies 2 novel mutations in the DAX1 gene which can further expand the mutation database and benefit patients in the diagnosis and treatment of AHC.
Collapse
Affiliation(s)
- C M Wu
- Department of Endocrinology, The Second Affiliated Hospital of Wenzhou Medical College, Wenzhou 325027, Zhejiang, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mortensen AH, MacDonald JW, Ghosh D, Camper SA. Candidate genes for panhypopituitarism identified by gene expression profiling. Physiol Genomics 2011; 43:1105-16. [PMID: 21828248 DOI: 10.1152/physiolgenomics.00080.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mutations in the transcription factors PROP1 and PIT1 (POU1F1) lead to pituitary hormone deficiency and hypopituitarism in mice and humans. The dysmorphology of developing Prop1 mutant pituitaries readily distinguishes them from those of Pit1 mutants and normal mice. This and other features suggest that Prop1 controls the expression of genes besides Pit1 that are important for pituitary cell migration, survival, and differentiation. To identify genes involved in these processes we used microarray analysis of gene expression to compare pituitary RNA from newborn Prop1 and Pit1 mutants and wild-type littermates. Significant differences in gene expression were noted between each mutant and their normal littermates, as well as between Prop1 and Pit1 mutants. Otx2, a gene critical for normal eye and pituitary development in humans and mice, exhibited elevated expression specifically in Prop1 mutant pituitaries. We report the spatial and temporal regulation of Otx2 in normal mice and Prop1 mutants, and the results suggest Otx2 could influence pituitary development by affecting signaling from the ventral diencephalon and regulation of gene expression in Rathke's pouch. The discovery that Otx2 expression is affected by Prop1 deficiency provides support for our hypothesis that identifying molecular differences in mutants will contribute to understanding the molecular mechanisms that control pituitary organogenesis and lead to human pituitary disease.
Collapse
Affiliation(s)
- Amanda H Mortensen
- Department of Human Genetics, Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
| | | | | | | |
Collapse
|
33
|
Nuclear Receptors in Regulation of Mouse ES Cell Pluripotency and Differentiation. PPAR Res 2011; 2007:61563. [PMID: 18274628 PMCID: PMC2233893 DOI: 10.1155/2007/61563] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Accepted: 06/11/2007] [Indexed: 12/25/2022] Open
Abstract
Embryonic stem (ES) cells have great therapeutic potential because they are capable of indefinite self-renewal and have the potential to differentiate into over 200 different cell types that compose the human body. The switch from the pluripotent phenotype to a differentiated cell involves many complex signaling pathways including those involving LIF/Stat3 and the transcription factors Sox2, Nanog and Oct-4. Many nuclear receptors play an important role in the maintenance of pluripotence (ERRβ, SF-1, LRH-1, DAX-1) repression of the ES cell phenotype (RAR, RXR, GCNF) and also the differentiation of ES cells (PPARγ). Here we review the roles of the nuclear receptors involved in regulating these important processes in ES cells.
Collapse
|
34
|
Dadhich RK, Barrionuevo FJ, Lupiañez DG, Real FM, Burgos M, Jiménez R. Expression of genes controlling testicular development in adult testis of the seasonally breeding iberian mole. Sex Dev 2011; 5:77-88. [PMID: 21412037 DOI: 10.1159/000323805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2010] [Indexed: 01/21/2023] Open
Abstract
Most testicular features undergo major circannual variation in seasonal breeding species. Although the ultimate cause of these variations is known to be the photoperiod in most cases, very little is known about the genetic mechanisms by which these changes are modulated in the testis. Many genes involved in testis development are known to be expressed in the adult testis as well. Since these genes encode genetic regulatory factors, it is reasonable to suspect that they could play some role in the control of the adult testis function. Using immunological detection techniques and RT-Q-PCR, we have studied the spatio-temporal expression pattern of WT1, SF1, SOX9, AMH, and DMRT1 in 4 representative stages of the circannual cycle of the testes of Talpa occidentalis, a mole species with strict seasonal reproduction. AMH is not expressed at any stage of the cycle, showing that inactive adult testes are functionally different from pre-pubertal, juvenile ones. The continuous presence of primary spermatocytes may explain the permanent repression of AMH in the mole testis. WT1 and SF1 are down-regulated and SOX9 is up-regulated in regressed mole testes, suggesting that the modulation of the expression of these genes may be involved in the control of circannual gonad variation. Furthermore, SOX9 and DMRT1 show clear spermatogenic stage-dependent expression patterns. Both genes are expressed more intensely during the proliferative stages of spermatogonia, although SOX9 expression is limited to Sertoli cells, whereas DMRT1 is expressed in both Sertoli and spermatogonial cells. Available data suggest that intratesticular levels of testosterone could regulate circannual spermatogenic variations of seasonal breeders by modulating the expression of DMRT1 to control spermatogonial proliferation.
Collapse
Affiliation(s)
- R K Dadhich
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, Centro de Investigación Biomédica, Armilla, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Gao L, Kim Y, Kim B, Lofgren SM, Schultz-Norton JR, Nardulli AM, Heckert LL, Jorgensen JS. Two regions within the proximal steroidogenic factor 1 promoter drive somatic cell-specific activity in developing gonads of the female mouse. Biol Reprod 2011; 84:422-34. [PMID: 20962249 PMCID: PMC3043126 DOI: 10.1095/biolreprod.110.084590] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Revised: 04/15/2010] [Accepted: 10/14/2010] [Indexed: 11/01/2022] Open
Abstract
Targets of steroidogenic factor 1 (SF1; also known as NR5A1 and AD4BP) have been identified within cells at every level of the hypothalamic-pituitary-gonadal and -adrenal axes, revealing SF1 to be a master regulator of major endocrine systems. Mouse embryos express SF1 in the genital ridge until Embryonic Day 13.5 (E13.5). Thereafter, expression persists in the male and is substantially lower in the female gonad until birth. We hypothesize that the sexually dimorphic expression of Sf1 during gonadogenesis is mediated by sex-specific regulation of its promoter. To investigate dimorphic regulation within the fetal gonad, we developed an experimental strategy using transient transfection of E13.5 gonad explant cultures and evaluated various Sf1 promoter constructs for sexually dimorphic DNA elements. The proximal Sf1 promoter correctly targeted reporter activity to SF1-expressing cells in both XY and XX gonads. Stepwise deletion of sequences from the Sf1 promoter revealed two regions that affected regulation within female gonads. Mutation of both sequences together did not cause further disruption of reporter activity, suggesting the two sites might work in concert to promote activity in female somatic cells. Results from gel mobility shift assays and fetal gonad-chromatin immunoprecipitation showed that TCFAP2 binds to one of the two female-specific sites within the proximal promoter of Sf1. Together, we show that transient transfection experiments performed within developing testes and ovaries are a powerful tool to uncover elements within the Sf1 promoter that contribute to sex-specific expression.
Collapse
Affiliation(s)
- Liying Gao
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Youngha Kim
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Bongki Kim
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | | | | | - Ann M. Nardulli
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Leslie L. Heckert
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Joan S. Jorgensen
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
36
|
Frapsauce C, Ravel C, Legendre M, Sibony M, Mandelbaum J, Donadille B, Achermann JC, Siffroi JP, Christin-Maitre S. Birth after TESE-ICSI in a man with hypogonadotropic hypogonadism and congenital adrenal hypoplasia linked to a DAX-1 (NR0B1) mutation. Hum Reprod 2011; 26:724-8. [PMID: 21227944 PMCID: PMC3037794 DOI: 10.1093/humrep/deq372] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
DAX1/NR0B1 mutations are responsible for X-linked congenital adrenal hypoplasia (AHC) associated with hypogonadotropic hypogonadism (HH). Few data are available concerning testicular function and fertility in men with DAX1 mutations. Azoospermia as well as failure of gonadotrophin treatment have been reported. We induced spermatogenesis in a patient who has a DAX1 mutation (c.1210C>T), leading to a stop codon in position 404 (p.Gln404X). His endocrine testing revealed a low testosterone level at 1.2 nmol/l (N: 12–40) with low FSH and LH levels at 2.1 IU/l (N: 1–5 IU/l) and 0.1 IU/l (N: 1–4 IU/l), respectively. Baseline semen analysis revealed azoospermia. Menotropin (Menopur®:150 IU, three times weekly) and human chorionic gonadotrophin (1500 IU, twice weekly) were used. After 20 months of treatment, as azoospermia persisted, bilateral multiple site testicular biopsies were performed. Histology revealed severe hypospermatogenesis. Rare spermatozoa were extracted from the right posterior fragment and ICSI was performed. Four embryos were obtained and, after a frozen–thawed single-embryo transfer, the patient's wife became pregnant and gave birth to a healthy boy. We report the first case of paternity after TESE–ICSI in a patient with DAX1 mutation, giving potential hope to these patients to father non-affected children. Furthermore, this case illustrates the fact that patients with X-linked AHC have a primary testicular defect in addition to HH.
Collapse
Affiliation(s)
- C Frapsauce
- UPMC, AP-HP, Hôpital Tenon, Service de Biologie de la Reproduction, 75020 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huang CCJ, Yao HHC. Diverse functions of Hedgehog signaling in formation and physiology of steroidogenic organs. Mol Reprod Dev 2010; 77:489-96. [PMID: 20422709 DOI: 10.1002/mrd.21174] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Adrenal, testis, and ovary are steroidogenic organs derived from a common primordium that consists of steroidogenic factor 1 (SF1)-positive precursor cells. SF1 not only defines the steroidogenic lineages in these organs but also controls their differentiation. Recent evidence implicates the Hedgehog (Hh) signaling pathway as a downstream regulator of SF1 in the appearance of steroidogenic cells in these organs. The Hh signaling pathway serves as a common crosstalk component, yet has evolved diverse functions in the expansion and differentiation of the steroidogenic cells in a tissue-specific manner. The purpose of this review is to compare and contrast the different roles of Hh signaling in these three organs during development.
Collapse
Affiliation(s)
- Chen-Che Jeff Huang
- Department of Veterinary Biosciences, University of Illinois, Urbana, IL 61802, USA
| | | |
Collapse
|
38
|
Val P, Swain A. Gene dosage effects and transcriptional regulation of early mammalian adrenal cortex development. Mol Cell Endocrinol 2010; 323:105-14. [PMID: 20025938 DOI: 10.1016/j.mce.2009.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Pierre Val
- Centre National de la Recherche Scientifique, Unité mixte de Recherche 6247, Génétique, Reproduction et Développement, Clermont Université, 63177 Aubière, France
| | | |
Collapse
|
39
|
Abstract
Genetic control of gonadal development proceeds through either the male or female molecular pathways, driving bipotential gonadal anlage differentiation into a testis or ovary. Antagonistic interactions between the 2 pathways determine the gonadal sex. Essentially sex determination is the enhancement of one of the 2 pathways according to genetic sex. Initially, Sry with other factors upregulates Sox9 expression in XY individuals. Afterwards the expression of Sox9 is maintained by a positive feedback loop with Fgf9 and prostaglandin D2 as well as by autoregulative ability of Sox9. If these factors reach high concentrations, then Sox9 and/or Fgf9 may inhibit the female pathway. Surprisingly, splicing, nuclear transport, and extramatrix proteins may be involved in sex determination. The male sex determination pathway switches on the expression of genes driving Sertoli cell differentiation. Sertoli cells orchestrate testicular differentiation. In the absence of Sry, the predomination of the female pathway results in the realization of a robust genetic program that drives ovarian differentiation.
Collapse
|
40
|
Hoivik EA, Lewis AE, Aumo L, Bakke M. Molecular aspects of steroidogenic factor 1 (SF-1). Mol Cell Endocrinol 2010; 315:27-39. [PMID: 19616058 DOI: 10.1016/j.mce.2009.07.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 07/01/2009] [Accepted: 07/08/2009] [Indexed: 12/24/2022]
Abstract
Steroidogenic factor 1 (SF-1, also called Ad4BP and NR5A1) is a nuclear receptor with critical roles in steroidogenic tissues, as well as in the brain and pituitary. In particular, SF-1 has emerged as an essential regulator of adrenal and gonadal functions and development. In the last few years, our knowledge on SF-1 has increased considerably at all levels, from the gene to the protein, and on its specific roles in different physiological processes. In this review, we discuss the current understanding on SF-1 with focus on the parameters that control the transcriptional capacity of SF-1 and the mechanisms that ensure proper stage- and tissue-specific expression of the gene encoding SF-1.
Collapse
Affiliation(s)
- Erling A Hoivik
- Department of Biomedicine, University of Bergen, Jonas Lies vei 9, N-5009 Bergen, Norway.
| | | | | | | |
Collapse
|
41
|
Barsoum IB, Yao HHC. Fetal Leydig cells: progenitor cell maintenance and differentiation. ACTA ACUST UNITED AC 2009; 31:11-5. [PMID: 19875489 DOI: 10.2164/jandrol.109.008318] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In most eutherian mammals, sexually dimorphic masculinization is established by androgen-producing fetal Leydig cells in the embryonic testis. Fetal Leydig cells, which lack expression of the testis-determining gene SRY, arise after the appearance of SRY-expressing Sertoli cells. Therefore, the appearance and differentiation of fetal Leydig cells are probably regulated by factors derived from Sertoli cells. Results from mouse genetic models have revealed that maintenance and differentiation of fetal Leydig cell population depends upon a balance between differentiation-promoting and differentiation-suppressing mechanisms. Although paracrine signaling via Sertoli cell-derived Hedgehog ligands is necessary and sufficient for fetal Leydig cell formation, cell-cell interaction via Notch signaling and intracellular transcription factors such as POD1 are implicated as suppressors of fetal Leydig cell differentiation. This review provides a model that summarizes the recent findings in fetal Leydig cell development.
Collapse
Affiliation(s)
- Ivraym B Barsoum
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61802, USA
| | | |
Collapse
|
42
|
Gassei K, Ehmcke J, Wood MA, Walker WH, Schlatt S. Immature rat seminiferous tubules reconstructed in vitro express markers of Sertoli cell maturation after xenografting into nude mouse hosts. Mol Hum Reprod 2009; 16:97-110. [PMID: 19770206 DOI: 10.1093/molehr/gap081] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sertoli cells undergo a maturation process during post-natal testicular development that leads to the adult-type Sertoli cell, which is required for spermatogenesis. Understanding Sertoli cell maturation is therefore necessary to gain insight into the underlying causes of impaired spermatogenesis and male infertility. The present study characterized the cellular and molecular differentiation of Sertoli cells in a xenograft model of mammalian testicular development. Immature rat Sertoli cells were cultured in a three-dimensional culture system to allow the formation of cord-like structures. The in vitro Sertoli cell cultures were then grafted into nude mice. Sertoli cell proliferation, morphological differentiation and mRNA expression of Sertoli cell maturation markers were evaluated in xenografts. Sertoli cell proliferation significantly decreased between 1 and 4 weeks (6.7 +/- 0.9 versus 1.2+/- 0.1%, P < 0.001), and was maintained at low levels thereafter. Sertoli cell cord-like structures significantly decreased between 1 and 4 weeks (59.6 versus 21%, P < 0.05), whereas Sertoli cell tubules were more frequently observed after 4 weeks (13.3 versus 73.1%, P < 0.05). Furthermore, expression of androgen binding protein, transferrin and follicle stimulating hormone receptor, markers for mature Sertoli cells, was detected after 1 week of grafting and increased significantly thereafter. We conclude from these results that rat Sertoli cells continue maturation after xenografting to the physiological environment of a host. This model of in vitro tubule formation will be helpful in future investigations addressing testicular maturation in the mammalian testis.
Collapse
Affiliation(s)
- K Gassei
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
43
|
Nagl F, Schönhofer K, Seidler B, Mages J, Allescher HD, Schmid RM, Schneider G, Saur D. Retinoic acid-induced nNOS expression depends on a novel PI3K/Akt/DAX1 pathway in human TGW-nu-I neuroblastoma cells. Am J Physiol Cell Physiol 2009; 297:C1146-56. [PMID: 19726747 DOI: 10.1152/ajpcell.00034.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuronal nitric oxide synthase (nNOS)-derived nitric oxide (NO) acts as a neurotransmitter and intracellular signaling molecule in the central and peripheral nervous system. NO regulates multiple processes like neuronal development, plasticity, and differentiation and is a mediator of neurotoxicity. The nNOS gene is highly complex with 12 alternative first exons, exon 1a-1l, transcribed from distinct promoters, leading to nNOS variants with different 5'-untranslated regions. Transcriptional control of the nNOS gene is not understood in detail. To investigate regulation of nNOS gene expression by retinoic acid (RA), we used the human neuroblastoma cell line TGW-nu-I as a model system. We show that RA induces nNOS transcription in a protein synthesis-dependent fashion. We identify the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway and the atypical orphan nuclear receptor DAX1 (NR0B1) as critical mediators involved in RA-induced nNOS gene transcription. RA treatment increases DAX1 expression via PI3K/Akt signaling. Upregulation of DAX1 expression in turn induces nNOS transcription in response to RA. These results identify nNOS as a target gene of a novel RA/PI3K/Akt/DAX1-dependent pathway in human neuroblastoma cells and stress the functional importance of the transcriptional regulator DAX1 for nNOS gene expression in response to RA treatment.
Collapse
Affiliation(s)
- Florian Nagl
- II. Medizinische Klinik, Technische Universität München, 81675 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Bogani D, Siggers P, Brixey R, Warr N, Beddow S, Edwards J, Williams D, Wilhelm D, Koopman P, Flavell RA, Chi H, Ostrer H, Wells S, Cheeseman M, Greenfield A. Loss of mitogen-activated protein kinase kinase kinase 4 (MAP3K4) reveals a requirement for MAPK signalling in mouse sex determination. PLoS Biol 2009; 7:e1000196. [PMID: 19753101 PMCID: PMC2733150 DOI: 10.1371/journal.pbio.1000196] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 08/06/2009] [Indexed: 11/29/2022] Open
Abstract
Sex determination in mammals is controlled by the presence or absence of the Y-linked gene SRY. In the developing male (XY) gonad, sex-determining region of the Y (SRY) protein acts to up-regulate expression of the related gene, SOX9, a transcriptional regulator that in turn initiates a downstream pathway of testis development, whilst also suppressing ovary development. Despite the requirement for a number of transcription factors and secreted signalling molecules in sex determination, intracellular signalling components functioning in this process have not been defined. Here we report a role for the phylogenetically ancient mitogen-activated protein kinase (MAPK) signalling pathway in mouse sex determination. Using a forward genetic screen, we identified the recessive boygirl (byg) mutation. On the C57BL/6J background, embryos homozygous for byg exhibit consistent XY gonadal sex reversal. The byg mutation is an A to T transversion causing a premature stop codon in the gene encoding MAP3K4 (also known as MEKK4), a mitogen-activated protein kinase kinase kinase. Analysis of XY byg/byg gonads at 11.5 d post coitum reveals a growth deficit and a failure to support mesonephric cell migration, both early cellular processes normally associated with testis development. Expression analysis of mutant XY gonads at the same stage also reveals a dramatic reduction in Sox9 and, crucially, Sry at the transcript and protein levels. Moreover, we describe experiments showing the presence of activated MKK4, a direct target of MAP3K4, and activated p38 in the coelomic region of the XY gonad at 11.5 d post coitum, establishing a link between MAPK signalling in proliferating gonadal somatic cells and regulation of Sry expression. Finally, we provide evidence that haploinsufficiency for Map3k4 accounts for T-associated sex reversal (Tas). These data demonstrate that MAP3K4-dependent signalling events are required for normal expression of Sry during testis development, and create a novel entry point into the molecular and cellular mechanisms underlying sex determination in mice and disorders of sexual development in humans.
Collapse
Affiliation(s)
- Debora Bogani
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Rachel Brixey
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Nick Warr
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Sarah Beddow
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Jessica Edwards
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Debbie Williams
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Dagmar Wilhelm
- The Institute of Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Peter Koopman
- The Institute of Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Harry Ostrer
- Human Genetics Program, New York University School of Medicine, New York, New York, United States of America
| | - Sara Wells
- The Mary Lyon Centre, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Michael Cheeseman
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
- The Mary Lyon Centre, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council (MRC) Harwell, Oxfordshire, United Kingdom
| |
Collapse
|
45
|
Buaas FW, Val P, Swain A. The transcription co-factor CITED2 functions during sex determination and early gonad development. Hum Mol Genet 2009; 18:2989-3001. [PMID: 19457926 DOI: 10.1093/hmg/ddp237] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025] Open
Abstract
The early bi-potential mammalian gonad requires the expression of a Y-linked gene, Sry, during a brief window of time to ensure proper testis development. WT1 and its direct target gene Sf1 function during sex determination as well as in the specified testes and ovaries. We have previously shown that the transcription co-factor CITED2 interacts with WT1 to stimulate the expression of Sf1 in the adrenogonadal primordium to ensure adrenal development. We now show through genetic interactions and expression analyses that Cited2 acts in the gonad with Wt1 and Sf1 to increase the expression of Sry levels to attain a critical threshold to efficiently initiate testis development. Reducing the gene dosage of Wt1 or Sf1 in Cited2 mutant gonads was sufficient to produce partial XY sex reversal while full sex reversal was attained in mutants containing a hypomorphic Sry(POS) allele. A direct correlation was observed between XY sex reversal and reduced expression levels of Sry and Sf1 during sex determination, which indicated that Sry is a downstream target of the CITED2/WT1/SF1 regulatory pathway. Our results provide in vivo evidence for the identification of the first transcription co-factor to function during mammalian sex determination, as part of the WT1/SF1 regulatory mechanism. This highlights the gene dosage sensitivity of the pathway's effect on Sry levels and embryonic gonad development.
Collapse
Affiliation(s)
- Frank William Buaas
- Section of Gene Function and Regulation, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | | | | |
Collapse
|
46
|
Zubair M, Oka S, Parker KL, Morohashi KI. Transgenic expression of Ad4BP/SF-1 in fetal adrenal progenitor cells leads to ectopic adrenal formation. Mol Endocrinol 2009; 23:1657-67. [PMID: 19628584 DOI: 10.1210/me.2009-0055] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Deficiency of adrenal 4 binding protein/steroidogenic factor 1 (Ad4BP/SF-1; NR5A1) impairs adrenal development in a dose-dependent manner, whereas overexpression of Ad4BP/SF-1 is associated with adrenocortical tumorigenesis. Despite its essential roles in adrenal development, the mechanism(s) by which Ad4BP/SF-1 regulates this process remain incompletely understood. We previously identified a fetal adrenal enhancer (FAdE) that stimulates Ad4BP/SF-1 expression in the fetal adrenal gland by a two-step mechanism in which homeobox proteins initiate Ad4BP/SF-1 expression, which then maintains FAdE activity in an autoregulatory loop. In the present study, we examined the effect of transgenic expression of Ad4BP/SF-1 controlled by FAdE on adrenal development. When Ad4BP/SF-1 was overexpressed using a FAdE-Ad4BP/SF-1 transgene, FAdE activity expanded outside of its normal field, resulting in increased adrenal size and the formation of ectopic adrenal tissue in the thorax. The increased size of the adrenal gland did not result from a corresponding increase in cell proliferation, suggesting rather that the increased levels of Ad4BP/SF-1 may divert uncommitted precursors to the steroidogenic lineage. The effects of FAdE-controlled Ad4BP/SF-1 overexpression in mice provide a novel model of ectopic adrenal formation that further supports the critical role of Ad4BP/SF-1 in the determination of steroidogenic cell fate in vivo.
Collapse
Affiliation(s)
- Mohamad Zubair
- National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
| | | | | | | |
Collapse
|
47
|
Ehrlund A, Anthonisen EH, Gustafsson N, Venteclef N, Robertson Remen K, Damdimopoulos AE, Galeeva A, Pelto-Huikko M, Lalli E, Steffensen KR, Gustafsson JA, Treuter E. E3 ubiquitin ligase RNF31 cooperates with DAX-1 in transcriptional repression of steroidogenesis. Mol Cell Biol 2009; 29:2230-2242. [PMID: 19237537 PMCID: PMC2663311 DOI: 10.1128/mcb.00743-08] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 06/05/2008] [Accepted: 02/02/2009] [Indexed: 11/20/2022] Open
Abstract
Genetic and experimental evidence points to a critical involvement of the atypical mammalian orphan receptor DAX-1 in reproductive development and steroidogenesis. Unlike conventional nuclear receptors, DAX-1 appears not to function as a DNA-bound transcription factor. Instead, it has acquired the capability to act as a transcriptional corepressor of steroidogenic factor 1 (SF-1). The interplay of DAX-1 and SF-1 is considered a central, presumably ligand-independent element of adrenogonadal development and function that requires tight regulation. This raises a substantial interest in identifying its modulators and the regulatory signals involved. Here, we uncover molecular mechanisms that link DAX-1 to the ubiquitin modification system via functional interaction with the E3 ubiquitin ligase RNF31. We demonstrate that RNF31 is coexpressed with DAX-1 in steroidogenic tissues and participates in repressing steroidogenic gene expression. We provide evidence for the in vivo existence of a corepressor complex containing RNF31 and DAX-1 at the promoters of the StAR and CYP19 genes. Our data suggest that RNF31 functions to stabilize DAX-1, which might be linked to DAX-1 monoubiquitination. In conclusion, RNF31 appears to be required for DAX-1 to repress transcription, provides means to regulate DAX-1 in ligand-independent ways, and emerges as a relevant coregulator of steroidogenic pathways governing physiology and disease.
Collapse
Affiliation(s)
- Anna Ehrlund
- Department of Biosciences and Nutrition, Karolinska Institutet, S-14157 Huddinge/Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ikeda Y, Tanaka H, Esaki M. Effects of gestational diethylstilbestrol treatment on male and female gonads during early embryonic development. Endocrinology 2008; 149:3970-9. [PMID: 18436715 PMCID: PMC2488225 DOI: 10.1210/en.2007-1599] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To study the effects of gestational exposure to estrogen on early gonadal differentiation, pregnant mice were treated by sc injection of diethylstilbestrol (DES) or vehicle from embryonic day (E) 8.5 to E14.5, and gonads at E11.5, E12.5, and E14.5 were examined. Quantitative real-time RT-PCR and in situ hybridization revealed that mRNA levels of steroidogenic factor 1 (SF-1), a key regulator of gonadal differentiation, and several male gonad-specific genes, including Müllerian-inhibiting substance (MIS), steroidogenic acute regulatory protein, cholesterol side-chain cleavage cytochrome P450, and Cerebellin 1 precursor protein, were significantly decreased in the DES-treated testis, compared with the control testis at E12.5 and/or E14.5. Immunohistochemistry demonstrated that the staining intensities for SF-1 and MIS in Sertoli cells were apparently reduced in the DES-treated testis, compared with those of the controls, at E12.5 and E14.5. Because MIS, steroidogenic acute regulatory protein, cholesterol side-chain cleavage cytochrome P450, and Cerebellin 1 precursor protein are activated under the regulation of SF-1, the down-regulation of these factors may be due to reduced SF-1 expression. Immunohistochemistry for laminin-1 demonstrated that ovigerous cords in the DES-treated ovary were smaller than those in controls at E14.5. Moreover, the number of 5-bromo-2'deoxyuridine-5-monophosphate-labeled cells in the DES-treated testis was significantly reduced at E12.5 and E14.5, compared with controls, and that in the DES-treated ovary remained higher than that in the control ovary at E14.5. The results suggest that exogenous estrogens can alter sex-specific genetic pathways governing early differentiation and cell proliferation of both male and female gonads.
Collapse
Affiliation(s)
- Yayoi Ikeda
- Department of Histology and Cell Biology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan.
| | | | | |
Collapse
|
49
|
Sekido R, Lovell-Badge R. Sex determination involves synergistic action of SRY and SF1 on a specific Sox9 enhancer. Nature 2008; 453:930-4. [PMID: 18454134 DOI: 10.1038/nature06944] [Citation(s) in RCA: 659] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Accepted: 03/20/2008] [Indexed: 12/17/2022]
Abstract
The mammalian Y chromosome acts as a dominant male determinant as a result of the action of a single gene, Sry, whose role in sex determination is to initiate testis rather than ovary development from early bipotential gonads. It does so by triggering the differentiation of Sertoli cells from supporting cell precursors, which would otherwise give follicle cells. The related autosomal gene Sox9 is also known from loss-of-function mutations in mice and humans to be essential for Sertoli cell differentiation; moreover, its abnormal expression in an XX gonad can lead to male development in the absence of Sry. These genetic data, together with the finding that Sox9 is upregulated in Sertoli cell precursors just after SRY expression begins, has led to the proposal that Sox9 could be directly regulated by SRY. However, the mechanism by which SRY action might affect Sox9 expression was not understood. Here we show that SRY binds to multiple elements within a Sox9 gonad-specific enhancer in mice, and that it does so along with steroidogenic factor 1 (SF1, encoded by the gene Nr5a1 (Sf1)), an orphan nuclear receptor. Mutation, co-transfection and sex-reversal studies all point to a feedforward, self-reinforcing pathway in which SF1 and SRY cooperatively upregulate Sox9 and then, together with SF1, SOX9 also binds to the enhancer to help maintain its own expression after that of SRY has ceased. Our results open up the field, permitting further characterization of the molecular mechanisms regulating sex determination and how they have evolved, as well as how they fail in cases of sex reversal.
Collapse
Affiliation(s)
- Ryohei Sekido
- Division of Developmental Genetics, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK.
| | | |
Collapse
|
50
|
O'Shaughnessy PJ, Hu L, Baker PJ. Effect of germ cell depletion on levels of specific mRNA transcripts in mouse Sertoli cells and Leydig cells. Reproduction 2008; 135:839-50. [PMID: 18390686 PMCID: PMC2592074 DOI: 10.1530/rep-08-0012] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Accepted: 04/02/2008] [Indexed: 01/18/2023]
Abstract
It has been shown that testicular germ cell development is critically dependent upon somatic cell activity but, conversely, the extent to which germ cells normally regulate somatic cell function is less clear. This study was designed, therefore, to examine the effect of germ cell depletion on Sertoli cell and Leydig cell transcript levels. Mice were treated with busulphan to deplete the germ cell population and levels of mRNA transcripts encoding 26 Sertoli cell-specific proteins and 6 Leydig cell proteins were measured by real-time PCR up to 50 days after treatment. Spermatogonia were lost from the testis between 5 and 10 days after treatment, while spermatocytes were depleted after 10 days and spermatids after 20 days. By 30 days after treatment, most tubules were devoid of germ cells. Circulating FSH and intratesticular testosterone were not significantly affected by treatment. Of the 26 Sertoli cell markers tested, 13 showed no change in transcript levels after busulphan treatment, 2 showed decreased levels, 9 showed increased levels and 2 showed a biphasic response. In 60% of cases, changes in transcript levels occurred after the loss of the spermatids. Levels of mRNA transcripts encoding Leydig cell-specific products related to steroidogenesis were unaffected by treatment. Results indicate (1) that germ cells play a major and widespread role in the regulation of Sertoli cell activity, (2) most changes in transcript levels are associated with the loss of spermatids and (3) Leydig cell steroidogenesis is largely unaffected by germ cell ablation.
Collapse
Affiliation(s)
- P J O'Shaughnessy
- Division of Cell Sciences, Institute of Comparative Medicine, University of Glasgow Veterinary School, Bearsden Road, Glasgow G61 1QH, UK.
| | | | | |
Collapse
|